1
|
Lugenbühl JF, Snijders C, Pernia CD, Estruch MS, Kenis G, Daskalakis NP. Corticosteroid-regulated gene transcription in SH-SY5Y-derived neurons: Insights into the mineralocorticoid and glucocorticoid receptor-mediated response. J Neuroendocrinol 2025:e70021. [PMID: 40108866 DOI: 10.1111/jne.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Post-traumatic stress disorder (PTSD) and major depressive disorder (MDD) are debilitating stress-related psychiatric disorders that can develop following exposure to traumatic events or chronic stress in some individuals. The neurobiological processes leading to disease remain largely unknown. Among others, these disorders are characterized by a dysregulated hypothalamic-pituitary-adrenal axis, which is regulated by the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). This leads to altered downstream corticosteroid-induced gene expression. In vitro models are promising tools to investigate specific neurobiological underpinnings of the stress response in the brain. Here, we investigated the suitability of SH-SY5Y-derived neurons as a cost-efficient system to study the role of GR and MR in the neuronal stress response. SH-SY5Y-derived neurons were characterized, exposed to corticosteroids, and analyzed on transcriptomic and proteomic levels. We show that (i) these neurons express sufficient and seemingly functional GR and MR to allow the study of corticosteroid-induced transcription, (ii) three corticosteroids cortisol, dexamethasone, and aldosterone, induced similar transcriptomic effects, (iii) the antagonist spironolactone mildly attenuated the effects of dexamethasone in FKBP5, DUSP1, and SUPV3L1. Mifepristone did not significantly alter the effect of aldosterone. (iv) Integrating transcriptomic alterations of these corticosteroid-exposed neurons with those of iPSC-derived neurons exposed to dexamethasone showed concordant corticosteroid-induced effects in the two in vitro systems. To determine translational validity, we compared the gene expression in these neurons with the transcriptome of postmortem brain samples from individuals with PTSD and MDD, yielding stronger negative correlations of corticosteroid effects in SH-SY5Y-derived neurons with PTSD signatures than with MDD signatures. Upon further refinement and validation, SH-SY5Y-derived neurons may serve as a simplistic tool to study neuronal corticosteroid-induced gene expression and the implicated molecular networks around GR and MR. Strengthening our insight into these receptors' functions improves our understanding of the hypothalamic-pituitary-adrenal axis, which is commonly altered in stress-related psychiatric disorders such as PTSD and MDD.
Collapse
Affiliation(s)
- Justina F Lugenbühl
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| | - Clara Snijders
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Cameron D Pernia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Marina Soliva Estruch
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
2
|
Perovic M, Pavlovic D, Palmer Z, Udo MSB, Citadin CT, Rodgers KM, Wu CYC, Zhang Q, Lin HW, Tesic V. Modulation of GABAergic system as a therapeutic option in stroke. Exp Neurol 2025; 384:115050. [PMID: 39522803 DOI: 10.1016/j.expneurol.2024.115050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Stroke is one of the leading causes of death and permanent adult disability worldwide. Despite the improvements in reducing the rate and mortality, the societal burden and costs of treatment associated with stroke management are increasing. Most of the therapeutic approaches directly targeting ischemic injury have failed to reduce short- and long-term morbidity and mortality and more effective therapeutic strategies are still needed to promote post-stroke functional recovery. Decades of stroke research have been focused on hyperexcitability and glutamate-induced excitotoxicity in the acute phase of ischemia and their relation to motor deficits. Recent advances in understanding the pathophysiology of stroke have been made with several lines of evidence suggesting that changes in the neurotransmission of the major inhibitory system via γ-Aminobutyric acid (GABA) play a particularly important role in functional recovery and deserve further attention. The present review provides an overview of how GABAergic neurotransmission changes correlate with stroke recovery and outlines GABAergic system modulators with special emphasis on neurosteroids that have been shown to affect stroke pathogenesis or plasticity or to protect against cognitive decline. Supporting evidence from both animal and human clinical studies is presented and the potential for GABA signaling-targeted therapies for stroke is discussed to translate this concept to human neural repair therapies. Age and sex are considered crucial parameters related to the pathophysiology of stroke and important factors in the development of therapeutic pharmacological strategies. Future work is needed to deepen our knowledge of the neurochemical changes after stroke, extend the conceptual framework, and allow for the development of more effective interventions that include the modulation of the inhibitory system.
Collapse
Affiliation(s)
- Milka Perovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Damjan Pavlovic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Zoe Palmer
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Mariana S B Udo
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Cristiane T Citadin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Krista M Rodgers
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Celeste Yin-Chien Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| |
Collapse
|
3
|
Wang Z, Liang C, Shi LL, Zhu CS, Wang S, Nakayama SF, Kido T, Sun XL, Shan J. Associations Between Heavy Metal Exposure from Milk and Steroid Hormones in Mothers. Biol Trace Elem Res 2024:10.1007/s12011-024-04466-0. [PMID: 39633227 DOI: 10.1007/s12011-024-04466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
Environmental exposure to heavy metals is ubiquitous. However, its relationship with steroid hormone levels is not well understood, particularly in pregnant women. This study investigated the association between prenatal heavy metal exposure and steroid hormone levels in an e-waste disposal area in China. We analyzed the Cd, Cr, Mn, Pb, Cu, and As concentrations in 102 human milk samples collected 4 weeks after delivery. Multiple regression analysis was used to assess the associations and interactions between heavy metals and steroidal hormones. We found positive associations between Mn and estrone and estriol (estrone: β = 0.713, 95%CI = 0.046, 1.381 and estriol: β = 1.290, 95%CI = 0.494, 2.085) and between Cd and progesterone (β = 0.280; 95%CI = 0.053, 0.506). We observed negative associations between Cr and estrone and estriol (estrone: β = - 0.757, 95%CI = - 1.473, - 0.041 and estriol: β = - 1.354, 95%CI = - 2.209, - 0.499). At last, we found that Pb was negatively associated with estrone (estrone: β = - 0.537, 95%CI = - 1.053, - 0.020). Our results suggest that exposure to heavy metals may affect steroid hormone levels in mothers living in an e-waste recycling area in China.
Collapse
Affiliation(s)
- Zheng Wang
- School of Medicine, Jiaxing University, Jiaxing, China
- School of Medicine, and The First Affiliated Hospital, Huzhou University, Huzhou, China
| | - Caixia Liang
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Li Li Shi
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Cheng-Sheng Zhu
- Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Shenghang Wang
- School of Public Health, Shandong University, Jinan, China
| | - Shoji F Nakayama
- Japan Environment and Children's Study Programme Office, National Institute for Environmental Studies, Tsukuba, Japan
| | - Teruhiko Kido
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Xian Liang Sun
- School of Medicine, and The First Affiliated Hospital, Huzhou University, Huzhou, China.
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan.
| | - Jiancong Shan
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China.
| |
Collapse
|
4
|
Nobue A, Ishikawa M. Sex-Specific Differences in Peripheral Nerve Properties: A Comparative Analysis of Conduction Velocity and Cross-Sectional Area in Upper and Lower Limbs. Diagnostics (Basel) 2024; 14:2711. [PMID: 39682618 DOI: 10.3390/diagnostics14232711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/26/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Peripheral nerve conduction velocity (NCV) and nerve cross-sectional area (nCSA) are crucial parameters in neurophysiological assessments, yet their sex-specific differences are not fully understood. This study investigated sex-based variations in NCV and nCSA between upper and lower limbs. Methods: Twenty participants (ten males and ten females) were recruited for this study. The NCV and nCSA of the ulnar and tibial nerves were measured in both the upper and lower limbs. NCV was measured using supramaximal electric stimulation, and nCSA was assessed using peripheral nerve ultrasonography at three regions for each nerve. Supramaximal electric stimulations were applied superficially to the ulnar and tibial nerves at each measurement point. Action potentials were recorded from the abductor digiti minimi and soleus muscles for the ulnar and tibial nerves, respectively. Results: The ulnar nCSA of the upper limbs was significantly greater in males than in females (p < 0.05). However, ulnar NCV was significantly higher in females than in males (p < 0.05). In the lower limbs, no sex differences were observed in tibial NCV or nCSA. Conclusions: These findings reveal sex-specific differences in upper limb peripheral nerve characteristics that may have important implications for clinical assessments and treatment strategies. The contrasting patterns between upper and lower limbs suggest that both developmental and functional factors influence peripheral nerve properties.
Collapse
Affiliation(s)
- Ayaka Nobue
- Faculty of Medical Science Technology, Morinomiya University of Medical Sciences, Osaka 559-8611, Japan
| | - Masaki Ishikawa
- Graduate School of Sport and Exercise Sciences, Osaka University of Health and Sport Sciences, Osaka 590-0459, Japan
| |
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
|
Cioffi L, Diviccaro S, Chrostek G, Caruso D, Garcia-Segura LM, Melcangi RC, Giatti S. Neuroactive steroids fluctuate with regional specificity in the central and peripheral nervous system across the rat estrous cycle. J Steroid Biochem Mol Biol 2024; 243:106590. [PMID: 39053702 DOI: 10.1016/j.jsbmb.2024.106590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Neuroactive steroids (i.e., sex steroid hormones and neurosteroids) are important physiological regulators of nervous function and potential neuroprotective agents for neurodegenerative and psychiatric disorders. Sex is an important component of such effects. However, even if fluctuations in sex steroid hormone level during the menstrual cycle are associated with neuropathological events in some women, the neuroactive steroid pattern in the brain across the ovarian cycle has been poorly explored. Therefore, we assessed the levels of pregnenolone, progesterone, and its metabolites (i.e., dihydroprogesterone, allopregnanolone and isoallopregnanolone), dehydroepiandrosterone, testosterone and its metabolites (i.e., dihydrotestosterone, 3α-diol and 17β-estradiol) across the rat ovarian cycle to determine whether their plasma fluctuations are similar to those occurring in the central (i.e., hippocampus and cerebral cortex) and peripheral (i.e., sciatic nerve) nervous system. Data obtained indicate that the plasma pattern of these molecules generally does not fully reflect the events occurring in the nervous system. In addition, for some neuroactive steroid levels, the pattern is not identical between the two brain regions and between the brain and peripheral nerves. Indeed, with the exception of progesterone, all other neuroactive steroids assessed here showed peculiar regional differences in their pattern of fluctuation in the nervous system during the estrous cycle. These observations may have important diagnostic and therapeutic consequences for neuropathological events influenced by the menstrual cycle.
Collapse
Affiliation(s)
- Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Gabriela Chrostek
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Luis Miguel Garcia-Segura
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain and Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy.
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| |
Collapse
|
7
|
Liang S, Zhao Y, Liu X, Wang Y, Yang H, Zhuo D, Fan F, Guo M, Luo G, Fan Y, Zhang L, Lv X, Chen X, Li SS, Jin X. Prenatal progesterone treatment modulates fetal brain transcriptome and impacts adult offspring behavior in mice. Physiol Behav 2024; 281:114549. [PMID: 38604593 DOI: 10.1016/j.physbeh.2024.114549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Maternal exposure to elevated levels of steroid hormones during pregnancy is associated with the development of chronic conditions in offspring that manifest in adulthood. However, the effects of progesterone (P4) administration during early pregnancy on fetal development and subsequent offspring behavior remain poorly understood. In this study, we aimed to investigate the effects of P4 treatment during early pregnancy on the transcript abundance in the fetal brain and assess the behavioral consequences in the offspring during adolescence and adulthood. Using RNA-seq analysis, we examined the impact of P4 treatment on the fetal brain transcriptome in a dosage-dependent manner. Our results revealed differential regulation of genes involved in neurotransmitter transport, synaptic transmission, and transcriptional regulation. Specifically, we observed bidirectional regulation of transcription factors (TFs) by P4 at different doses, highlighting the critical role of these TFs in neurodevelopment. To assess behavioral outcomes, we conducted open field and elevated plus maze tests. Offspring treated with low-dose P4 (LP4) displayed increased exploratory behavior during both adolescence and adulthood. In contrast, the high-dose P4 (HP4) group exhibited impaired exploration and heightened anxiety-like behaviors compared to the control mice. Moreover, in a novel object recognition test, HP4-treated offspring demonstrated impaired object recognition memory during both developmental stages. Additionally, both LP4 and HP4 groups showed reduced social interaction in the three-chamber test. These results suggest that prenatal exposure to P4 exerts a notable influence on the expression of genes associated with neurodevelopment and may induce alterations in behavioral characteristics in progeny, highlighting the need to monitor progesterone levels during pregnancy for long-term impacts on fetal brain development and behavior.
Collapse
Affiliation(s)
- Shuang Liang
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Ying Zhao
- School of Medicine, Nankai University, Tianjin, China
| | - Xiuwei Liu
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yan Wang
- Jiujiang Maternal and Child Health Hospital, China
| | | | - Donghai Zhuo
- School of Medicine, Nankai University, Tianjin, China
| | - Feifei Fan
- School of Medicine, Nankai University, Tianjin, China
| | - Miao Guo
- School of Medicine, Nankai University, Tianjin, China
| | - Gan Luo
- Tianjin Medical University, Tianjin, China
| | - Yonggang Fan
- School of Medicine, Nankai University, Tianjin, China
| | - Lingzhu Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Xinxin Lv
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Chen
- School of Medicine, Nankai University, Tianjin, China; Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Shan-Shan Li
- School of Medicine, Nankai University, Tianjin, China
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, China; Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China.
| |
Collapse
|
8
|
Zhang L, Verwer RWH, van Heerikhuize J, Lucassen PJ, Nathanielsz PW, Hol EM, Aronica E, Dhillo WS, Meynen G, Swaab DF. Progesterone receptor distribution in the human hypothalamus and its association with suicide. Acta Neuropathol Commun 2024; 12:16. [PMID: 38263257 PMCID: PMC10807127 DOI: 10.1186/s40478-024-01733-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
The human hypothalamus modulates mental health by balancing interactions between hormonal fluctuations and stress responses. Stress-induced progesterone release activates progesterone receptors (PR) in the human brain and triggers alterations in neuropeptides/neurotransmitters. As recent epidemiological studies have associated peripheral progesterone levels with suicide risks in humans, we mapped PR distribution in the human hypothalamus in relation to age and sex and characterized its (co-) expression in specific cell types. The infundibular nucleus (INF) appeared to be the primary hypothalamic structure via which progesterone modulates stress-related neural circuitry. An elevation of the number of pro-opiomelanocortin+ (POMC, an endogenous opioid precursor) neurons in the INF, which was due to a high proportion of POMC+ neurons that co-expressed PR, was related to suicide in patients with mood disorders (MD). MD donors who died of legal euthanasia were for the first time enrolled in a postmortem study to investigate the molecular signatures related to fatal suicidal ideations. They had a higher proportion of PR co-expressing POMC+ neurons than MD patients who died naturally. This indicates that the onset of endogenous opioid activation in MD with suicide tendency may be progesterone-associated. Our findings may have implications for users of progesterone-enriched contraceptives who also have MD and suicidal tendencies.
Collapse
Affiliation(s)
- Lin Zhang
- Neuropsychiatric Disorders Lab, Neuroimmunology Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Ronald W H Verwer
- Neuropsychiatric Disorders Lab, Neuroimmunology Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Joop van Heerikhuize
- Neuropsychiatric Disorders Lab, Neuroimmunology Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter W Nathanielsz
- Department of Animal Science, College of Agriculture and Natural Resources, University of Wyoming, Laramie, USA
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Waljit S Dhillo
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Gerben Meynen
- Faculty of Humanities, VU University Amsterdam, Amsterdam, the Netherlands
- Willem Pompe Institute for Criminal Law and Criminology and Utrecht Centre for Accountability and Liability Law (UCALL), Utrecht University, Utrecht, the Netherlands
| | - Dick F Swaab
- Neuropsychiatric Disorders Lab, Neuroimmunology Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands.
- Netherlands Institute for Neuroscience, Dept. Neuropsychiatric Disorders, University of Amsterdam, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
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
|
10
|
Terrin F, Tesoriere A, Plotegher N, Dalla Valle L. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson's Disease. Cells 2023; 12:1486. [PMID: 37296608 PMCID: PMC10252697 DOI: 10.3390/cells12111486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Sex hormones and genes on the sex chromosomes are not only key factors in the regulation of sexual differentiation and reproduction but they are also deeply involved in brain homeostasis. Their action is crucial for the development of the brain, which presents different characteristics depending on the sex of individuals. The role of these players in the brain is fundamental in the maintenance of brain function during adulthood as well, thus being important also with respect to age-related neurodegenerative diseases. In this review, we explore the role of biological sex in the development of the brain and analyze its impact on the predisposition toward and the progression of neurodegenerative diseases. In particular, we focus on Parkinson's disease, a neurodegenerative disorder that has a higher incidence in the male population. We report how sex hormones and genes encoded by the sex chromosomes could protect from the disease or alternatively predispose toward its development. We finally underline the importance of considering sex when studying brain physiology and pathology in cellular and animal models in order to better understand disease etiology and develop novel tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| | - Luisa Dalla Valle
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| |
Collapse
|
11
|
Lucchi C, Codeluppi A, Filaferro M, Vitale G, Rustichelli C, Avallone R, Mandrioli J, Biagini G. Human Microglia Synthesize Neurosteroids to Cope with Rotenone-Induced Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12040963. [PMID: 37107338 PMCID: PMC10135967 DOI: 10.3390/antiox12040963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
We obtained evidence that mouse BV2 microglia synthesize neurosteroids dynamically to modify neurosteroid levels in response to oxidative damage caused by rotenone. Here, we evaluated whether neurosteroids could be produced and altered in response to rotenone by the human microglial clone 3 (HMC3) cell line. To this aim, HMC3 cultures were exposed to rotenone (100 nM) and neurosteroids were measured in the culture medium by liquid chromatography with tandem mass spectrometry. Microglia reactivity was evaluated by measuring interleukin 6 (IL-6) levels, whereas cell viability was monitored by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. After 24 h (h), rotenone increased IL-6 and reactive oxygen species levels by approximately +37% over the baseline, without affecting cell viability; however, microglia viability was significantly reduced at 48 h (p < 0.01). These changes were accompanied by the downregulation of several neurosteroids, including pregnenolone, pregnenolone sulfate, 5α-dihydroprogesterone, and pregnanolone, except for allopregnanolone, which instead was remarkably increased (p < 0.05). Interestingly, treatment with exogenous allopregnanolone (1 nM) efficiently prevented the reduction in HMC3 cell viability. In conclusion, this is the first evidence that human microglia can produce allopregnanolone and that this neurosteroid is increasingly released in response to oxidative stress, to tentatively support the microglia's survival.
Collapse
Affiliation(s)
- Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Alessandro Codeluppi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Monica Filaferro
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanni Vitale
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cecilia Rustichelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Rossella Avallone
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department of Neurosciences, Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria di Modena, 41126 Modena, Italy
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
12
|
Ocañas SR, Ansere VA, Kellogg CM, Isola JVV, Chucair-Elliott AJ, Freeman WM. Chromosomal and gonadal factors regulate microglial sex effects in the aging brain. Brain Res Bull 2023; 195:157-171. [PMID: 36804773 PMCID: PMC10810555 DOI: 10.1016/j.brainresbull.2023.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Biological sex contributes to phenotypic sex effects through genetic (sex chromosomal) and hormonal (gonadal) mechanisms. There are profound sex differences in the prevalence and progression of age-related brain diseases, including neurodegenerative diseases. Inflammation of neural tissue is one of the most consistent age-related phenotypes seen with healthy aging and disease. The pro-inflammatory environment of the aging brain has primarily been attributed to microglial reactivity and adoption of heterogeneous reactive states dependent upon intrinsic (i.e., sex) and extrinsic (i.e., age, disease state) factors. Here, we review sex effects in microglia across the lifespan, explore potential genetic and hormonal molecular mechanisms of microglial sex effects, and discuss currently available models and methods to study sex effects in the aging brain. Despite recent attention to this area, significant further research is needed to mechanistically understand the regulation of microglial sex effects across the lifespan, which may open new avenues for sex informed prevention and treatment strategies.
Collapse
Affiliation(s)
- Sarah R Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Victor A Ansere
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Collyn M Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jose V V Isola
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
13
|
Antonelli A, Giannini A, Chedraui P, Monteleone P, Caretto M, Genazzani AD, Mannella P, Simoncini T, Genazzani AR. Mood disorders and hormonal status across women's life: a narrative review. Gynecol Endocrinol 2022; 38:1019-1027. [PMID: 36433781 DOI: 10.1080/09513590.2022.2149730] [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] [Indexed: 11/28/2022] Open
Abstract
Depressive disorders and anxiety states represent one of the most frequent psychiatric pathologies occurring transiently in vulnerable women throughout their life, from puberty to menopause. It is now known that sex hormones play a key role on the nervous system, interfering with neuronal plasticity and enhancing the processes of learning, memory, cognition, and mood. Numerous mechanisms are at the base of these processes, displaying interactions between estrogen and serotoninergic, dopaminergic, and GABAergic receptors at the central level. Therefore, given the sexual steroids fluctuations throughout the entire female lifespan, and considering the role played by sex hormones at the central level, it is not surprising to observe the onset of mood or neurodegenerative disorders over time. This is especially true for women in hormonal transition phase, such as puberty, postpartum and the menopausal transition. Moreover, all these conditions are characterized by hormone withdrawal, imbalance, or modifications due to menopausal hormone therapies or contraceptives which could prompt to a deterioration of mood and cognition impairment or to an improvement in the quality of life. More studies are needed to better understand the hormone-related effects on the nervous system, and the underlying pathways involved in transitional or chronic mood disorders, to promote new patient-specific therapeutic strategies more effective than the current ones and tailored according to the individual need and women's life period.
Collapse
Affiliation(s)
- Alice Antonelli
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Giannini
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Peter Chedraui
- Instituto de Investigación e Innovación en Salud Integral, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador
- Facultad de Ciencias de la Salud, Universidad Católica "Nuestra Señora de la Asunción", Asunción, Paraguay
| | - Patrizia Monteleone
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marta Caretto
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessandro D Genazzani
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Mannella
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Tommaso Simoncini
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea R Genazzani
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
14
|
Orsini CA, Truckenbrod LM, Wheeler AR. Regulation of sex differences in risk-based decision making by gonadal hormones: Insights from rodent models. Behav Processes 2022; 200:104663. [PMID: 35661794 PMCID: PMC9893517 DOI: 10.1016/j.beproc.2022.104663] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/22/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023]
Abstract
Men and women differ in their ability to evaluate options that vary in their rewards and the risks that are associated with these outcomes. Most studies have shown that women are more risk averse than men and that gonadal hormones significantly contribute to this sex difference. Gonadal hormones can influence risk-based decision making (i.e., risk taking) by modulating the neurobiological substrates underlying this cognitive process. Indeed, estradiol, progesterone and testosterone modulate activity in the prefrontal cortex, amygdala and nucleus accumbens associated with reward and risk-related information. The use of animal models of decision making has advanced our understanding of the intersection between the behavioral, neural and hormonal mechanisms underlying sex differences in risk taking. This review will outline the current state of this literature, identify the current gaps in knowledge and suggest the neurobiological mechanisms by which hormones regulate risky decision making. Collectively, this knowledge can be used to understand the potential consequences of significant hormonal changes, whether endogenously or exogenously induced, on risk-based decision making as well as the neuroendocrinological basis of neuropsychiatric diseases that are characterized by impaired risk taking, such as substance use disorder and schizophrenia.
Collapse
Affiliation(s)
- Caitlin A. Orsini
- Department of Psychology, University of Texas at Austin, Austin, TX, USA,Department of Neurology, University of Texas at Austin, Austin, TX, USA,Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA,Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA,Correspondence to: Department of Psychology & Neurology, Waggoner Center for Alcohol and Addiction Research, 108 E. Dean Keaton St., Stop A8000, Austin, TX 78712, USA. (C.A. Orsini)
| | - Leah M. Truckenbrod
- Department of Neurology, University of Texas at Austin, Austin, TX, USA,Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA,Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Alexa-Rae Wheeler
- Department of Neurology, University of Texas at Austin, Austin, TX, USA,Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA,Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
15
|
Kolatorova L, Vitku J, Suchopar J, Hill M, Parizek A. Progesterone: A Steroid with Wide Range of Effects in Physiology as Well as Human Medicine. Int J Mol Sci 2022; 23:7989. [PMID: 35887338 PMCID: PMC9322133 DOI: 10.3390/ijms23147989] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022] Open
Abstract
Progesterone is a steroid hormone traditionally linked with female fertility and pregnancy. In current reproductive medicine, progesterone and its analogues play crucial roles. While the discovery of its effects has a long history, over recent decades, various novel actions of this interesting steroid have been documented, of which its neuro- and immunoprotective activities are the most widely discussed. Discoveries of the novel biological activities of progesterone have also driven research and development in the field of progesterone analogues used in human medicine. Progestogen treatment has traditionally and predominately been used in maintaining pregnancy, the prevention of preterm labor, various gynecological pathologies, and in lowering the negative effects of menopause. However, there are also various other medical fields where progesterone and its analogues could find application in the future. The aim of this work is to show the mechanisms of action of progesterone and its metabolites, the physiological and pharmacological actions of progesterone and its synthetic analogues in human medicine, as well as the impacts of its production and use on the environment.
Collapse
Affiliation(s)
- Lucie Kolatorova
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic; (J.V.); (M.H.)
| | - Jana Vitku
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic; (J.V.); (M.H.)
| | - Josef Suchopar
- DrugAgency, a.s., Klokotska 833/1a, 142 00 Prague, Czech Republic;
| | - Martin Hill
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic; (J.V.); (M.H.)
| | - Antonin Parizek
- Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General Teaching Hospital, Apolinarska 18, 128 51 Prague, Czech Republic;
| |
Collapse
|
16
|
Zorrilla Veloz RI, McKenzie T, Palacios BE, Hu J. Nuclear hormone receptors in demyelinating diseases. J Neuroendocrinol 2022; 34:e13171. [PMID: 35734821 PMCID: PMC9339486 DOI: 10.1111/jne.13171] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Demyelination results from the pathological loss of myelin and is a hallmark of many neurodegenerative diseases. Despite the prevalence of demyelinating diseases, there are no disease modifying therapies that prevent the loss of myelin or promote remyelination. This review aims to summarize studies in the field that highlight the importance of nuclear hormone receptors in the promotion and maintenance of myelination and the relevance of nuclear hormone receptors as potential therapeutic targets for demyelinating diseases. These nuclear hormone receptors include the estrogen receptor, progesterone receptor, androgen receptor, vitamin D receptor, thyroid hormone receptor, peroxisome proliferator-activated receptor, liver X receptor, and retinoid X receptor. Pre-clinical studies in well-established animal models of demyelination have shown a prominent role of these nuclear hormone receptors in myelination through their promotion of oligodendrocyte maturation and development. The activation of the nuclear hormone receptors by their ligands also promotes the synthesis of myelin proteins and lipids in mouse models of demyelination. There are limited clinical studies that focus on how the activation of these nuclear hormone receptors could alleviate demyelination in patients with diseases such as multiple sclerosis (MS). However, the completed clinical trials have reported improved clinical outcome in MS patients treated with the ligands of some of these nuclear hormone receptors. Together, the positive results from both clinical and pre-clinical studies point to nuclear hormone receptors as promising therapeutic targets to counter demyelination.
Collapse
Affiliation(s)
- Rocío I Zorrilla Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Takese McKenzie
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridgitte E Palacios
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
17
|
Hata T, Nan H, Koh K, Ishiura H, Tsuji S, Takiyama Y. A clinical and genetic study of SPG31 in Japan. J Hum Genet 2022; 67:421-425. [DOI: 10.1038/s10038-022-01021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 11/09/2022]
|
18
|
Shah SB. COVID-19 and Progesterone: Part 1.SARS-CoV-2, Progesterone and its potential clinical use. ENDOCRINE AND METABOLIC SCIENCE 2021; 5:100109. [PMID: 34396353 PMCID: PMC8349425 DOI: 10.1016/j.endmts.2021.100109] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/28/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) infection is a global medical challenge. Experience based medicines and therapies are being attempted and vaccines are being developed. SARS-CoV-2 exhibits varied patterns of infection and clinical presentations with varied disease outcomes. These attributes are strongly suggestive of some variables that differ among individuals and that affect the course of SARS-CoV-2 infection and symptoms of COVID-19 (Corona Virus Disease of 2019). Sex hormones vary with ageing, between the sexes, among individuals and populations. Sex hormones are known to play a role in immunity and infections. Progesterone is a critical host factor to promote faster recovery following Influenza A virus infection. Anti-inflammatory effects of progesterone are noted. In part 1 of the current study the regulatory role of progesterone for SARS-CoV-2 infection and COVID-19 is analyzed. The role of progesterone at different stages of the SARS CoV-2 infection is investigated with respect to two types of immunity status: immune regulation and immune dysregulation. Progesterone could have various alleviating impacts from SARS-CoV-2 entry till recovery: reversing of hypoxia, stabilizing of blood pressure, controlling thrombosis, balancing electrolytes, reducing the viral load, regulation of immune responses, damage repair, and clearance of debris among others. The present research adds to the available evidence by providing a comprehensive and thorough evaluation of the regulatory role of progesterone in SARS COV-2 infection, COVID-19 pathogenesis, and immune dysregulation. The available evidence has implications for upcoming studies about pathophysiology of COVID-19, as well as the roles of progesterone and other hormones in other infectious diseases.
Collapse
|
19
|
Meknatkhah S, Mousavi MS, Sharif Dashti P, Azizzadeh Pormehr L, Riazi GH. The brain 3β-HSD up-regulation in response to deteriorating effects of background emotional stress: an animal model of multiple sclerosis. Metab Brain Dis 2021; 36:1253-1258. [PMID: 33721183 DOI: 10.1007/s11011-021-00708-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
The brain 3β-hydroxysteroid dehydrogenase (3β-HSD), is the enzyme that catalyzes the biosynthesis of a neuroprotective factor, progesterone. The regulation of 3β-HSD in response to stress exposure in the cuprizone-induced model of Multiple Sclerosis was investigated and the reaction related to the demyelination extremity. 32 female Wistar rats divided into four groups (i.e., control group (Cont), non-stress cuprizone treated (N-CPZ), physical stress- cuprizone treated (P-CPZ) and emotional stress- cuprizone treated (E-CPZ). A witness foot-shock model used to induce background stress for 5 days. An elevated-plus maze applied to validate the stress induction. Followed by 6 weeks of cuprizone treatment, the Y-maze test performed to confirm brain demyelination. 3β-HSD gene expression as an indicator of progesterone synthesis examined. At the behavioral level, both stressed groups reflected more impaired spatial memory compared to the N-CPZ group (p < 0.01), with more severe results in the E-CPZ group (p < 0.01). The results of mRNA expression of 3β-HSD illustrated significant elevation in all cuprizone treated groups (p < 0.001) with a higher up-regulation (p < 0.001) in the E-CPZ group. Background stress -particularly emotional type- exacerbates the demyelination caused by cuprizone treatment. The brain up-regulates the 3β-HSD gene expression as a protective response relative to the myelin degradation extent.
Collapse
Affiliation(s)
- Sogol Meknatkhah
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Monireh-Sadat Mousavi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Pouya Sharif Dashti
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Leila Azizzadeh Pormehr
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Gholam Hossein Riazi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
20
|
Chen S, Wang T, Yao J, Brinton RD. Allopregnanolone Promotes Neuronal and Oligodendrocyte Differentiation In Vitro and In Vivo: Therapeutic Implication for Alzheimer's Disease. Neurotherapeutics 2020; 17:1813-1824. [PMID: 32632771 PMCID: PMC7851314 DOI: 10.1007/s13311-020-00874-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Previous studies demonstrated that the endogenous neurosteroid allopregnanolone (Allo) promotes regeneration of rodent and human neural progenitor/neural stem cells (NSCs) in vitro and in vivo, and restores neurogenesis and cognitive function in the male triple transgenic mouse model of Alzheimer's disease (3xTgAD). In this study, we investigated Allo regulation of neuronal differentiation of adult mouse neural stem cells from both sexes. Outcomes indicated that the age-dependent shift from neuronal to glial differentiation was accelerated and magnified in 3xTgAD adult NSCs compared to that in age-matched non-Tg NSCs. Coincident with the decline in neuronal differentiation, the number of immature neurons declined earlier in 3xTgAD mice, which was consistent with observations in the aged Alzheimer's human brain. Allo treatment restored the neuron/astrocyte ratio derived from adult 3xTgAD NSCs and increased both NSC proliferation and differentiation in the 3xTgAD brain. Allo treatment also significantly increased expression of Olig2, an oligodendrocyte precursor cell marker, as well as Olig2-positive cells in the corpus callosum of 3xTgAD mice. Increased neuronal and oligodendrocyte differentiation was paralleled by an increase in the expression levels of insulin-like growth factor-1 (IGF-1) and IGF-1 receptor (IGF-1R). Collectively, these findings are consistent with Allo acting as a pleiotropic therapeutic to promote regeneration of gray and white matter in the Alzheimer's brain.
Collapse
Affiliation(s)
- Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, USA
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, USA
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, USA.
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
21
|
Vass RA, Bell EF, Colaizy TT, Schmelzel ML, Johnson KJ, Walker JR, Ertl T, Roghair RD. Hormone levels in preterm and donor human milk before and after Holder pasteurization. Pediatr Res 2020; 88:612-617. [PMID: 32000260 DOI: 10.1038/s41390-020-0789-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND After birth, breastfeeding is the exclusive source of hormonal signaling between mother and infant. Hospitalized infants often receive donor milk when their own mother's milk is unavailable. METHODS The presence of insulin, leptin, cortisol, progesterone, and testosterone was examined in samples from milk bank donors and mothers of preterm infants. We further investigated the effect of Holder pasteurization (HoP) on hormone levels. RESULTS Comparing nonpasteurized samples, leptin levels were nearly threefold higher in milk from mothers of preterm infants versus donated milk, and regardless of milk source, leptin levels were significantly decreased by HoP. Insulin concentrations were also decreased by HoP, and among mothers of preterm infants, obesity was associated with significantly higher content of leptin and insulin. While combined use of donor milk and HoP was associated with cortisol levels nearly threefold higher than those in nonpasteurized own mother's milk, progesterone and testosterone content did not differ by source or pasteurization. CONCLUSIONS The hormonal composition of breast milk is impacted by HoP and maternal obesity. Compared to nonpasteurized maternal milk, use of pasteurized donor milk dramatically decreases the intake of leptin while increasing the intake of cortisol. Further research is necessary to define optimal breast milk processing practices.
Collapse
Affiliation(s)
- Réka A Vass
- Departments of Neonatology and Obstetrics & Gynecology, University of Pécs Medical School, Pécs, Hungary.,MTA-PTE Human Reproduction Scientific Research Group, Pécs, Hungary
| | - Edward F Bell
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Tarah T Colaizy
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mendi L Schmelzel
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Karen J Johnson
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jacky R Walker
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Tibor Ertl
- Departments of Neonatology and Obstetrics & Gynecology, University of Pécs Medical School, Pécs, Hungary.,MTA-PTE Human Reproduction Scientific Research Group, Pécs, Hungary
| | - Robert D Roghair
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
22
|
Sinchak K, Mohr MA, Micevych PE. Hypothalamic Astrocyte Development and Physiology for Neuroprogesterone Induction of the Luteinizing Hormone Surge. Front Endocrinol (Lausanne) 2020; 11:420. [PMID: 32670203 PMCID: PMC7333179 DOI: 10.3389/fendo.2020.00420] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/26/2020] [Indexed: 12/26/2022] Open
Abstract
Neural circuits in female rats sequentially exposed to estradiol and progesterone underlie so-called estrogen positive feedback that induce the surge release of pituitary luteinizing hormone (LH) leading to ovulation and luteinization of the corpus hemorrhagicum. It is now well-established that gonadotropin releasing hormone (GnRH) neurons express neither the reproductively critical estrogen receptor-α (ERα) nor classical progesterone receptor (PGR). Estradiol from developing ovarian follicles acts on ERα-expressing kisspeptin neurons in the rostral periventricular region of the third ventricle (RP3V) to induce PGR expression, and kisspeptin release. Circulating estradiol levels that induce positive feedback also induce neuroprogesterone (neuroP) synthesis in hypothalamic astrocytes. This local neuroP acts on kisspeptin neurons that express PGR to augment kisspeptin expression and release needed to stimulate GnRH release, triggering the LH surge. In vitro and in vivo studies demonstrate that neuroP signaling in kisspeptin neurons occurs through membrane PGR activation of Src family kinase (Src). This signaling cascade has been also implicated in PGR signaling in the arcuate nucleus of the hypothalamus, suggesting that Src may be a common mode of membrane PGR signaling. Sexual maturation requires that signaling between neuroP synthesizing astrocytes, kisspeptin and GnRH neurons be established. Prior to puberty, estradiol does not facilitate the synthesis of neuroP in hypothalamic astrocytes. During pubertal development, levels of membrane ERα increase in astrocytes coincident with an increase of PKA phosphorylation needed for neuroP synthesis. Currently, it is not clear whether these developmental changes occur in existing astrocytes or are due to a new population of astrocytes born during puberty. However, strong evidence suggests that it is the former. Blocking new cell addition during puberty attenuates the LH surge. Together these results demonstrate the importance of pubertal maturation involving hypothalamic astrocytes, estradiol-induced neuroP synthesis and membrane-initiated progesterone signaling for the CNS control of ovulation and reproduction.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA, United States
| | - Margaret A Mohr
- The Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at UCLA, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Paul E Micevych
- The Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at UCLA, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
23
|
Frye CA, Chittur SV. Mating Enhances Expression of Hormonal and Trophic Factors in the Midbrain of Female Rats. Front Behav Neurosci 2020; 14:21. [PMID: 32351369 PMCID: PMC7176275 DOI: 10.3389/fnbeh.2020.00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/31/2020] [Indexed: 11/23/2022] Open
Abstract
Among female rats, mating enhances neurosteroid formation in the midbrain ventral tegmental area (VTA; independent of peripheral steroid-secreting glands, ovaries, and adrenals). The sources/targets for these actions are not well understood. In Experiment 1, proestrous rats engaged in a mating paradigm, or did not, and the midbrains had been assessed via the Affymetrix rat genome microarrays. In Experiment 2, the influence of gonadal and adrenal glands on the expression of these genes was assessed in rats that were proestrous, ovariectomized (OVX), or OVX and adrenalectomized (ADX). The microarrays revealed 53 target genes that were significantly up-regulated (>2.0-fold change) in response to mating. Mating significantly enhanced the midbrain mRNA expression of genes involved in hormonal and trophic actions: Gh1, S100g, and Klk1b3 in proestrous, but not OVX and/or ADX, rats; Fshb in all but OVX/ADX rats; and lutenizing hormone β and thyroid-stimulating hormone (TSH) β in all rats. Thus, mating enhances midbrain gene expression independent and dependent of peripheral glands.
Collapse
Affiliation(s)
- Cheryl A Frye
- Department of Psychology, The University at Albany-SUNY, Albany, NY, United States.,Department of Biological Sciences, The University at Albany-SUNY, Albany, NY, United States.,Center for Neuroscience Research, The University at Albany-SUNY, Albany, NY, United States.,Center for Life Sciences Research, The University at Albany-SUNY, Albany, NY, United States
| | - Sridar V Chittur
- Center for Functional Genomics, The University at Albany-SUNY, Albany, NY, United States
| |
Collapse
|
24
|
González-Orozco JC, Camacho-Arroyo I. Progesterone Actions During Central Nervous System Development. Front Neurosci 2019; 13:503. [PMID: 31156378 PMCID: PMC6533804 DOI: 10.3389/fnins.2019.00503] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/30/2019] [Indexed: 01/10/2023] Open
Abstract
Although progesterone is a steroid hormone mainly associated with female reproductive functions, such as uterine receptivity and maintenance of pregnancy, accumulating data have shown its physiological actions to extend to several non-reproductive functions in the central nervous system (CNS) both in males and females. In fact, progesterone is de novo synthesized in specific brain regions by neurons and glial cells and is involved in the regulation of various molecular and cellular processes underlying myelination, neuroprotection, neuromodulation, learning and memory, and mood. Furthermore, progesterone has been reported to be implicated in critical developmental events, such as cell differentiation and neural circuits formation. This view is supported by the increase in progesterone synthesis observed during pregnancy in both the placenta and the fetal brain. In the present review, we will focus on progesterone actions during CNS development.
Collapse
Affiliation(s)
- Juan Carlos González-Orozco
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
25
|
Shaw JC, Berry MJ, Dyson RM, Crombie GK, Hirst JJ, Palliser HK. Reduced Neurosteroid Exposure Following Preterm Birth and Its' Contribution to Neurological Impairment: A Novel Avenue for Preventative Therapies. Front Physiol 2019; 10:599. [PMID: 31156466 PMCID: PMC6529563 DOI: 10.3389/fphys.2019.00599] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
Children born preterm are at an increased risk of developing cognitive problems and neuro-behavioral disorders such as attention deficit hyperactivity disorder (ADHD) and anxiety. Whilst neonates born at all gestational ages, even at term, can experience poor cognitive outcomes due to birth-complications such as birth asphyxia, it is becoming widely known that children born preterm in particular are at significant risk for learning difficulties with an increased utilization of special education resources, when compared to their healthy term-born peers. Additionally, those born preterm have evidence of altered cerebral myelination with reductions in white matter volumes of the frontal cortex, hippocampus and cerebellum evident on magnetic resonance imaging (MRI). This disruption to myelination may underlie some of the pathophysiology of preterm-associated brain injury. Compared to a fetus of the same post-conceptional age, the preterm newborn loses access to in utero factors that support and promote healthy brain development. Furthermore, the preterm ex utero environment is hostile to the developing brain with a myriad of environmental, biochemical and excitotoxic stressors. Allopregnanolone is a key neuroprotective fetal neurosteroid which has promyelinating effects in the developing brain. Preterm birth leads to an abrupt loss of the protective effects of allopregnanolone, with a dramatic drop in allopregnanolone concentrations in the preterm neonatal brain compared to the fetal brain. This occurs in conjunction with reduced myelination of the hippocampus, subcortical white matter and cerebellum; thus, damage to neurons, astrocytes and especially oligodendrocytes of the developing nervous system can occur in the vulnerable developmental window prior to term as a consequence reduced allopregnanolone. In an effort to prevent preterm-associated brain injury a number of therapies have been considered, but to date, other than antenatal magnesium sulfate and corticosteroid therapy, none have become part of standard clinical care for vulnerable infants. Therefore, there remains an urgent need for improved therapeutic options to prevent brain injury in preterm neonates. The actions of the placentally derived neurosteroid allopregnanolone on GABAA receptor signaling has a major role in late gestation neurodevelopment. The early loss of this intrauterine neurotrophic support following preterm birth may be pivotal to development of neurodevelopmental morbidity. Thus, restoring the in utero neurosteroid environment for preterm neonates may represent a new and clinically feasible treatment option for promoting better trajectories of myelination and brain development, and therefore reducing neurodevelopmental disorders in children born preterm.
Collapse
Affiliation(s)
- Julia C. Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Mary J. Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, Wellington, New Zealand
- Centre for Translational Physiology, University of Otago, Wellington, Wellington, New Zealand
| | - Rebecca M. Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, Wellington, New Zealand
- Centre for Translational Physiology, University of Otago, Wellington, Wellington, New Zealand
| | - Gabrielle K. Crombie
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Jonathan J. Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Hannah K. Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
26
|
Abstract
Sex differences exist in the regulation of energy homeostasis. Better understanding of the underlying mechanisms for sexual dimorphism in energy balance may facilitate development of gender-specific therapies for human diseases, e.g. obesity. Multiple organs, including the brain, liver, fat and muscle, play important roles in the regulations of feeding behavior, energy expenditure and physical activity, which therefore contribute to the maintenance of energy balance. It has been increasingly appreciated that this multi-organ system is under different regulations in male vs. female animals. Much of effort has been focused on roles of sex hormones (including androgens, estrogens and progesterone) and sex chromosomes in this sex-specific regulation of energy balance. Emerging evidence also indicates that other factors (not sex hormones/receptors and not encoded by the sex chromosomes) exist to regulate energy homeostasis differentially in males vs. females. In this review, we summarize factors and signals that have been shown to regulate energy homeostasis in a sexually dimorphic fashion and propose a framework where these factors and signals may be integrated to mediate sex differences in energy homeostasis.
Collapse
Affiliation(s)
- Chunmei Wang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030
| | - Yong Xu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030
| |
Collapse
|
27
|
Salem HF, Kharshoum RM, Abou-Taleb HA, AbouTaleb HA, AbouElhassan KM. Progesterone-loaded nanosized transethosomes for vaginal permeation enhancement: formulation, statistical optimization, and clinical evaluation in anovulatory polycystic ovary syndrome. J Liposome Res 2018; 29:183-194. [DOI: 10.1080/08982104.2018.1524483] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Heba F. Salem
- Faculty of Pharmacy, Department of Pharmaceutics and Industrial Pharmacy, Beni Suef University, Beni Suef, Egypt
| | - Rasha M. Kharshoum
- Faculty of Pharmacy, Department of Pharmaceutics and Industrial Pharmacy, Beni Suef University, Beni Suef, Egypt
| | - Heba A. Abou-Taleb
- Faculty of Pharmacy, Department of Pharmaceutics and Clinical Pharmacy, Nahda University (NUB), Beni Suef, Egypt
| | - Hisham A. AbouTaleb
- Faculty of Medicine, Department of Obstetrics and Gynecology, Assiut University, Assiut, Egypt
| | - Kariman M. AbouElhassan
- Faculty of Pharmacy, Department of Pharmaceutics and Clinical Pharmacy, Nahda University (NUB), Beni Suef, Egypt
| |
Collapse
|
28
|
Qiu ZK, Liu X, Tang D, Zhang Z, Fan QH, Pan YY, Chen YY, Huang MY, Zhu T, Wang YL, Cheng XF, Chen JS. Cytoprotective effects of paeoniflorin are associated with translocator protein 18 kDa. Biomed Pharmacother 2018; 107:19-23. [PMID: 30075369 DOI: 10.1016/j.biopha.2018.07.112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 01/07/2023] Open
Abstract
Paeoniflorin (PF) is one of the important active components in peony that are known to produce the neuroprotective effects. However, the involved cytoprotective factors on brain astrocytes are remain unclear. Translocator protein 18 kDa (TSPO) and its downstream neurosteroids biosynthesis play a significant role in cytoprotection. Based on these, the role of TSPO and neurosteroids biosynthesis in the cytoprotective effects of PF is evaluated. The astrocyte cells were cultured and AC-5216 (TSPO ligand) was selected as the positive control drug. The cytoprotective effects of PF and the levels of neurosteroids were quantified by water-soluble tetrazolium assay and enzyme linked immunosorbent assay, respectively. The cytoprotective activities of PF were relevant to neurosteroids (e.g. progsterone and allopregnanolone) biosynthesis, while these effects were totally blocked by PK11195, trilostane and finasteride, respectively. In summary, the cytoprotective effects of PF maybe mediated by TSPO and neurosteroids biosynthesis. The findings may provide the new insights into the cytoprotective effects of PF.
Collapse
Affiliation(s)
- Zhi-Kun Qiu
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Xu Liu
- Pharmacy Department of General Hospital of Chinese People's Armed Police Forces, Beijing 100039, PR China
| | - Dan Tang
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Zhe Zhang
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Qing-Hong Fan
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Yun-Yun Pan
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou 510080, PR China
| | - Ying-Yu Chen
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou 510080, PR China
| | - Mei-Yan Huang
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Tao Zhu
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou 510080, PR China
| | - Yu-Lu Wang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, PR China
| | - Xiao-Fang Cheng
- Tuina and Pain Management Department, Shenzhen LongGang Hospital of Traditional Chinese Medicine (Beijing University of Chinese Medicine Shenzhen Hospital), Shenzhen 518172, PR China.
| | - Ji-Sheng Chen
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou 510080, PR China.
| |
Collapse
|
29
|
|
30
|
Eaton J, Pradhan DS, Barske J, Fusani L, Canoine V, Schlinger BA. 3β-HSD expression in the CNS of a manakin and finch. Gen Comp Endocrinol 2018; 256:43-49. [PMID: 28935582 PMCID: PMC5742301 DOI: 10.1016/j.ygcen.2017.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 09/01/2017] [Accepted: 09/16/2017] [Indexed: 12/26/2022]
Abstract
The prohormone, dehydroepiandrosterone (DHEA) circulates in vertebrate blood with the potential for actions on target tissues including the central nervous system (CNS). Many actions of DHEA require its conversion into more active products, some of which are catalyzed by the enzyme 3β-hydroxysteroid-dehydrogenase/isomerase (3β-HSD). Studies of birds show both expression and activity of 3β-HSD in brain and its importance in regulating social behavior. In oscine songbirds, 3β-HSD is expressed at reasonably high levels in brain, possibly linked to their complex neural circuitry controlling song. Studies also indicate that circulating DHEA may serve as the substrate for neural 3β-HSD to produce active steroids that activate behavior during non-breeding seasons. In the golden-collared manakin (Manacus vitellinus), a sub-oscine bird, low levels of courtship behavior are displayed by males when circulating testosterone levels are basal. Therefore, we asked whether DHEA circulates in blood of manakins and whether the brain expresses 3β-HSD mRNA. Given that the spinal cord is a target of androgens and likely important in regulating acrobatic movements, we also examined expression of this enzyme in the manakin spinal cord. For comparison, we examined expression levels with those of an oscine songbird, the zebra finch (Taeniopygia guttata), a species in which brain, but not spinal cord, 3β-HSD has been well studied. DHEA was detected in manakin blood at levels similar to that seen in other species. As described previously, 3β-HSD was expressed in all zebra finch brain regions examined. By contrast, expression of 3β-HSD was only detected in the manakin hypothalamus where levels were greater than zebra finches. In spinal cord, 3β-HSD was detected in some but not all regions in both species. These data point to species differences and indicate that manakins have the substrate and neural machinery to convert circulating DHEA into potentially active androgens and/or estrogens.
Collapse
Affiliation(s)
- Joy Eaton
- Department of Integrative Biology and Physiology, University of California, Los Angeles, United States
| | - Devaleena S Pradhan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, United States; Laboratory for Neuroendocrinology, University of California, Los Angeles, United States.
| | - Julia Barske
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, United States
| | - Leonida Fusani
- Department of Cognitive Biology, University of Vienna, Austria; Konrad Lorenz Institute of Ethology, University of Veterinary Medicine, Vienna, Austria
| | - Virginie Canoine
- Department of Behavioural Biology, University of Vienna, Austria
| | - Barney A Schlinger
- Department of Integrative Biology and Physiology, University of California, Los Angeles, United States; Laboratory for Neuroendocrinology, University of California, Los Angeles, United States; Department of Ecology and Evolutionary Biology, University of California, Los Angeles, United States
| |
Collapse
|
31
|
Expression and regulation of CYP17A1 and 3β-hydroxysteroid dehydrogenase in cells of the nervous system: Potential effects of vitamin D on brain steroidogenesis. Neurochem Int 2017; 113:46-55. [PMID: 29162485 DOI: 10.1016/j.neuint.2017.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/16/2017] [Accepted: 11/16/2017] [Indexed: 11/21/2022]
Abstract
Steroids are reported to have diverse functions in the nervous system. Enzymatic production of steroid hormones has been reported in different cell types, including astrocytes and neurons. However, the information on some of the steroidogenic enzymes involved is insufficient in many respects. Contradictory results have been reported concerning the relative importance of different cell types in the nervous system for expression of CYP17A1 and 3β-hydroxysteroid dehydrogenase (3β-HSD). 3β-HSD is important in all basic steroidogenic pathways and CYP17A1 is required to form sex hormones. In the current investigation we studied the expression of these enzymes in cultured primary rat astrocytes, in neuron-enriched cells from rat cerebral cortex and in human neuroblastoma SH-SY5Y cells, a cell line often used as an in vitro model of neuronal function and differentiation. As part of this study we also examined potential effects on CYP17A1 and 3β-HSD by vitamin D, a compound previously shown to have regulatory effects in steroid hormone-producing cells outside the brain. The results of our study indicate that astrocytes are a major site for expression of 3β-HSD whereas expression of CYP17A1 is found in both astrocytes and neurons. The current data suggest that neurons, contrary to some previous reports, are not involved in 3β-HSD reactions. Previous studies have shown that vitamin D can influence gene expression and hormone production by steroidogenic enzymes in some cells. We found that vitamin D suppressed CYP17A1-mediated activity by 20% in SH-SY5Ycells and astrocytes. Suppression of CYP17A1 mRNA levels was considerably stronger, about 50% in SH-SY5Y cells and 75% in astrocytes. In astrocytes 3β-HSD was also suppressed by vitamin D, about 20% at the enzyme activity level and 60% at the mRNA level. These data suggest that vitamin D-mediated regulation of CYP17A1 and 3β-HSD, particularly on the transcriptional level, may play a role in the nervous system.
Collapse
|
32
|
Bennett GA, Palliser HK, Shaw JC, Palazzi KL, Walker DW, Hirst JJ. Maternal stress in pregnancy affects myelination and neurosteroid regulatory pathways in the guinea pig cerebellum. Stress 2017; 20:580-588. [PMID: 28969480 DOI: 10.1080/10253890.2017.1378637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Prenatal stress predisposes offspring to behavioral pathologies. These may be attributed to effects on cerebellar neurosteroids and GABAergic inhibitory signaling, which can be linked to hyperactivity disorders. The aims were to determine the effect of prenatal stress on markers of cerebellar development, a key enzyme in neurosteroid synthesis and the expression of GABAA receptor (GABAAR) subunits involved in neurosteroid signaling. We used a model of prenatal stress (strobe light exposure, 2 h on gestational day 50, 55, 60 and 65) in guinea pigs, in which we have characterized anxiety and neophobic behavioral outcomes. The cerebellum and plasma were collected from control and prenatally stressed offspring at term (control fetus: n = 9 male, n = 7 female; stressed fetus: n = 7 male, n = 8 female) and postnatal day (PND) 21 (control: n = 8 male, n = 8 female; stressed: n = 9 male, n = 6 female). We found that term female offspring exposed to prenatal stress showed decreased expression of mature oligodendrocytes (∼40% reduction) and these deficits improved to control levels by PND21. Reactive astrocyte expression was lower (∼40% reduction) following prenatal stress. GABAAR subunit (δ and α6) expression and circulating allopregnanolone concentrations were not affected by prenatal stress. Prenatal stress increased expression (∼150-250% increase) of 5α-reductase type-1 mRNA in the cerebellum, which may be a neuroprotective response to promote GABAergic inhibition and aid in repair. These observations indicate that prenatal stress exposure has marked effects on the development of the cerebellum. These findings suggest cerebellar changes after prenatal stress may contribute to adverse behavioral outcomes after exposure to these stresses.
Collapse
Affiliation(s)
- Greer A Bennett
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| | - Hannah K Palliser
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| | - Julia C Shaw
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| | - Kerrin L Palazzi
- c Clinical Research Design , Information Technology and Statistical Support (CReDITSS), Hunter Medical Research Institute (HMRI) , Newcastle , New South Wales , Australia
| | - David W Walker
- d School of Health and Biomedical Sciences , RMIT University , Bundoora , Victoria , Australia
| | - Jonathan J Hirst
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| |
Collapse
|
33
|
Cyr KJ, Avaldi OM, Wikswo JP. Circadian hormone control in a human-on-a-chip: In vitro biology's ignored component? Exp Biol Med (Maywood) 2017; 242:1714-1731. [PMID: 29065796 PMCID: PMC5832251 DOI: 10.1177/1535370217732766] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Organs-on-Chips (OoCs) are poised to reshape dramatically the study of biology by replicating in vivo the function of individual and coupled human organs. Such microphysiological systems (MPS) have already recreated complex physiological responses necessary to simulate human organ function not evident in two-dimensional in vitro biological experiments. OoC researchers hope to streamline pharmaceutical development, accelerate toxicology studies, limit animal testing, and provide new insights beyond the capability of current biological models. However, to develop a physiologically accurate Human-on-a-Chip, i.e., an MPS homunculus that functions as an interconnected, whole-body, model organ system, one must couple individual OoCs with proper fluidic and metabolic scaling. This will enable the study of the effects of organ-organ interactions on the metabolism of drugs and toxins. Critical to these efforts will be the recapitulation of the complex physiological signals that regulate the endocrine, metabolic, and digestive systems. To date, with the exception of research focused on reproductive organs on chips, most OoC research ignores homuncular endocrine regulation, in particular the circadian rhythms that modulate the function of all organ systems. We outline the importance of cyclic endocrine regulation and the role that it may play in the development of MPS homunculi for the pharmacology, toxicology, and systems biology communities. Moreover, we discuss the critical end-organ hormone interactions that are most relevant for a typical coupled-OoC system, and the possible research applications of a missing endocrine system MicroFormulator (MES-µF) that could impose biological rhythms on in vitro models. By linking OoCs together through chemical messenger systems, advanced physiological phenomena relevant to pharmacokinetics and pharmacodynamics studies can be replicated. The concept of a MES-µF could be applied to other standard cell-culture systems such as well plates, thereby extending the concept of circadian hormonal regulation to much of in vitro biology. Impact statement Historically, cyclic endocrine modulation has been largely ignored within in vitro cell culture, in part because cultured cells typically have their media changed every day or two, precluding hourly adjustment of hormone concentrations to simulate circadian rhythms. As the Organ-on-Chip (OoC) community strives for greater physiological realism, the contribution of hormonal oscillations toward regulation of organ systems has been examined only in the context of reproductive organs, and circadian variation of the breadth of other hormones on most organs remains unaddressed. We illustrate the importance of cyclic endocrine modulation and the role that it plays within individual organ systems. The study of cyclic endocrine modulation within OoC systems will help advance OoC research to the point where it can reliably replicate in vitro key regulatory components of human physiology. This will help translate OoC work into pharmaceutical applications and connect the OoC community with the greater pharmacology and physiology communities.
Collapse
Affiliation(s)
- Kevin J. Cyr
- Vanderbilt Institute for Integrative Biosystems Research and Education
- Systems Biology and Bioengineering Undergraduate Research Experience
| | - Omero M. Avaldi
- Vanderbilt Institute for Integrative Biosystems Research and Education
- Systems Biology and Bioengineering Undergraduate Research Experience
| | - John P. Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education
- Department of Biomedical Engineering
- Department of Molecular Physiology and Biophysics
- Department of Physics and Astronomy, Vanderbilt University, Nashville TN, 37235, USA
| |
Collapse
|
34
|
Marquina-Sánchez B, González-Jorge J, Hansberg-Pastor V, Wegman-Ostrosky T, Baranda-Ávila N, Mejía-Pérez S, Camacho-Arroyo I, González-Arenas A. The interplay between intracellular progesterone receptor and PKC plays a key role in migration and invasion of human glioblastoma cells. J Steroid Biochem Mol Biol 2017; 172:198-206. [PMID: 27717886 DOI: 10.1016/j.jsbmb.2016.10.001] [Citation(s) in RCA: 13] [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: 04/29/2016] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 10/20/2022]
Abstract
Intracellular progesterone receptors (PRs) and protein kinases C (PKCs) are known regulators of cancer cell proliferation and metastasis. Both PRs and PKCs are found overexpressed in grade IV human astrocytomas, also known as glioblastomas, which are the most frequent and aggressive brain tumors. In the present study, we investigated whether PR activation by PKC induces the migration and invasion of glioblastoma derived cell lines and if PKCα and δ isoforms are involved in PR activation. We observed that PKC activation with tetradecanoylphorbol acetate (TPA) increases the migration and invasion capacity of two human glioblastoma derived human cell lines (U251 MG and U87) and that the treatment with the PR receptor antagonist RU486 blocks these processes. Interestingly, the pharmacological inhibition of the isoenzymes PKCα and PKCδ also resulted in a blocked PR transcriptional activity. Also, TPA-dependent PR activation increases the expression of progesterone-induced blocking factor (PIBF), a known PR target gene. These results hint to an existing cross-talk between PKCs and PRs in regulating the infiltration process of human glioblastomas.
Collapse
Affiliation(s)
- Brenda Marquina-Sánchez
- Departamento de Medicina Genómica y Toxicología Ambiental, Programa de Investigación en Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jesús González-Jorge
- Departamento de Medicina Genómica y Toxicología Ambiental, Programa de Investigación en Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Valeria Hansberg-Pastor
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Mexico
| | - Talia Wegman-Ostrosky
- Dirección de Investigación, Instituto Nacional Cancerología, Ciudad de México, Mexico
| | - Noemi Baranda-Ávila
- División de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico
| | - Sonia Mejía-Pérez
- Subdirección de Neurocirugía, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Programa de Investigación en Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
35
|
Bansal R, Singh R. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Med Res Rev 2017; 38:1126-1158. [PMID: 28697282 DOI: 10.1002/med.21458] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Neurodegeneration is a complex process, which leads to progressive brain damage due to loss of neurons. Despite exhaustive research, the cause of neuronal loss in various degenerative disorders is not entirely understood. Neuroprotective steroids constitute an important line of attack, which could play a major role against the common mechanisms associated with various neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Natural endogenous steroids induce the neuroprotection by protecting the nerve cells from neuronal injury through multiple mechanisms, therefore the structural modifications of the endogenous steroids could be helpful in the generation of new therapeutically useful neuroprotective agents. The review article will keep the readers apprised of the detailed description of natural as well as synthetic neuroprotective steroids from the medicinal chemistry point of view, which would be helpful in drug discovery efforts aimed toward neurodegenerative diseases.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranjit Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
36
|
Shaw JC, Palliser HK, Palazzi K, Hirst JJ. Administration of Progesterone Throughout Pregnancy Increases Maternal Steroids Without Adverse Effect on Mature Oligodendrocyte Immunostaining in the Guinea Pig. Reprod Sci 2017. [PMID: 28631553 DOI: 10.1177/1933719117715125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Progesterone is administered to pregnant women at risk of premature labor, despite systematic reviews showing conflicting outcomes regarding its use, highlighting doubt over the effectiveness of the therapy. Progesterone can be rapidly metabolized into a number of steroids, but to date, there has been a lack of investigation into the fetal steroid profiles following administration and whether this impacts fetal neurodevelopment. The objective of this study was to determine the effect of progesterone treatment on allopregnanolone and cortisol levels in the fetus and on a marker of myelination in the fetal brain. We used a guinea pig model where pregnant dams were administered vehicle (β-cyclodextrin) or progesterone orally throughout pregnancy (GA29-61). Maternal and fetal fluids and tissues were collected at both preterm (GA61) and term (GA68) ages. Maternal and fetal progesterone and cortisol were analyzed by enzyme immunoassay and allopregnanolone by radioimmunoassay. Measurement of myelination of fetal brains (hippocampus, cingulum, and subcortical white matter) at preterm and term ages was performed by immunohistochemistry staining for myelin basic protein. We found that dams receiving progesterone had significantly elevated progesterone and cortisol concentrations, but there was no effect on allopregnanolone. Interestingly, the increased cortisol concentrations were not reflected in the fetuses, and there was no effect of progesterone treatment on myelination. Therefore, we conclude that in our guinea pig model, maternal administration of progesterone has no effect on cortisol levels or markers of mature oligodendrocytes in the fetus and suggest this is potentially due to the protective cortisol barrier in the placenta.
Collapse
Affiliation(s)
- Julia C Shaw
- 1 School of Biomedical Sciences and Pharmacy, University of Newcastle, New South Wales, Australia.,2 Mothers and Babies Research Centre, Hunter Medical Research Institute, New South Wales, Australia
| | - Hannah K Palliser
- 1 School of Biomedical Sciences and Pharmacy, University of Newcastle, New South Wales, Australia.,2 Mothers and Babies Research Centre, Hunter Medical Research Institute, New South Wales, Australia
| | - Kerrin Palazzi
- 3 Clinical Research Design, Information Technology and Statistical Support, Hunter Medical Research Institute, New South Wales, Australia
| | - Jonathan J Hirst
- 1 School of Biomedical Sciences and Pharmacy, University of Newcastle, New South Wales, Australia.,2 Mothers and Babies Research Centre, Hunter Medical Research Institute, New South Wales, Australia
| |
Collapse
|
37
|
Hagage-Dobensky D, Aserin A, Garti N. Progesterone solubilization within reverse hexagonal mesophase. Colloids Surf A Physicochem Eng Asp 2017. [DOI: 10.1016/j.colsurfa.2016.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
38
|
Proliferative and Invasive Effects of Progesterone-Induced Blocking Factor in Human Glioblastoma Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1295087. [PMID: 28168193 PMCID: PMC5266854 DOI: 10.1155/2017/1295087] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/24/2016] [Accepted: 12/15/2016] [Indexed: 11/22/2022]
Abstract
Progesterone-induced blocking factor (PIBF) is a progesterone (P4) regulated protein expressed in different types of high proliferative cells including astrocytomas, the most frequent and aggressive brain tumors. It has been shown that PIBF increases the number of human astrocytoma cells. In this work, we evaluated PIBF regulation by P4 and the effects of PIBF on proliferation, migration, and invasion of U87 and U251 cells, both derived from human glioblastomas. PIBF mRNA expression was upregulated by P4 (10 nM) from 12 to 24 h. Glioblastoma cells expressed two PIBF isoforms, 90 and 57 kDa. The content of the shorter isoform was increased by P4 at 24 h, while progesterone receptor antagonist RU486 (10 μM) blocked this effect. PIBF (100 ng/mL) increased the number of U87 cells on days 4 and 5 of treatment and induced cell proliferation on day 4. Wound-healing assays showed that PIBF increased the migration of U87 (12–48 h) and U251 (24 and 48 h) cells. Transwell invasion assays showed that PIBF augmented the number of invasive cells in both cell lines at 24 h. These data suggest that PIBF promotes proliferation, migration, and invasion of human glioblastoma cells.
Collapse
|
39
|
Mittelman-Smith MA, Rudolph LM, Mohr MA, Micevych PE. Rodent Models of Non-classical Progesterone Action Regulating Ovulation. Front Endocrinol (Lausanne) 2017; 8:165. [PMID: 28790975 PMCID: PMC5522857 DOI: 10.3389/fendo.2017.00165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/27/2017] [Indexed: 11/13/2022] Open
Abstract
It is becoming clear that steroid hormones act not only by binding to nuclear receptors that associate with specific response elements in the nucleus but also by binding to receptors on the cell membrane. In this newly discovered manner, steroid hormones can initiate intracellular signaling cascades which elicit rapid effects such as release of internal calcium stores and activation of kinases. We have learned much about the translocation and signaling of steroid hormone receptors from investigations into estrogen receptor α, which can be trafficked to, and signal from, the cell membrane. It is now clear that progesterone (P4) can also elicit effects that cannot be exclusively explained by transcriptional changes. Similar to E2 and its receptors, P4 can initiate signaling at the cell membrane, both through progesterone receptor and via a host of newly discovered membrane receptors (e.g., membrane progesterone receptors, progesterone receptor membrane components). This review discusses the parallels between neurotransmitter-like E2 action and the more recently investigated non-classical P4 signaling, in the context of reproductive behaviors in the rodent.
Collapse
Affiliation(s)
- Melinda A. Mittelman-Smith
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
- *Correspondence: Melinda A. Mittelman-Smith,
| | - Lauren M. Rudolph
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
| | - Margaret A. Mohr
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
| | - Paul E. Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
40
|
Worsley R, Santoro N, Miller KK, Parish SJ, Davis SR. Hormones and Female Sexual Dysfunction: Beyond Estrogens and Androgens--Findings from the Fourth International Consultation on Sexual Medicine. J Sex Med 2016; 13:283-90. [PMID: 26944460 DOI: 10.1016/j.jsxm.2015.12.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/18/2015] [Indexed: 01/20/2023]
Abstract
INTRODUCTION In recent years, multiple hormones have been investigated in relation to female sexual function. Because consumers can easily purchase products claiming to contain these hormones, a clear statement regarding the current state of knowledge is required. AIM To review the contribution of hormones, other than estrogens and androgens, to female sexual functioning and the evidence that specific endocrinopathies in women are associated with female sexual dysfunction (FSD) and to update the previously published International Society of Sexual Medicine Consensus on this topic. METHODS The literature was searched using several online databases with an emphasis on studies examining the physiologic role of oxytocin, prolactin, and progesterone in female sexual function and any potential therapeutic effect of these hormones. The association between common endocrine disorders, such as polycystic ovary syndrome, pituitary disorders, and obesity, and FSD also was examined. MAIN OUTCOME MEASURES Quality of data published in the literature and recommendations were based on the Grading of Recommendations Assessment, Development and Education system. RESULTS There is no evidence to support the use of oxytocin or progesterone for FSD. Treating hyperprolactinemia might lessen FSD. Polycystic ovary syndrome, obesity, and metabolic syndrome could be associated with FSD, but data are limited. There is a strong association between diabetes mellitus and FSD. CONCLUSION Further research is required; in particular, high-quality, large-scale studies of women with common endocrinopathies are needed to determine the impact of these prevalent disorders on female sexual function.
Collapse
Affiliation(s)
- Roisin Worsley
- The Women's Health Research Program, School of Public Health and Preventive Medicine, Melbourne, Victoria, Australia
| | - Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Karen K Miller
- Neuroendocrine Research Program in Women's Health, Massachusetts General Hospital; Massachusetts General Hospital Neuroendocrine and Pituitary Clinical Center; Harvard Medical School, Boston, MA, USA
| | - Sharon J Parish
- Departments of Clinical Psychiatry Clinical Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Susan R Davis
- The Women's Health Research Program, School of Public Health and Preventive Medicine, Melbourne, Victoria, Australia.
| |
Collapse
|
41
|
Siddiqui AN, Siddiqui N, Khan RA, Kalam A, Jabir NR, Kamal MA, Firoz CK, Tabrez S. Neuroprotective Role of Steroidal Sex Hormones: An Overview. CNS Neurosci Ther 2016; 22:342-50. [PMID: 27012165 PMCID: PMC6492877 DOI: 10.1111/cns.12538] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/21/2016] [Accepted: 02/21/2016] [Indexed: 12/11/2022] Open
Abstract
Progesterone, estrogens, and testosterone are the well-known steroidal sex hormones, which have been reported to have "nonreproductive "effects in the brain, specifically in the neuroprotection and neurotrophy. In the last one decade, there has been a surge in the research on the role of these hormones in neuroprotection and their positive impact on different brain injuries. The said interest has been sparked by a desire to understand the action and mechanisms of these steroidal sex hormones throughout the body. The aim of this article was to highlight the potential outcome of the steroidal hormones, viz. progesterone, estrogens, and testosterone in terms of their role in neuroprotection and other brain injuries. Their possible mechanism of action at both genomic and nongenomic level will be also discussed. As far as our knowledge goes, we are for the first time reporting neuroprotective effect and possible mechanism of action of these hormones in a single article.
Collapse
Affiliation(s)
- Ali Nasir Siddiqui
- Department of Pharmaceutical Medicine, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Nahida Siddiqui
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Rashid Ali Khan
- Department of Pharmaceutical Medicine, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Abul Kalam
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Nasimudeen R Jabir
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Hebersham, NSW, Australia
| | | | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
42
|
Micevych PE, Wong AM, Mittelman-Smith MA. Estradiol Membrane-Initiated Signaling and Female Reproduction. Compr Physiol 2016; 5:1211-22. [PMID: 26140715 DOI: 10.1002/cphy.c140056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The discoveries of rapid, membrane-initiated steroid actions and central nervous system steroidogenesis have changed our understanding of the neuroendocrinology of reproduction. Classical nuclear actions of estradiol and progesterone steroids affecting transcription are essential. However, with the discoveries of membrane-associated steroid receptors, it is becoming clear that estradiol and progesterone have neurotransmitter-like actions activating intracellular events. Ultimately, membrane-initiated actions can influence transcription. Estradiol membrane-initiated signaling (EMS) modulates female sexual receptivity and estrogen feedback regulating the luteinizing hormone (LH) surge. In the arcuate nucleus, EMS activates a lordosis-regulating circuit that extends to the medial preoptic nucleus and subsequently to the ventromedial nucleus (VMH)--the output from the limbic and hypothalamic regions. Here, we discuss how EMS leads to an active inhibition of lordosis behavior. To stimulate ovulation, EMS facilitates astrocyte synthesis of progesterone (neuroP) in the hypothalamus. Regulation of GnRH release driving the LH surge is dependent on estradiol-sensitive kisspeptin (Kiss1) expression in the rostral periventricular nucleus of the third ventricle (RP3V). NeuroP activation of the LH surge depends on Kiss1, but the specifics of signaling have not been well elucidated. RP3V Kiss1 neurons appear to integrate estradiol and progesterone information which feeds back onto GnRH neurons to stimulate the LH surge. In a second population of Kiss1 neurons, estradiol suppresses the surge but maintains tonic LH release, another critical component of the estrous cycle. Together, evidence suggests that regulation of reproduction involves membrane action of steroids, some of which are synthesized in the brain.
Collapse
Affiliation(s)
- Paul E Micevych
- UCLA - David Geffen School of Medicine Los Angeles, California, USA
| | - Angela May Wong
- UCLA - David Geffen School of Medicine Los Angeles, California, USA
| | | |
Collapse
|
43
|
Hsieh JT, Lei B, Sheng H, Venkatraman T, Lascola CD, Warner DS, James ML. Sex-Specific Effects of Progesterone on Early Outcome of Intracerebral Hemorrhage. Neuroendocrinology 2016; 103:518-30. [PMID: 26356626 DOI: 10.1159/000440883] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/07/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preclinical evidence suggests that progesterone improves recovery after intracerebral hemorrhage (ICH); however, gonadal hormones have sex-specific effects. Therefore, an experimental model of ICH was used to assess recovery after progesterone administration in male and female rats. METHODS ICH was induced in male and female Wistar rats via stereotactic intrastriatal injection of clostridial collagenase (0.5 U). Animals were randomized to receive vehicle or 8 mg/kg progesterone intraperitoneally at 2 h, then subcutaneously at 5, 24, 48, and 72 h after injury. Outcomes included relevant physiology during the first 3 h, hemorrhage and edema evolution over the first 24 h, proinflammatory transcription factor and cytokine regulation at 24 h, rotarod latency and neuroseverity score over the first 7 days, and microglial activation/macrophage recruitment at 7 days after injury. RESULTS Rotarod latency (p = 0.001) and neuroseverity score (p = 0.01) were improved in progesterone-treated males, but worsened in progesterone-treated females (p = 0.028 and p = 0.008, respectively). Progesterone decreased cerebral edema (p = 0.04), microglial activation/macrophage recruitment (p < 0.001), and proinflammatory transcription factor phosphorylated nuclear factor-x03BA;B p65 expression (p = 0.0038) in males but not females, independent of tumor necrosis factor-α, interleukin-6, and toll-like receptor-4 expression. Cerebral perfusion was increased in progesterone-treated males at 4 h (p = 0.043) but not 24 h after injury. Hemorrhage volume, arterial blood gases, glucose, and systolic blood pressure were not affected. CONCLUSIONS Progesterone administration improved early neurobehavioral recovery and decreased secondary neuroinflammation after ICH in male rats. Paradoxically, progesterone worsened neurobehavioral recovery and did not modify neuroinflammation in female rats. Future work should isolate mechanisms of sex-specific progesterone effects after ICH.
Collapse
|
44
|
|
45
|
Sarabia-Estrada R, Bañuelos-Pineda J, Osuna Carrasco LP, Jiménez-Vallejo S, Jiménez-Estrada I, Rivas-Celis E, Dueñas-Jiménez JM, Dueñas-Jiménez SH. Aberrant gastrocnemius muscle innervation by tibial nerve afferents after implantation of chitosan tubes impregnated with progesterone favored locomotion recovery in rats with transected sciatic nerve. J Neurosurg 2015; 123:270-82. [DOI: 10.3171/2014.12.jns132519] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT
Transection of peripheral nerves produces loss of sensory and/or motor function. After complete nerve cutting, the distal and proximal segment ends retract, but if both ends are bridged with unaltered chitosan, progesterone-impregnated chitosan, or silicone tubes, an axonal repair process begins. Progesterone promotes nerve repair and has neuroprotective effects thwarting regulation of neuron survival, inflammation, and edema. It also modulates aberrant axonal sprouting and demyelination. The authors compared the efficacy of nerve recovery after implantation of progesterone-loaded chitosan, unaltered chitosan, or silicone tubes after sciatic nerve transection in rats.
METHODS
After surgical removal of a 5-mm segment of the proximal sciatic nerve, rats were implanted with progesterone-loaded chitosan, unaltered chitosan, or silicone tubes in the transected nerve for evaluating progesterone and chitosan effects on sciatic nerve repair and ipsilateral hindlimb kinematic function, as well as on gastrocnemius electro-myographic responses. In some experiments, tube implantation was performed 90 minutes after nerve transection.
RESULTS
At 90 days after sciatic nerve transection and tube implantation, rats with progesterone-loaded chitosan tubes showed knee angular displacement recovery and better outcomes for step length, velocity of locomotion, and normal hindlimb raising above the ground. In contrast, rats with chitosan-only tubes showed reduced normal raising and pendulum-like hindlimb movements. Aberrant fibers coming from the tibial nerve innervated the gastrocnemius muscle, producing electromyographic responses. Electrical responses in the gastrocnemius muscle produced by sciatic nerve stimulation occurred only when the distal nerve segment was stimulated; they were absent when the proximal or intratubular segment was stimulated. A clear sciatic nerve morphology with some myelinated fiber fascicles appeared in the tube section in rats with progesterone-impregnated chitosan tubes. Some gastrocnemius efferent fibers were partially repaired 90 days after nerve resection. The better outcome in knee angle displacement may be partially attributable to the aberrant neuromuscular synaptic effects, since nerve conduction in the gastrocnemius muscle could be blocked in the progesterone-impregnated chitosan tubes. In addition, in the region of the gap produced by the nerve resection, the number of axons and amount of myelination were reduced in the sciatic nerve implanted with chitosan, progesterone-loaded chitosan, and silicone tubes. At 180 days after sciatic nerve sectioning, the knee kinematic function recovered to a level observed in control rats of a similar age. In rats with progesterone-loaded chitosan tubes, stimulation of the proximal and intratubular sciatic nerve segments produced an electromyographic response. The axon morphology of the proximal and intratubular segments of the sciatic nerve resembled that of the contralateral nontransected nerve.
CONCLUSIONS
Progesterone-impregnated chitosan tubes produced aberrant innervation of the gastrocnemius muscle, which allowed partial recovery of gait locomotion and could be adequate for reinnervating synergistic denervated muscles while a parent innervation is reestablished. Hindlimb kinematic parameters differed between younger (those at 90 days) and older (those at 180 days) rats.
Collapse
Affiliation(s)
| | | | | | | | - Ismael Jiménez-Estrada
- 3Department of Physiology, Biophysics and Neurosciences, CINVESTAV, IPN, México City D.F., México
| | | | | | | |
Collapse
|
46
|
Abstract
Neurosteroids, like allopregnanolone and pregnanolone, are endogenous regulators of neuronal excitability. Inside the brain, they are highly selective and potent modulators of GABAA receptor activity. Their anticonvulsant, anesthetics and anxiolytic properties are useful for the treatments of several neurological and psychiatric disorders via reducing the risks of side effects obtained with the commercial drugs. The principal disadvantages of endogenous neurosteroids administration are their rapid metabolism and their low oral bioavailability. Synthetic steroids analogues with major stability or endogenous neurosteroids stimulation synthesis might constitute promising novel strategies for the treatment of several disorders. Numerous studies indicate that the 3α-hydroxyl configuration is the key for binding and activity, but modifications in the steroid nucleus may emphasize different pharmacophores. So far, several synthetic steroids have been developed with successful neurosteroid-like effects. In this work, we summarize the properties of various synthetic steroids probed in trials throughout the analysis of several neurosteroids-like actions.
Collapse
Affiliation(s)
- Mariana Rey
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, (C1428ADN) Ciudad Autónoma de Buenos Aires, Argentina
| | - Héctor Coirini
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, (C1428ADN) Ciudad Autónoma de Buenos Aires, Argentina ; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
47
|
González-Arenas A, Peña-Ortiz MÁ, Hansberg-Pastor V, Marquina-Sánchez B, Baranda-Ávila N, Nava-Castro K, Cabrera-Wrooman A, González-Jorge J, Camacho-Arroyo I. PKCα and PKCδ activation regulates transcriptional activity and degradation of progesterone receptor in human astrocytoma cells. Endocrinology 2015; 156:1010-22. [PMID: 25514083 DOI: 10.1210/en.2014-1137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Progesterone regulates cancer cell proliferation and invasion through its receptors (PR-A and PR-B), whose phosphorylation modifies their transcriptional activity and induce their degradation. We identified by in silico analysis a putative residue (Ser400) in PR that might be phosphorylated by protein kinase C (PKC), a family of enzymes involved in the proliferation and infiltration of astrocytomas, the most frequent and aggressive brain tumors. A grade III human astrocytoma-derived cell line was used to study the role of PKC in PR phosphorylation, transcriptional activity, and degradation. Treatment with PKC activator [tetradecanoyl phorbol acetate (TPA)] increased PR phosphorylation in Ser400 after 5 minutes, which in turn induced PR transcriptional activity and its subsequent degradation by the 26S proteasome 3-5 hours after treatment. Silencing or inhibition of PKCα and PKCδ blocked PR phosphorylation and degradation induced by TPA. Both PR isoforms were associated with PKCα and reached the maximum association after 5 minutes of TPA addition. These data correlated with immunnofluorescence assays in which nuclear colocalization of PKCα with PR increased after TPA treatment. We observed a 2-fold increase in cell proliferation after PKC activation with TPA that was reduced with the PR antagonist, RU486. The PR S400A mutant revealed that this residue is essential for PKC-mediated PR phosphorylation and degradation. Our results show a key participation of PKCα and PKCδ in PR regulation and function.
Collapse
Affiliation(s)
- Aliesha González-Arenas
- Departamento de Biología (A.G.-A., M.A.P.-O., V.H.-P., B.M.-S., K.N.-C., A.C.-W., J.G.-J., I.C.-A.), Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, Distrito Federal, México; and División de Investigación Básica (N.B.-A.), Instituto Nacional de Cancerología, México City 14080, Distrito Federal, México
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Saher G, Stumpf SK. Cholesterol in myelin biogenesis and hypomyelinating disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1083-94. [PMID: 25724171 DOI: 10.1016/j.bbalip.2015.02.010] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/05/2015] [Accepted: 02/12/2015] [Indexed: 02/05/2023]
Abstract
The largest pool of free cholesterol in mammals resides in myelin membranes. Myelin facilitates rapid saltatory impulse propagation by electrical insulation of axons. This function is achieved by ensheathing axons with a tightly compacted stack of membranes. Cholesterol influences myelination at many steps, from the differentiation of myelinating glial cells, over the process of myelin membrane biogenesis, to the functionality of mature myelin. Cholesterol emerged as the only integral myelin component that is essential and rate-limiting for the development of myelin in the central and peripheral nervous system. Moreover, disorders that interfere with sterol synthesis or intracellular trafficking of cholesterol and other lipids cause hypomyelination and neurodegeneration. This review summarizes recent results on the roles of cholesterol in CNS myelin biogenesis in normal development and under different pathological conditions. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Gesine Saher
- Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany.
| | - Sina Kristin Stumpf
- Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
49
|
Elsworth JD, Groman SM, Jentsch JD, Leranth C, Redmond DE, Kim JD, Diano S, Roth RH. Primate phencyclidine model of schizophrenia: sex-specific effects on cognition, brain derived neurotrophic factor, spine synapses, and dopamine turnover in prefrontal cortex. Int J Neuropsychopharmacol 2015; 18:pyu048. [PMID: 25522392 PMCID: PMC4438537 DOI: 10.1093/ijnp/pyu048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/15/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cognitive deficits are a core symptom of schizophrenia, yet they remain particularly resistant to treatment. The model provided by repeatedly exposing adult nonhuman primates to phencyclidine has generated important insights into the neurobiology of these deficits, but it remains possible that administration of this psychotomimetic agent during the pre-adult period, when the dorsolateral prefrontal cortex in human and nonhuman primates is still undergoing significant maturation, may provide a greater understanding of schizophrenia-related cognitive deficits. METHODS The effects of repeated phencyclidine treatment on spine synapse number, dopamine turnover and BDNF expression in dorsolateral prefrontal cortex, and working memory accuracy were examined in pre-adult monkeys. RESULTS One week following phencyclidine treatment, juvenile and adolescent male monkeys demonstrated a greater loss of spine synapses in dorsolateral prefrontal cortex than adult male monkeys. Further studies indicated that in juvenile males, a cognitive deficit existed at 4 weeks following phencyclidine treatment, and this impairment was associated with decreased dopamine turnover, decreased brain derived neurotrophic factor messenger RNA, and a loss of dendritic spine synapses in dorsolateral prefrontal cortex. In contrast, female juvenile monkeys displayed no cognitive deficit at 4 weeks after phencyclidine treatment and no alteration in dopamine turnover or brain derived neurotrophic factor messenger RNA or spine synapse number in dorsolateral prefrontal cortex. In the combined group of male and female juvenile monkeys, significant linear correlations were detected between dopamine turnover, spine synapse number, and cognitive performance. CONCLUSIONS As the incidence of schizophrenia is greater in males than females, these findings support the validity of the juvenile primate phencyclidine model and highlight its potential usefulness in understanding the deficits in dorsolateral prefrontal cortex in schizophrenia and developing novel treatments for the cognitive deficits associated with schizophrenia.
Collapse
Affiliation(s)
- John D Elsworth
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano).
| | - Stephanie M Groman
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| | - James D Jentsch
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| | - Csaba Leranth
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| | - D Eugene Redmond
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| | - Jung D Kim
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| | - Sabrina Diano
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| | - Robert H Roth
- Neuropsychopharmacology Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (Drs Elsworth, Groman, Redmond, and Roth); Department of Psychology and Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California (Dr Jentsch); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Drs Leranth, Kim, and Diano)
| |
Collapse
|
50
|
Estradiol and Progesterone Administration After pMCAO Stimulates the Neurological Recovery and Reduces the Detrimental Effect of Ischemia Mainly in Hippocampus. Mol Neurobiol 2014; 52:1690-1703. [DOI: 10.1007/s12035-014-8963-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/23/2014] [Indexed: 10/24/2022]
|