101
|
Joksimovic SL, Covey DF, Jevtovic-Todorovic V, Todorovic SM. Neurosteroids in Pain Management: A New Perspective on an Old Player. Front Pharmacol 2018; 9:1127. [PMID: 30333753 PMCID: PMC6176051 DOI: 10.3389/fphar.2018.01127] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/14/2018] [Indexed: 12/11/2022] Open
Abstract
Since the discovery of the nervous system’s ability to produce steroid hormones, numerous studies have demonstrated their importance in modulating neuronal excitability. These central effects are mostly mediated through different ligand-gated receptor systems such as GABAA and NMDA, as well as voltage-dependent Ca2+ or K+ channels. Because these targets are also implicated in transmission of sensory information, it is not surprising that numerous studies have shown the analgesic properties of neurosteroids in various pain models. Physiological (nociceptive) pain has protective value for an organism by promoting survival in life-threatening conditions. However, more prolonged pain that results from dysfunction of nerves (neuropathic pain), and persists even after tissue injury has resolved, is one of the main reasons that patients seek medical attention. This review will focus mostly on the analgesic perspective of neurosteroids and their synthetic 5α and 5β analogs in nociceptive and neuropathic pain conditions.
Collapse
Affiliation(s)
- Sonja L Joksimovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Douglas F Covey
- Department of Developmental Biology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States.,Taylor Family Institute for Innovative Psychiatric Research, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States.,Neuroscience Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
102
|
Abstract
INTRODUCTION Epilepsy is one of the most serious neurological conditions, affecting almost 50 million people around the world. Despite more than 20 antiepileptic drugs (AEDs) available, seizures are still uncontrolled in one third of patients. Areas covered: The present paper reviews current compounds in preclinical and clinical development for the treatment of focal epilepsies and new potential molecular targets recently identified. Expert opinion: 1OP-2198, Cannabidavirin, Everolimus, FV-082, Ganaxolone, Minocycline, NAX 810-2, Padsevonil and Selurampanel seem to be particularly promising in focal epilepsy. Some of them, Everolimus and Ganaxolone, are already completing Phase III development while others are still at a preclinical stage. Everolimus represents the first example of precision-medicine in epilepsy and the first generation of disease-modifying agents but data on long-term safety are needed. Among AEDs in Phase II development, Cannabidavirin, Padsevonil and Selurampanel may represent a promising fourth generation of compounds for focal epilepsies if they successfully proceed to subsequent stages. Data on general tolerability, effects of cognition and behavior as well as the potential for interactions in polytherapy will be key element for the success or decline of these drugs.
Collapse
Affiliation(s)
- Marco Mula
- a Institute of Medical and Biomedical Education , St George's University of London , London , UK.,b Atkinson Morley Regional Neuroscience Centre , St George's University Hospitals NHS Foundation Trust , London , UK
| |
Collapse
|
103
|
Tsutsui K, Haraguchi S, Vaudry H. 7α-Hydroxypregnenolone regulating locomotor behavior identified in the brain and pineal gland across vertebrates. Gen Comp Endocrinol 2018; 265:97-105. [PMID: 28919448 DOI: 10.1016/j.ygcen.2017.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/30/2017] [Accepted: 09/13/2017] [Indexed: 01/29/2023]
Abstract
The brain synthesizes steroids de novo from cholesterol, which are called neurosteroids. Based on extensive studies on neurosteroids over the past thirty years, it is now accepted that neurosteroidogenesis in the brain is a conserved property across vertebrates. However, the formation of bioactive neurosteroids in the brain is still incompletely elucidated in vertebrates. In fact, we recently identified 7α-hydroxypregnenolone (7α-OH PREG) as a novel bioactive neurosteroid stimulating locomotor behavior in the brain of several vertebrates. The follow-up studies have demonstrated that the stimulatory action of brain 7α-OH PREG on locomotor behavior is mediated by the dopaminergic system across vertebrates. More recently, we have further demonstrated that the pineal gland, an endocrine organ located close to the brain, is a major site of the formation of bioactive neurosteroids. In addition to the brain, the pineal gland actively produces 7α-OH PREG de novo from cholesterol as a major pineal neurosteroid that acts on the brain to control locomotor rhythms. This review summarizes the identification, biosynthesis and mode of action of brain and pineal 7α-OH PREG, a new bioactive neurosteroid regulating locomotor behavior, across vertebrates.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo 162-8480, Japan.
| | - Shogo Haraguchi
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo 162-8480, Japan; Department of Biochemistry, Showa University School of Medicine, Tokyo 142-8555, Japan
| | - Hubert Vaudry
- INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandy University, 76000 Rouen, France
| |
Collapse
|
104
|
Kuang H, Yang F, Zhang Y, Wang T, Chen G. The Impact of Egg Nutrient Composition and Its Consumption on Cholesterol Homeostasis. CHOLESTEROL 2018; 2018:6303810. [PMID: 30210871 PMCID: PMC6126094 DOI: 10.1155/2018/6303810] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/08/2018] [Indexed: 02/07/2023]
Abstract
Nutrient deficiencies and excess are involved in many aspects of human health. As a source of essential nutrients, eggs have been used worldwide to support the nutritional needs of human societies. On the other hand, eggs also contain a significant amount of cholesterol, a lipid molecule that has been associated with the development of cardiovascular diseases. Whether the increase of egg consumption will lead to elevated cholesterol absorption and disruption of cholesterol homeostasis has been a concern of debate for a while. Cholesterol homeostasis is regulated through its dietary intake, endogenous biosynthesis, utilization, and excretion. Recently, some research interests have been paid to the effects of egg consumption on cholesterol homeostasis through the intestinal cholesterol absorption. Nutrient components in eggs such as phospholipids may contribute to this process. The goals of this review are to summarize the recent progress in this area and to discuss some potential benefits of egg consumption.
Collapse
Affiliation(s)
- Heqian Kuang
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| | - Fang Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Yan Zhang
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| | - Tiannan Wang
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
105
|
Dieni CV, Sullivan JA, Faralli M, Contemori S, Biscarini A, Pettorossi VE, Panichi R. 17 beta-estradiol synthesis modulates cerebellar dependent motor memory formation in adult male rats. Neurobiol Learn Mem 2018; 155:276-286. [PMID: 30125696 DOI: 10.1016/j.nlm.2018.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 12/30/2022]
Abstract
Neurosteroid 17 beta-estradiol (E2) is a steroid synthesized de novo in the nervous system that might influence neuronal activity and behavior. Nevertheless, the impact of E2 on the functioning of those neural systems in which it is slightly synthesized is less questioned. The vestibulo-ocular reflex (VOR) adaptation, may provide an ideal arena for investigating this issue. Indeed, E2 modulates cerebellar parallel fiber-Purkinje cell synaptic plasticity that underlies encoding of VOR adaptation. Moreover, aromatase expression in the cerebellum of adult rodents is maintained at very low levels and localized to Purkinje cells. The significance of age-related maintenance of low levels of aromatase expression in the cerebellum on behavior, however, has yet to be explored. Our aim in this study was to determine whether E2 synthesis exerts an effective and persistent modulation of VOR adaptation in adult male rats. To answer this question, we investigated the acute effect of blocking E2 synthesis on gain increases and decreases in VOR adaptation using an oral dose (2.5 mg/kg) of the aromatase inhibitor Letrozole in peri-pubertal and post-pubertal male rats. We found that Letrozole acutely impaired gain increases and decreases in VOR adaptation without altering basal ocular-motor performance and that these effects were similar in peri-pubertal and post-pubertal rats. Thus, in adult male rats neurosteroid E2 effectively modulates VOR adaptation in both of the periods studied. These findings imply that the adult cerebellum uses E2 synthesis for modulating motor memory formation and suggest that low and extremely localized E2 production may play a role in adaptive phenomena.
Collapse
Affiliation(s)
- Cristina V Dieni
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Mario Faralli
- Department of Medical-Surgical Specialization, Otolaryngology and Cervicofacial Surgery Division, University of Perugia, 06127 Perugia, Italy
| | - Samuele Contemori
- Department of Experimental Medicine Section of Physiology and Biochemistry, University of Perugia, 06127 Perugia, Italy
| | - Andrea Biscarini
- Department of Experimental Medicine Section of Physiology and Biochemistry, University of Perugia, 06127 Perugia, Italy
| | - Vito E Pettorossi
- Department of Experimental Medicine Section of Physiology and Biochemistry, University of Perugia, 06127 Perugia, Italy
| | - Roberto Panichi
- Department of Experimental Medicine Section of Physiology and Biochemistry, University of Perugia, 06127 Perugia, Italy.
| |
Collapse
|
106
|
Haque MN, Moon IS. Stigmasterol upregulates immediate early genes and promotes neuronal cytoarchitecture in primary hippocampal neurons as revealed by transcriptome analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 46:164-175. [PMID: 30097115 DOI: 10.1016/j.phymed.2018.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/21/2018] [Accepted: 04/04/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND The hippocampus is a vulnerable brain region that is implicated in learning and memory impairment by two pathophysiological features, that is, neurite regression and synaptic dysfunction, and stigmasterol (ST), a cholesterol-equivalent phytosterol, is known to facilitate neuromodulatory effects. PURPOSE To investigate the neuromodulatory effects of ST on the development of central nervous system neurons and the molecular bases of these effects in primary hippocampal neurons. METHODS Rat embryonic (E18-19) brain neurons were cultured in the absence or presence of ST (75 µM). Neuritogenic activities of ST were evident by increases in various morphometric parameters. To identify underlying affected genes, total RNA was isolated on day in vitro 12 (DIV 12) and mRNA high throughput sequencing (mRNA-Seq) was performed. Affected key genes for neuronal development were identified using bioinformatics tools and their upregulations were confirmed by immunocytochemistry. RESULTS Among the differentially expressed 17,337 RefSeq genes, 445 genes (up/down 293/157) passed the p-value < 0.05 criterion, 52 genes (up/down; 37/13) had a p-value < 0.05 and a false discovery rate (FDR) q-value of < 0.2, and 24 genes (up/down; 20/4) passed the more stringent criterion of both p < 0.05 and q < 0.05. After applying a stringent FDR q-value cutoff of < 0.2, it was found ST induced many immediate early genes (IEGs), and that a major proportion of upregulated genes were related to central nervous system (CNS) development (neurite outgrowth or synaptic transmission). In a Venn diagram for CNS development Gene Ontologies (GOs) (i.e., axon development, dendrite development, modulation of synaptic transmission), Reln emerged as a central player in these processes, and highly interconnected 'hub' genes, including Dcx, Egr1, Ntrk2, and Slc24a2, were revealed by gene co-expression networks. Finally, transcriptomic data was confirmed by immunocytochemistry of primary hippocampal neurons. CONCLUSION The study indicates that ST upregulates genes for neuritogenesis and synaptogenesis, and suggests ST be viewed as a potential resource for improving brain functions.
Collapse
Affiliation(s)
- Md Nazmul Haque
- Department of Anatomy, Dongguk University Graduate School of Medicine, Gyeongju 38066, Republic of Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University Graduate School of Medicine, Gyeongju 38066, Republic of Korea.
| |
Collapse
|
107
|
DHEA inhibits acute microglia-mediated inflammation through activation of the TrkA-Akt1/2-CREB-Jmjd3 pathway. Mol Psychiatry 2018; 23:1410-1420. [PMID: 28894299 DOI: 10.1038/mp.2017.167] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/05/2017] [Accepted: 06/20/2017] [Indexed: 12/16/2022]
Abstract
Dehydroepiandrosterone (DHEA) is the most abundant circulating steroid hormone in humans, produced by the adrenals, the gonads and the brain. DHEA was previously shown to bind to the nerve growth factor receptor, tropomyosin-related kinase A (TrkA), and to thereby exert neuroprotective effects. Here we show that DHEA reduces microglia-mediated inflammation in an acute lipopolysaccharide-induced neuro-inflammation model in mice and in cultured microglia in vitro. DHEA regulates microglial inflammatory responses through phosphorylation of TrkA and subsequent activation of a pathway involving Akt1/Akt2 and cAMP response element-binding protein. The latter induces the expression of the histone 3 lysine 27 (H3K27) demethylase Jumonji d3 (Jmjd3), which thereby controls the expression of inflammation-related genes and microglial polarization. Together, our data indicate that DHEA-activated TrkA signaling is a potent regulator of microglia-mediated inflammation in a Jmjd3-dependent manner, thereby providing the platform for potential future therapeutic interventions in neuro-inflammatory pathologies.
Collapse
|
108
|
Neuroprotective Effects of Dehydroepiandrosterone Sulfate Through Inhibiting Expression of Matrix Metalloproteinase-9 from Bradykinin-Challenged Astroglia. Mol Neurobiol 2018; 56:736-747. [PMID: 29796990 DOI: 10.1007/s12035-018-1125-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 05/11/2018] [Indexed: 10/16/2022]
Abstract
Dehydroepiandrosterone sulfate (DHEAS), one of the most important neuroactive steroids, is produced in the adrenals and the brain. DHEAS is believed to play a critical role in modulating different forms of cellular control, including processes associated with human neural systems. Its production rate and level in serum, adrenals, and brain gradually decrease with advancing age. The decline of DHEAS level was associated with age-related neuronal dysfunction and degeneration, most probably because the steroids protect the central nervous system (CNS) neurons against neurotoxic challenges. Moreover, increasing studies show that matrix metalloproteinases (MMPs), MMP-9 especially, are upregulated by proinflammatory mediators in the CNS disorders. The increased MMP-9 as an inflammatory biomarker of several CNS disorders that may participate in the CNS inflammation and neurodegeneration. Herein, we investigate the effects of DHEAS on brain inflammation by the model we have defined of bradykinin (BK)-induced MMP-9 expression in rat brain astrocyte (RBA) and its mechanism. The results showed that DHEAS significantly reduce MMP-9 induced by BK. Pretreatment with DHEAS can inhibit BK-stimulated phosphorylation of c-Src and PYK2. Moreover, DHEAS attenuated BK-stimulated NADPH oxidase (Nox)-derived reactive oxygen species (ROS) production, suggesting that DHEAS has an antioxidative effect. We further demonstrated that DHEAS blocked activation of ERK1/2, Akt, and c-Fos/AP-1 by BK. Finally, DHEAS decreased MMP-9-related events including RBA migration and neuronal apoptosis. The results will provide new insights into the anti-inflammatory action of DHEAS, supporting that DHEAS may have a neuroprotective effect in the improvement of the CNS disorders by reducing neuroinflammation.
Collapse
|
109
|
Weger M, Diotel N, Weger BD, Beil T, Zaucker A, Eachus HL, Oakes JA, do Rego JL, Storbeck KH, Gut P, Strähle U, Rastegar S, Müller F, Krone N. Expression and activity profiling of the steroidogenic enzymes of glucocorticoid biosynthesis and the fdx1 co-factors in zebrafish. J Neuroendocrinol 2018; 30:e12586. [PMID: 29486070 DOI: 10.1111/jne.12586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/06/2018] [Accepted: 02/22/2018] [Indexed: 01/23/2023]
Abstract
The spatial and temporal expression of steroidogenic genes in zebrafish has not been fully characterised. Because zebrafish are increasingly employed in endocrine and stress research, a better characterisation of steroidogenic pathways is required to target specific steps in the biosynthetic pathways. In the present study, we have systematically defined the temporal and spatial expression of steroidogenic enzymes involved in glucocorticoid biosynthesis (cyp21a2, cyp11c1, cyp11a1, cyp11a2, cyp17a1, cyp17a2, hsd3b1, hsd3b2), as well as the mitochondrial electron-providing ferredoxin co-factors (fdx1, fdx1b), during zebrafish development. Our studies showed an early expression of all these genes during embryogenesis. In larvae, expression of cyp11a2, cyp11c1, cyp17a2, cyp21a2, hsd3b1 and fdx1b can be detected in the interrenal gland, which is the zebrafish counterpart of the mammalian adrenal gland, whereas the fdx1 transcript is mainly found in the digestive system. Gene expression studies using quantitative reverse transcriptase-PCR and whole-mount in situ hybridisation in the adult zebrafish brain revealed a wide expression of these genes throughout the encephalon, including neurogenic regions. Using ultra-high-performance liquid chromatography tandem mass spectrometry, we were able to demonstrate the presence of the glucocorticoid cortisol in the adult zebrafish brain. Moreover, we demonstrate de novo biosynthesis of cortisol and the neurosteroid tetrahydrodeoxycorticosterone in the adult zebrafish brain from radiolabelled pregnenolone. Taken together, the present study comprises a comprehensive characterisation of the steroidogenic genes and the fdx co-factors facilitating glucocorticoid biosynthesis in zebrafish. Furthermore, we provide additional evidence of de novo neurosteroid biosynthesising in the brain of adult zebrafish facilitated by enzymes involved in glucocorticoid biosynthesis. Our study provides a valuable source for establishing the zebrafish as a translational model with respect to understanding the roles of the genes for glucocorticoid biosynthesis and fdx co-factors during embryonic development and stress, as well as in brain homeostasis and function.
Collapse
Affiliation(s)
- M Weger
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - N Diotel
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - B D Weger
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland
| | - T Beil
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - A Zaucker
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - H L Eachus
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, The Bateson Centre, Sheffield, UK
| | - J A Oakes
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, The Bateson Centre, Sheffield, UK
| | - J L do Rego
- Plateforme d'Analyse Comportementale (SCAC), Institut de Recherche et d'Innovation Biomédicale, Inserm U1234, Université de Rouen, Rouen Cedex, France
| | - K-H Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - P Gut
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland
| | - U Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - S Rastegar
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - F Müller
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - N Krone
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, The Bateson Centre, Sheffield, UK
| |
Collapse
|
110
|
Diotel N, Charlier TD, Lefebvre d'Hellencourt C, Couret D, Trudeau VL, Nicolau JC, Meilhac O, Kah O, Pellegrini E. Steroid Transport, Local Synthesis, and Signaling within the Brain: Roles in Neurogenesis, Neuroprotection, and Sexual Behaviors. Front Neurosci 2018; 12:84. [PMID: 29515356 PMCID: PMC5826223 DOI: 10.3389/fnins.2018.00084] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/02/2018] [Indexed: 01/18/2023] Open
Abstract
Sex steroid hormones are synthesized from cholesterol and exert pleiotropic effects notably in the central nervous system. Pioneering studies from Baulieu and colleagues have suggested that steroids are also locally-synthesized in the brain. Such steroids, called neurosteroids, can rapidly modulate neuronal excitability and functions, brain plasticity, and behavior. Accumulating data obtained on a wide variety of species demonstrate that neurosteroidogenesis is an evolutionary conserved feature across fish, birds, and mammals. In this review, we will first document neurosteroidogenesis and steroid signaling for estrogens, progestagens, and androgens in the brain of teleost fish, birds, and mammals. We will next consider the effects of sex steroids in homeostatic and regenerative neurogenesis, in neuroprotection, and in sexual behaviors. In a last part, we will discuss the transport of steroids and lipoproteins from the periphery within the brain (and vice-versa) and document their effects on the blood-brain barrier (BBB) permeability and on neuroprotection. We will emphasize the potential interaction between lipoproteins and sex steroids, addressing the beneficial effects of steroids and lipoproteins, particularly HDL-cholesterol, against the breakdown of the BBB reported to occur during brain ischemic stroke. We will consequently highlight the potential anti-inflammatory, anti-oxidant, and neuroprotective properties of sex steroid and lipoproteins, these latest improving cholesterol and steroid ester transport within the brain after insults.
Collapse
Affiliation(s)
- Nicolas Diotel
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - Thierry D. Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Christian Lefebvre d'Hellencourt
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - David Couret
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | | | - Joel C. Nicolau
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Olivier Meilhac
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | - Olivier Kah
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Elisabeth Pellegrini
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
111
|
Nguyen TV. Developmental effects of androgens in the human brain. J Neuroendocrinol 2018; 30. [PMID: 28489322 DOI: 10.1111/jne.12486] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/25/2022]
Abstract
Neuroendocrine theories of brain development posit that androgens play a crucial role in sex-specific cortical growth, although little is known about the differential effects of testosterone and dehydroepiandrosterone (DHEA) on cortico-limbic development and cognition during adolescence. In this context, the National Institutes of Health Study of Normal Brain Development, a longitudinal study of typically developing children and adolescents aged 4-24 years (n=433), offers a unique opportunity to examine the developmental effects of androgens on cortico-limbic maturation and cognition. Using data from this sample, our group found that higher testosterone levels were associated with left-sided decreases in cortical thickness (CTh) in post-pubertal boys, particularly in the prefrontal cortex, compared to right-sided increases in CTh in somatosensory areas in pre-pubertal girls. Prefrontal-amygdala and prefrontal-hippocampal structural covariance (considered to reflect structural connectivity) also varied according to testosterone levels, with the testosterone-related brain phenotype predicting higher aggression levels and lower executive function, particularly in boys. By contrast, DHEA was associated with a pre-pubertal increase in CTh of several regions involved in cognitive control in both boys and girls. Covariance within several cortico-amygdalar structural networks also varied as a function of DHEA levels, with the DHEA-related brain phenotype predicting improvements in visual attention in both boys and girls. DHEA-related cortico-hippocampal structural covariance, on the other hand, predicted higher scores on a test of working memory. Interestingly, there were significant interactions between testosterone and DHEA, such that DHEA tended to mitigate the anti-proliferative effects of testosterone on brain structure. In sum, testosterone-related effects on the developing brain may lead to detrimental effects on cortical functions (ie, higher aggression and lower executive function), whereas DHEA-related effects may optimise cortical functions (ie, better attention and working memory), perhaps by decreasing the influence of amygdalar and hippocampal afferents on cortical functions.
Collapse
Affiliation(s)
- T-V Nguyen
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Department of Obstetrics-Gynecology, McGill University Health Center, Montreal, QC, Canada
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
112
|
Barendse MEA, Simmons JG, Byrne ML, Seal ML, Patton G, Mundy L, Wood SJ, Olsson CA, Allen NB, Whittle S. Brain structural connectivity during adrenarche: Associations between hormone levels and white matter microstructure. Psychoneuroendocrinology 2018; 88:70-77. [PMID: 29175736 DOI: 10.1016/j.psyneuen.2017.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 01/04/2023]
Abstract
Levels of the adrenal hormones dehydroepiandrosterone (DHEA), its sulfate (DHEAS), and testosterone, have all been linked to behavior and mental health during adrenarche, and preclinical studies suggest that these hormones influence brain development. However, little is known about how variation in these hormones is associated with white matter structure during this period of life. The current study aimed to examine associations between DHEA, DHEAS, and testosterone, and white matter microstructure during adrenarche. To avoid the confounding effect of age on hormone levels, we tested these associations in 87 children within a narrow age range (mean age 9.56 years, SD=0.34) but varying in hormone levels. All children provided saliva samples directly after waking and completed a diffusion-weighted MRI scan. Higher levels of DHEA were associated with higher mean diffusivity (MD) in a widespread cluster of white matter tracts, which was partially explained by higher radial diffusivity (RD) and partially by higher axial diffusivity (AD). In addition, there was an interaction between DHEA and testosterone, with higher levels of testosterone being associated with higher fractional anisotropy (FA) and lower MD and RD when DHEA levels were relatively high, but with lower FA and higher MD and RD when DHEA levels were low. These findings suggest that relatively early exposure to DHEA, as well as an imbalance between the adrenal hormones, may be associated with alterations in white matter microstructure. These findings highlight the potential relevance of adrenarcheal hormones for structural brain development.
Collapse
Affiliation(s)
- Marjolein E A Barendse
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Parkville, VIC, 3052, Australia.
| | - Julian G Simmons
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Parkville, VIC, 3052, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Michelle L Byrne
- Department of Psychology, University of Oregon, Eugene, OR, 97403, USA
| | - Marc L Seal
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - George Patton
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Lisa Mundy
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Stephen J Wood
- Orygen, the National Centre of Excellence for Youth Mental Health, Parkville, VIC, 3052, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia; School of Psychology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Craig A Olsson
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia; Centre for Social and Early Emotional Development, School of Psychology, Deakin University, Geelong, VIC, 3125, Australia
| | - Nicholas B Allen
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia; Department of Psychology, University of Oregon, Eugene, OR, 97403, USA
| | - Sarah Whittle
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Parkville, VIC, 3052, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| |
Collapse
|
113
|
Hojo Y, Kawato S. Neurosteroids in Adult Hippocampus of Male and Female Rodents: Biosynthesis and Actions of Sex Steroids. Front Endocrinol (Lausanne) 2018; 9:183. [PMID: 29740398 PMCID: PMC5925962 DOI: 10.3389/fendo.2018.00183] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
The brain is not only the target of steroid hormones but also is able to locally synthesize steroids de novo. Evidence of the local production of steroids in the brain has been accumulating in various vertebrates, including teleost fish, amphibia, birds, rodents, non-human primates, and humans. In this review, we mainly focus on the local production of sex steroids in the hippocampal neurons of adult rodents (rats and mice), a center for learning and memory. From the data of the hippocampus of adult male rats, hippocampal principal neurons [pyramidal cells in CA1-CA3 and granule cells in dentate gyrus (DG)] have a complete system for biosynthesis of sex steroids. Liquid chromatography with tandem-mass-spectrometry (LC-MS/MS) enabled us to accurately determine the levels of hippocampal sex steroids including 17β-estradiol (17β-E2), testosterone (T), and dihydrotestosterone (DHT), which are much higher than those in blood. Next, we review the steroid synthesis in the hippocampus of female rats, since previous knowledge had been biased toward the data from males. Recently, we clarified that the levels of hippocampal steroids fluctuate in adult female rats across the estrous cycle. Accurate determination of hippocampal steroids at each stage of the estrous cycle is of importance for providing the account for the fluctuation of female hippocampal functions, including spine density, long-term potentiation (LTP) and long-term depression (LTD), and learning and memory. These functional fluctuations in female had been attributed to the level of circulation-derived steroids. LC-MS/MS analysis revealed that the dendritic spine density in CA1 of adult female hippocampus correlates with the levels of hippocampal progesterone and 17β-E2. Finally, we introduce the direct evidence of the role of hippocampus-synthesized steroids in hippocampal function including neurogenesis, LTP, and memory consolidation. Mild exercise (2 week of treadmill running) elevated synthesis of DHT in the hippocampus, but not in the testis, of male rats, resulting in enhancement of neurogenesis in DG. Concerning synaptic plasticity, hippocampus-synthesized E2 is required for LTP induction, whereas hippocampus-synthesized DHT is required for LTD induction. Furthermore, hippocampus-synthesized E2 is involved in memory consolidation tested by object recognition and object placement tasks, both of which are hippocampus-dependent.
Collapse
Affiliation(s)
- Yasushi Hojo
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
- *Correspondence: Yasushi Hojo,
| | - Suguru Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| |
Collapse
|
114
|
Loiseau C, Cayetanot F, Joubert F, Perrin-Terrin AS, Cardot P, Fiamma MN, Frugiere A, Straus C, Bodineau L. Current Perspectives for the use of Gonane Progesteronergic Drugs in the Treatment of Central Hypoventilation Syndromes. Curr Neuropharmacol 2018; 16:1433-1454. [PMID: 28721821 PMCID: PMC6295933 DOI: 10.2174/1570159x15666170719104605] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/30/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Central alveolar hypoventilation syndromes (CHS) encompass neurorespiratory diseases resulting from congenital or acquired neurological disorders. Hypercapnia, acidosis, and hypoxemia resulting from CHS negatively affect physiological functions and can be lifethreatening. To date, the absence of pharmacological treatment implies that the patients must receive assisted ventilation throughout their lives. OBJECTIVE To highlight the relevance of determining conditions in which using gonane synthetic progestins could be of potential clinical interest for the treatment of CHS. METHODS The mechanisms by which gonanes modulate the respiratory drive were put into the context of those established for natural progesterone and other synthetic progestins. RESULTS The clinical benefits of synthetic progestins to treat respiratory diseases are mixed with either positive outcomes or no improvement. A benefit for CHS patients has only recently been proposed. We incidentally observed restoration of CO2 chemosensitivity, the functional deficit of this disease, in two adult CHS women by desogestrel, a gonane progestin, used for contraception. This effect was not observed by another group, studying a single patient. These contradictory findings are probably due to the complex nature of the action of desogestrel on breathing and led us to carry out mechanistic studies in rodents. Our results show that desogestrel influences the respiratory command by modulating the GABAA and NMDA signaling in the respiratory network, medullary serotoninergic systems, and supramedullary areas. CONCLUSION Gonanes show promise for improving ventilation of CHS patients, although the conditions of their use need to be better understood.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Laurence Bodineau
- Address correspondence to this author at the Sorbonne Universités, UPMC Univ. Paris 06, INSERM, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, F-75013, Paris, France; Tel: 33 1 40 77 97 15; Fax: 33 1 40 77 97 89; E-mail:
| |
Collapse
|
115
|
Arukwe A, Ibor OR, Adeogun AO. Biphasic modulation of neuro- and interrenal steroidogenesis in juvenile African sharptooth catfish (Clarias gariepinus) exposed to waterborne di-(2-ethylhexyl) phthalate. Gen Comp Endocrinol 2017; 254:22-37. [PMID: 28919451 DOI: 10.1016/j.ygcen.2017.09.007] [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: 06/01/2017] [Revised: 09/04/2017] [Accepted: 09/13/2017] [Indexed: 12/27/2022]
Abstract
Receptor (i.e. genomic) and non-receptor (or non-genomic) effects of endocrine toxicology have received limited or almost non-existent attention for tropical species and regions. In the present study, we have evaluated the effects of di-(2-ethylhexyl) phthalate (DEHP) on neuro- and interrenal steroidogenesis of the African catfish (Clarias gariepinus) using molecular, immunochemical and physiological approaches. Juvenile fish (mean weight and length: 5.6±0.6g and 8.2±1.2cm, respectively), were randomly distributed into ten 120L rectangular glass tanks containing 60L of dechlorinated tap water, at 50 fish per exposure group. The fish were exposed to environmentally relevant concentrations of DEHP, consisting of 0 (ethanol solvent control), 10, 100, 200, and 400μg DEHP/L water and performed in two replicates. Brain, liver and head kidney samples were collected at day 3, 7 and 14 after exposure, and analysed for star, p450scc, cyp19a1, cyp17, cyp11β-, 3β-, 17β- and 20β-hsd, and 17β-ohase mRNA expression using real-time PCR. The StAR, P450scc and CYP19 proteins were measured using immunoblotting method, while estradiol-17β (E2) and testosterone (T) were measured in liver homogenate using enzyme immunoassay (EIA). Our data showed a consistent and unique pattern of biphasic effect on star and steroidogenic enzyme genes with increases at low concentration (10μg/L) and thereafter, a concentration-dependent decrease in both the brain and head kidney, that paralleled the expression of StAR, P450scc and CYP19 proteins. Cellular E2 and T levels showed an apparent DEHP concentration-dependent increase at day 14 of exposure. The observed consistency in the current findings and in view of previous reports on contaminants-induced alterations in neuro- and interrenal steroidogenesis, the broader toxicological and endocrine disruptor implication of our data indicate potentials for overt reproductive, metabolic, physiological and general health consequences for the exposed organisms.
Collapse
Affiliation(s)
- Augustine Arukwe
- Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, N-7491 Trondheim, Norway.
| | - Oju R Ibor
- Department of Zoology, University of Ibadan, Ibadan, Nigeria; Department of Zoology and Environmental Biology, University of Calabar, Calabar, Nigeria
| | - Aina O Adeogun
- Department of Zoology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
116
|
Veetil AT, Chakraborty K, Xiao K, Minter MR, Sisodia SS, Krishnan Y. Cell-targetable DNA nanocapsules for spatiotemporal release of caged bioactive small molecules. NATURE NANOTECHNOLOGY 2017; 12:1183-1189. [PMID: 28825714 DOI: 10.1038/nnano.2017.159] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/06/2017] [Indexed: 05/23/2023]
Abstract
Achieving triggered release of small molecules with spatial and temporal precision at designated cells within an organism remains a challenge. By combining a cell-targetable, icosahedral DNA-nanocapsule loaded with photoresponsive polymers, we show cytosolic delivery of small molecules with the spatial resolution of single endosomes in specific cells in Caenorhabditis elegans. Our technology can report on the extent of small molecules released after photoactivation as well as pinpoint the location at which uncaging of the molecules occurred. We apply this technology to release dehydroepiandrosterone (DHEA), a neurosteroid that promotes neurogenesis and neuron survival, and determined the timescale of neuronal activation by DHEA, using light-induced release of DHEA from targeted DNA nanocapsules. Importantly, sequestration inside the DNA capsule prevents photocaged DHEA from activating neurons prematurely. Our methodology can in principle be generalized to diverse neurostimulatory molecules.
Collapse
Affiliation(s)
- Aneesh T Veetil
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behaviour, University of Chicago, Chicago, Illinois 60637, USA
| | - Kasturi Chakraborty
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behaviour, University of Chicago, Chicago, Illinois 60637, USA
| | - Kangni Xiao
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behaviour, University of Chicago, Chicago, Illinois 60637, USA
| | - Myles R Minter
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behaviour, University of Chicago, Chicago, Illinois 60637, USA
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637, USA
- The Microbiome Center, University of Chicago, Chicago, Illinois 60637, USA
| | - Sangram S Sisodia
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behaviour, University of Chicago, Chicago, Illinois 60637, USA
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637, USA
- The Microbiome Center, University of Chicago, Chicago, Illinois 60637, USA
| | - Yamuna Krishnan
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behaviour, University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
117
|
Bazanova OM, Nikolenko ED, Barry RJ. Reactivity of alpha rhythms to eyes opening (the Berger effect) during menstrual cycle phases. Int J Psychophysiol 2017; 122:56-64. [DOI: 10.1016/j.ijpsycho.2017.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 03/30/2017] [Accepted: 05/01/2017] [Indexed: 02/02/2023]
|
118
|
Nguyen TV, Wu M, Lew J, Albaugh MD, Botteron KN, Hudziak JJ, Fonov VS, Collins DL, Campbell BC, Booij L, Herba C, Monnier P, Ducharme S, McCracken JT. Dehydroepiandrosterone impacts working memory by shaping cortico-hippocampal structural covariance during development. Psychoneuroendocrinology 2017; 86:110-121. [PMID: 28946055 PMCID: PMC5659912 DOI: 10.1016/j.psyneuen.2017.09.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 11/18/2022]
Abstract
Existing studies suggest that dehydroepiandrosterone (DHEA) may be important for human brain development and cognition. For example, molecular studies have hinted at the critical role of DHEA in enhancing brain plasticity. Studies of human brain development also support the notion that DHEA is involved in preserving cortical plasticity. Further, some, though not all, studies show that DHEA administration may lead to improvements in working memory in adults. Yet these findings remain limited by an incomplete understanding of the specific neuroanatomical mechanisms through which DHEA may impact the CNS during development. Here we examined associations between DHEA, cortico-hippocampal structural covariance, and working memory (216 participants [female=123], age range 6-22 years old, mean age: 13.6 +/-3.6 years, each followed for a maximum of 3 visits over the course of 4 years). In addition to administering performance-based, spatial working memory tests to these children, we also collected ecological, parent ratings of working memory in everyday situations. We found that increasingly higher DHEA levels were associated with a shift toward positive insular-hippocampal and occipito-hippocampal structural covariance. In turn, DHEA-related insular-hippocampal covariance was associated with lower spatial working memory but higher overall working memory as measured by the ecological parent ratings. Taken together with previous research, these results support the hypothesis that DHEA may optimize cortical functions related to general attentional and working memory processes, but impair the development of bottom-up, hippocampal-to-cortical connections, resulting in impaired encoding of spatial cues.
Collapse
Affiliation(s)
- Tuong-Vi Nguyen
- Department of Psychiatry, McGill University, Montreal, QC, H3A1A1, Canada; Department of Obstetrics-Gynecology, McGill University Health Center, Montreal, QC, H4A 3J1, Canada; Research Institute of the McGill University Health Center, Montreal, QC, H4A 3J1, Canada.
| | - Mia Wu
- Department of Psychology, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Jimin Lew
- Department of Psychology, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Matthew D Albaugh
- Department of Psychology, University of Vermont, College of Medicine, Burlington, VT, 05405, USA
| | - Kelly N Botteron
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA; Brain Development Cooperative Group, United States
| | - James J Hudziak
- Department of Psychology, University of Vermont, College of Medicine, Burlington, VT, 05405, USA; Brain Development Cooperative Group, United States
| | - Vladimir S Fonov
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada
| | - D Louis Collins
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada
| | - Benjamin C Campbell
- Department of Anthropology, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Linda Booij
- Department of Psychiatry, McGill University, Montreal, QC, H3A1A1, Canada; Department of Psychology, Concordia University, Montreal, QC, H4B 1R6, Canada; CHU Sainte Justine Hospital Research Centre, University of Montreal, Montreal, QC, H3T1C5, Canada
| | - Catherine Herba
- CHU Sainte Justine Hospital Research Centre, University of Montreal, Montreal, QC, H3T1C5, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, QC, Canada
| | - Patricia Monnier
- Department of Obstetrics-Gynecology, McGill University Health Center, Montreal, QC, H4A 3J1, Canada; Research Institute of the McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Simon Ducharme
- Department of Psychiatry, McGill University, Montreal, QC, H3A1A1, Canada; McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada; Department of Neurology & Neurosurgery, McGill University, Montreal, QC, H3A 1A1, Canada
| | - James T McCracken
- Brain Development Cooperative Group, United States; Department of Child and Adolescent Psychiatry, University of California in Los Angeles, Los Angeles, CA, 90024, USA
| |
Collapse
|
119
|
Miller WL. Steroidogenesis: Unanswered Questions. Trends Endocrinol Metab 2017; 28:771-793. [PMID: 29031608 DOI: 10.1016/j.tem.2017.09.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/05/2017] [Accepted: 09/05/2017] [Indexed: 01/06/2023]
Abstract
Until the mid-1980s studies of steroidogenesis largely depended on identifying steroid structures and measuring steroid concentrations in body fluids. The molecular biology revolution radically revolutionized studies of steroidogenesis with the cloning of known steroidogenic enzymes, by identifying novel factors, and delineating the genetic basis of known and newly discovered diseases. Unfortunately, this dramatic success has led many young research-oriented endocrinologists to regard steroidogenesis as a 'solved area'. However, many important and exciting questions remain, especially concerning the mechanisms of cholesterol delivery to the steroidogenic machinery, the biochemistry of androgen synthesis, the regulation and biological role of adrenarche, fetal adrenal development and involution, the roles of steroids made in 'extraglandular' cells, and the search for genetic disorders. This review outlines some of these questions, but this list is necessarily incomplete.
Collapse
Affiliation(s)
- Walter L Miller
- Center for Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA 94143-0556, USA.
| |
Collapse
|
120
|
Kalamarz-Kubiak H, Gozdowska M, Guellard T, Kulczykowska E. How does oestradiol influence the AVT/IT system in female round gobies during different reproductive phases? Biol Open 2017; 6:1493-1501. [PMID: 28860130 PMCID: PMC5665460 DOI: 10.1242/bio.024844] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this in vitro gradient perfusion study, we determined whether there is a functional relationship between oestradiol and the arginine vasotocin/isotocin (AVT/IT) system in the female round goby (Neogobius melanostomus). Brain explants were perfused in medium supplemented with 17β-oestradiol (E2) at doses mimicking the plasma levels of this hormone in nature during the spawning-capable phase and regressing phase. We aimed to establish which pathway, genomic or non-genomic, is involved in this mechanism in different reproductive phases. For this purpose, brain explants were perfused in medium supplemented with Fulvestrant (ICI 182.780) or Actinomycin D (Act D) separately or in combination with E2 The contents of AVT and IT in the perfusion media were determined using high-performance liquid chromatography (HPLC) with fluorescence and UV detection. During the spawning-capable phase, the effect of E2 on AVT release is mediated through oestrogen receptors (ERs) via both genomic and non-genomic pathways, while IT release is mediated through ERs via a genomic pathway only. In the regressing phase, release of both nonapeptides is mediated through ERs via a genomic pathway. This is the first study to present a feasible mechanism of oestradiol action on the AVT/IT system in female fish during different phases of the reproductive cycle.
Collapse
Affiliation(s)
- Hanna Kalamarz-Kubiak
- Department of Genetics and Marine Biotechnology, Institute of Oceanology Polish, Academy of Sciences, Powstańców Warszawy 55, 81-712 Sopot, Poland
| | - Magdalena Gozdowska
- Department of Genetics and Marine Biotechnology, Institute of Oceanology Polish, Academy of Sciences, Powstańców Warszawy 55, 81-712 Sopot, Poland
| | - Tatiana Guellard
- Department of Genetics and Marine Biotechnology, Institute of Oceanology Polish, Academy of Sciences, Powstańców Warszawy 55, 81-712 Sopot, Poland
| | - Ewa Kulczykowska
- Department of Genetics and Marine Biotechnology, Institute of Oceanology Polish, Academy of Sciences, Powstańców Warszawy 55, 81-712 Sopot, Poland
| |
Collapse
|
121
|
Acute inhibition of estradiol synthesis impacts vestibulo-ocular reflex adaptation and cerebellar long-term potentiation in male rats. Brain Struct Funct 2017; 223:837-850. [PMID: 28942480 DOI: 10.1007/s00429-017-1514-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/06/2017] [Indexed: 10/18/2022]
Abstract
The vestibulo-ocular reflex (VOR) adaptation is an ideal model for investigating how the neurosteroid 17 beta-estradiol (E2) contributes to the modification of behavior by regulating synaptic activities. We hypothesized that E2 impacts VOR adaptation by affecting cerebellar synaptic plasticity at the parallel fiber-Purkinje cell (PF) synapse. To verify this hypothesis, we investigated the acute effect of blocking E2 synthesis on gain increases and decreases in adaptation of the VOR in male rats using an oral dose (2.5 mg/kg) of the aromatase inhibitor letrozole. We also assessed the effect of letrozole on synaptic plasticity at the PF synapse in vitro, using cerebellar slices from male rats. We found that letrozole acutely impaired both gain increases and decreases adaptation of the VOR without altering basal ocular-motor performance. Moreover, letrozole prevented long-term potentiation at the PF synapse (PF-LTP) without affecting long-term depression (PF-LTD). Thus, in male rats neurosteroid E2 has a relevant impact on VOR adaptation and affects exclusively PF-LTP. These findings suggest that E2 might regulate changes in VOR adaptation by acting locally on cerebellar and extra-cerebellar synaptic plasticity sites.
Collapse
|
122
|
Korade Ž, Liu W, Warren EB, Armstrong K, Porter NA, Konradi C. Effect of psychotropic drug treatment on sterol metabolism. Schizophr Res 2017; 187:74-81. [PMID: 28202290 PMCID: PMC5554466 DOI: 10.1016/j.schres.2017.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/08/2023]
Abstract
Cholesterol metabolism is vital for brain function. Previous work in cultured cells has shown that a number of psychotropic drugs inhibit the activity of 7-dehydrocholesterol reductase (DHCR7), an enzyme that catalyzes the final steps in cholesterol biosynthesis. This leads to the accumulation of 7-dehydrocholesterol (7DHC), a molecule that gives rise to oxysterols, vitamin D, and atypical neurosteroids. We examined levels of cholesterol and the cholesterol precursors desmosterol, lanosterol, 7DHC and its isomer 8-dehydrocholesterol (8DHC), in blood samples of 123 psychiatric patients on various antipsychotic and antidepressant drugs, and 85 healthy controls, to see if the observations in cell lines hold true for patients as well. Three drugs, aripiprazole, haloperidol and trazodone increased circulating 7DHC and 8DHC levels, while five other drugs, clozapine, escitalopram/citalopram, lamotrigine, olanzapine, and risperidone, did not. Studies in rat brain verified that haloperidol dose-dependently increased 7DHC and 8DHC levels, while clozapine had no effect. We conclude that further studies should investigate the role of 7DHC and 8DHC metabolites, such as oxysterols, vitamin D, and atypical neurosteroids, in the deleterious and therapeutic effects of psychotropic drugs. Finally, we recommend that drugs that increase 7DHC levels should not be prescribed during pregnancy, as children born with DHCR7 deficiency have multiple congenital malformations.
Collapse
Affiliation(s)
- Željka Korade
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, UNMC, Omaha, NE 68198, United States
| | - Wei Liu
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Nashville, TN 37235, United States
| | - Emily B Warren
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240, United States
| | - Kristan Armstrong
- Department of Psychiatry, Vanderbilt University, Nashville, TN 37212, United States
| | - Ned A Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Nashville, TN 37235, United States
| | - Christine Konradi
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240, United States; Department of Psychiatry, Vanderbilt University, Nashville, TN 37212, United States.
| |
Collapse
|
123
|
Bonetto G, Charalampopoulos I, Gravanis A, Karagogeos D. The novel synthetic microneurotrophin BNN27 protects mature oligodendrocytes against cuprizone-induced death, through the NGF receptor TrkA. Glia 2017; 65:1376-1394. [PMID: 28567989 DOI: 10.1002/glia.23170] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/12/2017] [Accepted: 05/08/2017] [Indexed: 12/23/2022]
Abstract
BNN27, a member of a chemical library of C17-spiroepoxy derivatives of the neurosteroid DHEA, has been shown to regulate neuronal survival through its selective interaction with NGF receptors (TrkA and p75NTR ), but its role on glial populations has not been studied. Here, we present evidence that BNN27 provides trophic action (rescue from apoptosis), in a TrkA-dependent manner, to mature oligodendrocytes when they are challenged with the cuprizone toxin in culture. BNN27 treatment also increases oligodendrocyte maturation and diminishes microglia activation in vitro. The effect of BNN27 in the cuprizone mouse model of demyelination in vivo has also been investigated. In this model, that does not directly involve the adaptive immune system, BNN27 can protect from demyelination without affecting the remyelinating process. BNN27 preserves mature oligodendrocyte during demyelination, while reducing microgliosis and astrogliosis. Our findings suggest that BNN27 may serve as a lead molecule to develop neurotrophin-like blood-brain barrier (BBB)-permeable protective agents of oligodendrocyte populations and myelin, with potential applications in the treatment of demyelinating disorders.
Collapse
Affiliation(s)
- Giulia Bonetto
- Department of Basic Science, Faculty of Medicine, University of Crete, Crete, Greece
- Institute of Molecular Biology & Biotechnology - FoRTH, Heraklion, Crete, Greece
| | | | - Achille Gravanis
- Institute of Molecular Biology & Biotechnology - FoRTH, Heraklion, Crete, Greece
- Department of Pharmacology, Faculty of Medicine, University of Crete, Crete, Greece
| | - Domna Karagogeos
- Department of Basic Science, Faculty of Medicine, University of Crete, Crete, Greece
- Institute of Molecular Biology & Biotechnology - FoRTH, Heraklion, Crete, Greece
| |
Collapse
|
124
|
Botsakis K, Mourtzi T, Panagiotakopoulou V, Vreka M, Stathopoulos GT, Pediaditakis I, Charalampopoulos I, Gravanis A, Delis F, Antoniou K, Zisimopoulos D, Georgiou CD, Panagopoulos NT, Matsokis N, Angelatou F. BNN-20, a synthetic microneurotrophin, strongly protects dopaminergic neurons in the "weaver" mouse, a genetic model of dopamine-denervation, acting through the TrkB neurotrophin receptor. Neuropharmacology 2017; 121:140-157. [PMID: 28461162 DOI: 10.1016/j.neuropharm.2017.04.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 12/23/2022]
Abstract
Neurotrophic factors are among the most promising treatments aiming at slowing or stopping and even reversing Parkinson's disease (PD). However, in most cases, they cannot readily cross the human blood-brain-barrier (BBB). Herein, we propose as a therapeutic for PD the small molecule 17-beta-spiro-[5-androsten-17,2'-oxiran]-3beta-ol (BNN-20), a synthetic analogue of DHEA, which crosses the BBB and is deprived of endocrine side-effects. Using the "weaver" mouse, a genetic model of PD, which exhibits progressive dopaminergic neurodegeneration in the Substantia Nigra (SN), we have shown that long-term administration (P1-P21) of BNN-20 almost fully protected the dopaminergic neurons and their terminals, via i) a strong anti-apoptotic effect, probably mediated through the Tropomyosin receptor kinase B (TrkB) neurotrophin receptor's PI3K-Akt-NF-κB signaling pathway, ii) by exerting an efficient antioxidant effect, iii) by inducing significant anti-inflammatory activity and iv) by restoring Brain-Derived Neurotrophic Factor (BDNF) levels. By intercrossing "weaver" with NGL mice (dual GFP/luciferase-NF-κΒ reporter mice, NF-κΒ.GFP.Luc), we obtained Weaver/NGL mice that express the NF-κB reporter in all somatic cells. Acute BNN-20 administration to Weaver/NGL mice induced a strong NF-κB-dependent transcriptional response in the brain as detected by bioluminescence imaging, which was abolished by co-administration of the TrkB inhibitor ANA-12. This indicates that BNN-20 exerts its beneficial action (at least in part) through the TrkB-PI3K-Akt-NF-κB signaling pathway. These results could be of clinical relevance, as they suggest BNN-20 as an important neuroprotective agent acting through the TrkB neurotrophin receptor pathway, mimicking the action of the endogenous neurotrophin BDNF. Thus BNN-20 could be proposed for treatment of PD.
Collapse
Affiliation(s)
- Konstantinos Botsakis
- Department of Physiology, School of Medicine, University of Patras, Patras, 26 500, Greece
| | - Theodora Mourtzi
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Patras, 26 500, Greece
| | - Vasiliki Panagiotakopoulou
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Patras, 26 500, Greece
| | - Malamati Vreka
- Department of Physiology, School of Medicine, University of Patras, Patras, 26 500, Greece
| | | | - Iosif Pediaditakis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion 71110, Greece
| | | | - Achilleas Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion 71110, Greece; Institute of Molecular Biology & Biotechnology Foundation for Research & Technology - Hellas, GR, 70013, Heraklion, Crete, Greece
| | - Foteini Delis
- Department of Pharmacology, School of Medicine, University of Ioannina, Ioannina 45110, Greece
| | - Katerina Antoniou
- Department of Pharmacology, School of Medicine, University of Ioannina, Ioannina 45110, Greece
| | | | | | - Nikolaos T Panagopoulos
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Patras, 26 500, Greece
| | - Nikolaos Matsokis
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Patras, 26 500, Greece
| | - Fevronia Angelatou
- Department of Physiology, School of Medicine, University of Patras, Patras, 26 500, Greece.
| |
Collapse
|
125
|
Khakpay R, Azaddar M, Khakpay F, Hatami Nemati H. Analgesic Effect of 17β-Estradiol on Nucleus Paragigantocellularis Lateralis of Male Rats Mediated Via GABA A Receptors. Basic Clin Neurosci 2017; 8:51-60. [PMID: 28446950 PMCID: PMC5396174 DOI: 10.15412/j.bcn.03080107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Introduction: Beside its autonomic functions, the nucleus paragigantocellularis lateralis (LPGi) is involved in the descending pain modulation. 17β-Estradiol is a neuroactive steroid found in several brain areas such as LPGi. Intra-LPGi microinjection of 17β-estradiol can elicit the analgesic responses. 17β-Estradiol modulates nociception by binding to estrogenic receptors as well as allosteric interaction with other membrane-bound receptors like GABAA receptors. This study aimed to examine the role of GABAA receptors in the pain modulating effect of intra-LPGi injection of 17β-estradiol. Methods: To study the antinociceptive effects of 17β-estradiol, cannulation into the LPGi nucleus of male Wistar rats was performed. About 500 nL of drug was administered 15 minutes prior to formalin injection (50 μL of 4%). Then, formalin-induced flexing and licking behaviors were recorded for 60 minutes. For evaluating the role of GABAA receptors in the estradiol-induced pain modulation, 17β-estradiol was administered into the LPGi nucleus 15 minutes after the injection of 25 ng/μL bicuculline (the GABAA receptor antagonist). Then, the formalin-induced responses were recorded. Results: The results of the current study showed that intra-LPGi injection of 17β-estradiol decreased the flexing duration in both phases of formalin test (P<0.001); but it only attenuated the second phase of licking behavior (P<0.001). 17β-estradiol attenuated the second phase of formalin test of both behaviors (P<0.001). Bicuculline prevented the antinociceptive effect of intra-LPGi 17β-estradiol in both first and second phases of formalin-induced responses (P<0.001). Conclusion: According to the results of this study, the analgesic effect of intra-LPGi 17β-estradiol on the formalin-induced inflammatory pain might be mediated via GABAA receptors.
Collapse
Affiliation(s)
- Roghaieh Khakpay
- Department of Animal Science, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Maryam Azaddar
- Department of Animal Science, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Fatemeh Khakpay
- Department of Biology, Faculty of Basics Sciences, Varamin Branch, Islamic Azad University, Pishva, Iran
| | - Homeira Hatami Nemati
- Department of Animal Science, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
126
|
Zethraeus N, Dreber A, Ranehill E, Blomberg L, Labrie F, von Schoultz B, Johannesson M, Hirschberg AL. A first-choice combined oral contraceptive influences general well-being in healthy women: a double-blind, randomized, placebo-controlled trial. Fertil Steril 2017; 107:1238-1245. [PMID: 28433366 DOI: 10.1016/j.fertnstert.2017.02.120] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/28/2017] [Accepted: 02/24/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine whether there is a causal effect of oral contraceptive (OC) treatment on general well-being and depressed mood in healthy women. DESIGN Double-blind, randomized, and placebo-controlled trial. SETTING University hospital. PATIENT(S) Three hundred and forty healthy women aged 18-35 years randomized to treatment, of whom 332 completed the data collection at follow-up evaluation. INTERVENTION(S) A combined OC (150 μg levonorgestrel and 30 μg ethinylestradiol) or placebo for 3 months of treatment. MAIN OUTCOME MEASURE(S) Primary outcome measures: global score of Psychological General Well-Being Index (PGWBI) and the Beck Depression Inventory (BDI); secondary outcome measures: six separate dimensions of the PGWBI. RESULT(S) The OC treatment statistically significantly decreased general well-being compared with placebo -4.12 (95% CI, -7.18 to -1.06). Furthermore, OC decreased the following PGWBI dimensions compared with placebo: positive well-being -3.90 (95% CI, -7.78 to -0.01), self-control -6.63 (95% CI, -11.20 to -2.06), and vitality -6.84 (95% CI, -10.80 to -2.88). The effect of OC on depressive symptoms and on the PGWBI dimension depressed mood were not statistically significant. CONCLUSION(S) This study demonstrates a statistically significant reduction in general well-being by a first-choice OC in comparison with placebo in healthy women. We found no statistically significant effects on depressive symptoms. A reduction in general well-being should be of clinical importance.
Collapse
Affiliation(s)
- Niklas Zethraeus
- Department of Learning, Informatics, Management and Ethics, Karolinska Institutet, Stockholm, Sweden
| | - Anna Dreber
- Department of Economics, Stockholm School of Economics, Stockholm, Sweden
| | - Eva Ranehill
- Center for Economic Research, ETH, Zurich, Switzerland; Department of Economics, University of Gothenburg, Gothenburg, Sweden
| | - Liselott Blomberg
- Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Bo von Schoultz
- Department of Economics, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Johannesson
- Department of Economics, Stockholm School of Economics, Stockholm, Sweden
| | - Angelica Lindén Hirschberg
- Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
127
|
Zhang M, Liu J, Zhou MM, Wu H, Hou Y, Li YF, Yin Y, Zheng L, Cai J, Liao FF, Liu FY, Yi M, Wan Y. Anxiolytic effects of hippocampal neurosteroids in normal and neuropathic rats with spared nerve injury. J Neurochem 2017; 141:137-150. [PMID: 28129443 DOI: 10.1111/jnc.13965] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/16/2016] [Accepted: 01/11/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Meng Zhang
- Neuroscience Research Institute; Peking University; Beijing China
- Department of Pathology; Beijing Children's Hospital, Capital Medical University; Beijing China
| | - Jia Liu
- Institute of Systems Biomedicine; Peking University; Beijing China
| | - Meng-Meng Zhou
- Neuroscience Research Institute; Peking University; Beijing China
| | - Honghai Wu
- Department of Pharmacy; Bethune International Peace Hospital; Shijiazhuang China
| | - Yanning Hou
- Department of Pharmacy; Bethune International Peace Hospital; Shijiazhuang China
| | - Yun-Feng Li
- Department of New Drug Evaluation; Beijing Institute of Pharmacology and Toxicology; Beijing China
| | - Yuxin Yin
- Institute of Systems Biomedicine; Peking University; Beijing China
| | - Lemin Zheng
- Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education; Peking University; Beijing China
| | - Jie Cai
- Neuroscience Research Institute; Peking University; Beijing China
| | - Fei-Fei Liao
- Neuroscience Research Institute; Peking University; Beijing China
| | - Feng-Yu Liu
- Neuroscience Research Institute; Peking University; Beijing China
| | - Ming Yi
- Neuroscience Research Institute; Peking University; Beijing China
| | - You Wan
- Neuroscience Research Institute; Peking University; Beijing China
- Key Laboratory for Neuroscience; Ministry of Education/National Health and Family Planning Commission; Peking University; Beijing China
- Department of Neurobiology; School of Basic Medical Sciences; Peking University; Beijing China
| |
Collapse
|
128
|
Afroz S, Shen H, Smith SS. α4βδ GABA A receptors reduce dendritic spine density in CA1 hippocampus and impair relearning ability of adolescent female mice: Effects of a GABA agonist and a stress steroid. Neuroscience 2017; 347:22-35. [PMID: 28189613 DOI: 10.1016/j.neuroscience.2017.01.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/22/2017] [Accepted: 01/31/2017] [Indexed: 01/10/2023]
Abstract
Synaptic pruning underlies the transition from an immature to an adult CNS through refinements of neuronal circuits. Our recent study indicates that pubertal synaptic pruning is triggered by the inhibition generated by extrasynaptic α4βδ GABAA receptors (GABARs) which are increased for 10 d on dendritic spines of CA1 pyramidal cells at the onset of puberty (PND 35-44) in the female mouse, suggesting α4βδ GABARs as a novel target for the regulation of adolescent synaptic pruning. In the present study we used a pharmacological approach to further examine the role of these receptors in altering spine density during puberty of female mice and the impact of these changes on spatial learning, assessed in adulthood. Two drugs were chronically administered during the pubertal period (PND 35-44): the GABA agonist gaboxadol (GBX, 0.1mg/kg, i.p.), to enhance current gated by α4βδ GABARs and the neurosteroid/stress steroid THP (3α-OH-5β-pregnan-20-one, 10mg/kg, i.p.) to decrease expression of α4βδ. Spine density was determined on PND 56 with Golgi staining. Spatial learning and relearning were assessed using the multiple object relocation task and an active place avoidance task on PND 56. Pubertal GBX decreased spine density post-pubertally by 70% (P<0.05), while decreasing α4βδ expression with THP increased spine density by twofold (P<0.05), in both cases, with greatest effects on the mushroom spines. Adult relearning ability was compromised in both hippocampus-dependent tasks after pubertal administration of either drug. These findings suggest that an optimal spine density produced by α4βδ GABARs is necessary for optimal cognition in adults.
Collapse
Affiliation(s)
- Sonia Afroz
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA
| | - Hui Shen
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA; School of Biomedical Engineering, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| |
Collapse
|
129
|
van Rooyen D, du Toit T, Louw-du Toit R, Africander D, Swart P, Swart AC. The metabolic fate and receptor interaction of 16α-hydroxyprogesterone and its 5α-reduced metabolite, 16α-hydroxy-dihydroprogesterone. Mol Cell Endocrinol 2017; 441:86-98. [PMID: 27664517 DOI: 10.1016/j.mce.2016.09.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/17/2016] [Accepted: 09/21/2016] [Indexed: 01/14/2023]
Abstract
16α-hydroxyprogesterone (16OHP4) is not well characterised in terms of metabolism and receptor interaction. We therefore investigated its metabolism by adrenal CYP11B and peripheral steroidogenic enzymes, SRD5A and AKR1C2. UHPLC-MS/MS analyses identified novel steroids: the biosynthesis of 4-pregnen-11β,16α-diol-3,20-dione catalysed by CYP11B2; the 5α-reduction of the latter and 16OHP4 catalysed by SRD5A yielding 5α-pregnan-11β,16α-diol-3,20-diovne and 5α-pregnan-16α-ol-3,20-dione (16OH-DHP4); and 16OH-DHP4 converted by AKR1C2 to 5α-pregnan-3α,16α-diol-20-one. Receptor studies showed 16OHP4, 16OH-DHP4, progesterone and dihydroprogesterone (DHP4) were weak partial AR agonists; 16OHP4, 16OH-DHP4 and DHP4 exhibited weak partial agonist activity towards PR-B with DHP4 also exhibiting partial agonist activity towards PR-A. Data showed that while the 5α-reduction of P4 decreased PR activation significantly, 16OHP4 and 16OH-DHP4 exhibited comparable receptor activation. Although the clinical relevance of 16OHP4 remains unclear the elevated 16OHP4 levels characteristic of 21OHD, CAH, PCOS, prostate cancer, testicular feminization syndrome and cryptorchidism likely contribute towards these clinical conditions, inducing receptor-activated target genes.
Collapse
Affiliation(s)
- Desmaré van Rooyen
- Biochemistry Department, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Therina du Toit
- Biochemistry Department, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Renate Louw-du Toit
- Biochemistry Department, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Donita Africander
- Biochemistry Department, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Pieter Swart
- Biochemistry Department, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Amanda C Swart
- Biochemistry Department, Stellenbosch University, Stellenbosch 7600, South Africa.
| |
Collapse
|
130
|
Di Mauro M, Tozzi A, Calabresi P, Pettorossi VE, Grassi S. Different synaptic stimulation patterns influence the local androgenic and estrogenic neurosteroid availability triggering hippocampal synaptic plasticity in the male rat. Eur J Neurosci 2017; 45:499-509. [DOI: 10.1111/ejn.13455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Michela Di Mauro
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| | - Alessandro Tozzi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
- Fondazione Santa Lucia – I.R.C.C.S. Rome Italy
| | - Paolo Calabresi
- Fondazione Santa Lucia – I.R.C.C.S. Rome Italy
- Dipartimento di Medicina Clinica Neurologica Università di Perugia Perugia Italy
| | - Vito Enrico Pettorossi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| | - Silvarosa Grassi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| |
Collapse
|
131
|
Piekarski DJ, Johnson CM, Boivin JR, Thomas AW, Lin WC, Delevich K, M Galarce E, Wilbrecht L. Does puberty mark a transition in sensitive periods for plasticity in the associative neocortex? Brain Res 2017; 1654:123-144. [PMID: 27590721 PMCID: PMC5283387 DOI: 10.1016/j.brainres.2016.08.042] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/17/2016] [Accepted: 08/29/2016] [Indexed: 02/08/2023]
Abstract
Postnatal brain development is studded with sensitive periods during which experience dependent plasticity is enhanced. This enables rapid learning from environmental inputs and reorganization of cortical circuits that matches behavior with environmental contingencies. Significant headway has been achieved in characterizing and understanding sensitive period biology in primary sensory cortices, but relatively little is known about sensitive period biology in associative neocortex. One possible mediator is the onset of puberty, which marks the transition to adolescence, when animals shift their behavior toward gaining independence and exploring their social world. Puberty onset correlates with reduced behavioral plasticity in some domains and enhanced plasticity in others, and therefore may drive the transition from juvenile to adolescent brain function. Pubertal onset is also occurring earlier in developed nations, particularly in unserved populations, and earlier puberty is associated with vulnerability for substance use, depression and anxiety. In the present article we review the evidence that supports a causal role for puberty in developmental changes in the function and neurobiology of the associative neocortex. We also propose a model for how pubertal hormones may regulate sensitive period plasticity in associative neocortex. We conclude that the evidence suggests puberty onset may play a causal role in some aspects of associative neocortical development, but that further research that manipulates puberty and measures gonadal hormones is required. We argue that further work of this kind is urgently needed to determine how earlier puberty may negatively impact human health and learning potential. This article is part of a Special Issue entitled SI: Adolescent plasticity.
Collapse
Affiliation(s)
- David J Piekarski
- Department of Psychology, University of California, Berkeley, Berkeley CA 94720, USA
| | - Carolyn M Johnson
- Department of Psychology, University of California, Berkeley, Berkeley CA 94720, USA
| | - Josiah R Boivin
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco CA 94158, USA
| | - A Wren Thomas
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley CA 94720, USA
| | - Wan Chen Lin
- Department of Psychology, University of California, Berkeley, Berkeley CA 94720, USA
| | - Kristen Delevich
- Department of Psychology, University of California, Berkeley, Berkeley CA 94720, USA
| | - Ezequiel M Galarce
- School of Public Health, University of California, Berkeley, Berkeley CA 94720, USA
| | - Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, Berkeley CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley CA 94720, USA.
| |
Collapse
|
132
|
Garay L, Gonzalez Giqueaux P, Guennoun R, Schumacher M, Gonzalez Deniselle MC, De Nicola AF. Progesterone treatment modulates mRNA OF neurosteroidogenic enzymes in a murine model of multiple sclerosis. J Steroid Biochem Mol Biol 2017; 165:421-429. [PMID: 27597394 DOI: 10.1016/j.jsbmb.2016.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 11/20/2022]
Abstract
Previous studies of experimental autoimmune encephalomyelitis (EAE) have shown that progesterone decreases inflammatory cell infiltration and proinflammatory factors, increases myelination and attenuates clinical grade of EAE mice. To elucidate potential mediators of these effects, we analyzed the mRNA expression of neurosteroidogenic enzymes in the spinal cord, in view of the protective role of steroids in EAE. We also analyzed mitochondrial morphology and dynamics (fusion and fission proteins), considering the role of mitochondria in neurosteroidogenesis. EAE was induced in C57Bl6 mice using MOG40-54 and killed on day 16 after induction. Using qPCR, we found in steroid-untreated EAE mice decreased mRNAs for the steroidogenic acute regulatory protein (Star), voltage-dependent anion channel (VDAC), P450scc (cholesterol side-chain cleavage), 5α-reductase, 3α-hydroxysteroid dehydrogenase (3α-HSD) and aromatase, whereas levels of 3β-hydroxysteroid dehydrogenase (3β-HSD) showed a large intra-group variance. We also found increased mRNA expression of 18Kd translocator protein (TSPO), which likely resulted from the reactive microgliosis in this model. EAE mice also showed pathological mitochondrial morphology and reduced expression of fission and fusion protein mRNAs. Most importantly, pretreatment with progesterone a week before EAE induction increased Star,VDAC, P450scc, 5α-reductase type I, 3α-HSD and aromatase mRNAs and did not modify 3β-HSD. TSPO mRNA was decreased, consequent with the inhibition of microgliosis. Mitochondrial morphology was improved and fission/fusion protein mRNAs were enhanced by progesterone treatment. Furthermore, progesterone protective effects on mitochondrial and endoplasmic reticulum may allow the recovery of neurosteroidogenesis. In this way, endogenously synthesized neurosteroids may reinforce the beneficial effects of exogenous progesterone previously shown in MS mice.
Collapse
Affiliation(s)
- Laura Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry(,) Faculty of Medicine, University of Buenos Aires, Argentina
| | - Paula Gonzalez Giqueaux
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Physiology, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry(,) Faculty of Medicine, University of Buenos Aires, Argentina.
| |
Collapse
|
133
|
Zethraeus N, Dreber A, Ranehill E, Blomberg L, Labrie F, von Schoultz B, Johannesson M, Hirschberg AL. Combined Oral Contraceptives and Sexual Function in Women-a Double-Blind, Randomized, Placebo-Controlled Trial. J Clin Endocrinol Metab 2016; 101:4046-4053. [PMID: 27525531 DOI: 10.1210/jc.2016-2032] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
CONTEXT There is a lack of knowledge about how oral contraceptives may affect sexual function. OBJECTIVE To determine whether there is a causal effect of oral contraceptives on sexuality. We hypothesized that a widely used pill impairs sexuality. DESIGN A double-blind, randomized, placebo-controlled trial. Enrollment began in February 2012 and was completed in August 2015. SETTING Karolinska University Hospital, Stockholm, Sweden. PARTICIPANTS A total of 340 healthy women, aged 18-35 years, were randomized to treatment, and 332 completed the study. INTERVENTIONS A combined oral contraceptive (150 μg levonorgestrel and 30 μg ethinylestradiol) or placebo for 3 months of treatment. MAIN OUTCOME MEASURES The primary outcome was the aggregate score on the Profile of Female Sexual Function (PFSF). Secondary outcomes were the seven domains of the PFSF, the Sexual Activity Log, and the Personal Distress Scale. RESULTS Overall sexual function was similar in women in the oral contraceptive and placebo groups. The PFSF domains desire (-4.4; 95% confidence interval [CI], -8.49 to -0.38; P = .032), arousal (-5.1; 95% CI, -9.63 to -0.48; P = .030), and pleasure (-5.1; 95% CI, -9.97 to -0.32; P = .036) were significantly reduced in comparison to placebo, whereas orgasm, concern, responsiveness, and self-image were similar between groups. The mean frequency of satisfying sexual episodes and personal distress were also similar between groups. CONCLUSIONS This study shows no negative impact of a levonorgestrel-containing oral contraceptive on overall sexual function, although three of seven sexual function domains were adversely affected.
Collapse
Affiliation(s)
- Niklas Zethraeus
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Anna Dreber
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Eva Ranehill
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Liselott Blomberg
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Fernand Labrie
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Bo von Schoultz
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Magnus Johannesson
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| | - Angelica Lindén Hirschberg
- Department of Learning, Informatics, Management and Ethics (N.Z.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Economics (A.D., M.J.), Stockholm School of Economics, 113 83 Stockholm, Sweden; Department of Economics (E.R.), University of Zurich, 8006 Zurich, Switzerland; Department of Women's and Children's Health (L.B., B.v.S., A.L.H), Karolinska Institutet, and Department of Obstetrics and Gynecology, Karolinska University Hospital, 171 76 Stockholm, Sweden; and Laval University (F.L.), Quebec City G1V 0A6, Canada
| |
Collapse
|
134
|
Pleil KE, Helms CM, Sobus JR, Daunais JB, Grant KA, Kash TL. Effects of chronic alcohol consumption on neuronal function in the non-human primate BNST. Addict Biol 2016. [PMID: 26223349 DOI: 10.1111/adb.12289] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alterations in hypothalamic-pituitary-adrenal axis function contribute to many of the adverse behavioral effects of chronic voluntary alcohol drinking, including alcohol dependence and mood disorders; limbic brain structures such as the bed nucleus of the stria terminalis (BNST) may be key sites for these effects. Here, we measured circulating levels of several steroid hormones and performed whole-cell electrophysiological recordings from acutely prepared BNST slices of male rhesus monkeys allowed to self-administer alcohol for 12 months or a control solution. Initial comparisons revealed that BNST neurons in alcohol-drinking monkeys had decreased membrane resistance, increased frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) with no change in spontaneous excitatory postsynaptic currents (sEPSCs). We then used a combined variable cluster analysis and linear mixed model statistical approach to determine whether specific factors including stress and sex hormones, age and measures of alcohol consumption and intoxication are related to these BNST measures. Modeling results showed that specific measures of alcohol consumption and stress-related hormone levels predicted differences in membrane conductance in BNST neurons. Distinct groups of adrenal stress hormones were negatively associated with the frequency of sIPSCs and sEPSCs, and alcohol drinking measures and basal neuronal membrane properties were additional positive predictors of inhibitory, but not excitatory, PSCs. The amplitude of sEPSCs was highly positively correlated with age, independent of other variables. Together, these results suggest that chronic voluntary alcohol consumption strongly influences limbic function in non-human primates, potentially via interactions with or modulation by other physiological variables, including stress steroid hormones and age.
Collapse
Affiliation(s)
- Kristen E. Pleil
- Bowles Center for Alcohol Studies & Department of Pharmacology, School of Medicine; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Christa M. Helms
- Division of Neuroscience, Oregon National Primate Research Center; Oregon Health & Science University; Beaverton OR USA
| | - Jon R. Sobus
- Human Exposure and Atmospheric Sciences Division, National Exposure Research Laboratory, Office of Research and Development; U.S. Environmental Protection Agency; Research Triangle Park NC USA
| | - James B. Daunais
- Department of Physiology & Pharmacology; Wake Forest School of Medicine; Winston-Salem NC USA
| | - Kathleen A. Grant
- Division of Neuroscience, Oregon National Primate Research Center; Oregon Health & Science University; Beaverton OR USA
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies & Department of Pharmacology, School of Medicine; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| |
Collapse
|
135
|
Glajch KE, Ferraiuolo L, Mueller KA, Stopford MJ, Prabhkar V, Gravanis A, Shaw PJ, Sadri-Vakili G. MicroNeurotrophins Improve Survival in Motor Neuron-Astrocyte Co-Cultures but Do Not Improve Disease Phenotypes in a Mutant SOD1 Mouse Model of Amyotrophic Lateral Sclerosis. PLoS One 2016; 11:e0164103. [PMID: 27716798 PMCID: PMC5055348 DOI: 10.1371/journal.pone.0164103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/20/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease caused by loss of motor neurons. ALS patients experience rapid deterioration in muscle function with an average lifespan of 3–5 years after diagnosis. Currently, the most effective therapeutic only extends lifespan by a few months, thus highlighting the need for new and improved therapies. Neurotrophic factors (NTFs) are important for neuronal development, maintenance, and survival. NTF treatment has previously shown efficacy in pre-clinical ALS models. However, clinical trials using NTFs produced no major improvements in ALS patients, due in part to the limited blood brain barrier (BBB) penetration. In this study we assessed the potential neuroprotective effects of a novel class of compounds known as MicroNeurotrophins (MNTs). MNTs are derivatives of Dehydroepiandrosterone (DHEA), an endogenous neurosteroid that can cross the BBB and bind to tyrosine kinase receptors mimicking the pro-survival effects of NTFs. Here we sought to determine whether MNTs were neuroprotective in two different models of ALS. Our results demonstrate that BNN27 (10 μM) attenuated loss of motor neurons co-cultured with astrocytes derived from human ALS patients with SOD1 mutations via the reduction of oxidative stress. Additionally, in the G93A SOD1 mouse, BNN27 (10 mg/kg) treatment attenuated motor behavioral impairment in the paw grip endurance and rotarod tasks at postnatal day 95 in female but not male mice. In contrast, BNN27 (10 mg/kg and 50 mg/kg) treatment did not alter any other behavioral outcome or neuropathological marker in male or female mice. Lastly, BNN27 was not detected in post-mortem brain or spinal cord tissue of treated mice due to the rapid metabolism of BNN27 by mouse hepatocytes relative to human hepatocytes. Together, these findings demonstrate that BNN27 treatment failed to yield significant neuroprotective effects in the G93A SOD1 model likely due to its rapid rate of metabolism in mice.
Collapse
Affiliation(s)
- Kelly E. Glajch
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Laura Ferraiuolo
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Kaly A. Mueller
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Matthew J. Stopford
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Varsha Prabhkar
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Institute of Molecular Biology & Biotechnology-FORTH, Heraklion 71003, Greece
| | - Pamela J. Shaw
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Ghazaleh Sadri-Vakili
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
- * E-mail:
| |
Collapse
|
136
|
Jin RO, Mason S, Mellon SH, Epel ES, Reus VI, Mahan L, Rosser RL, Hough CM, Burke HM, Mueller SG, Wolkowitz OM. Cortisol/DHEA ratio and hippocampal volume: A pilot study in major depression and healthy controls. Psychoneuroendocrinology 2016; 72:139-46. [PMID: 27428086 PMCID: PMC5203799 DOI: 10.1016/j.psyneuen.2016.06.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 11/19/2022]
Abstract
Structural imaging studies investigating the relationship between hippocampal volume (HCV) and peripheral measures of glucocorticoids (GCs) have produced conflicting results in both normal populations and in individuals with MDD, raising the possibility of other modulating factors. In preclinical studies, dehydroepiandrosterone (DHEA) and its sulfate ester (DHEAS; together abbreviated, DHEA(S)) have been shown to antagonize the actions of GCs on the central nervous system. Therefore, considering the relationship of HCV to both of these hormones simultaneously may be important, although it has rarely been done in human populations. Using high-resolution magnetic resonance imaging (MRI), the present pilot study examined the relationship between morning serum cortisol, DHEA(S), and HCV in nineteen normal controls and eighteen unmedicated subjects with Major Depressive Disorder (MDD). Serum cortisol and DHEA(S) were not significantly correlated with HCV across all subjects (cortisol: r=-0.165, p=0.33; DHEA: r=0.164, p=0.35; DHEAS: r=0.211, p=0.22, respectively). However, the ratios of cortisol/DHEA(S) were significantly negatively correlated with HCV in combined group (Cortisol/DHEA: r=-0.461, p=0.005; Cortisol/DHEAS: r=-0.363, p=0.03). Significant or near-significant correlations were found between some hormonal measurements and HCV in the MDDs alone (DHEA: r=0.482, p=0.059; DHEAS: r=0.507, p=0.045; cort/DHEA: r=-0.589, p=0.02; cort/DHEAS: r=-0.424p=0.10), but not in the controls alone (DHEA: r=0.070, p=0.79; DHEAS: r=0.077, p=0.77; cort/DHEA: r=-0.427, p=0.09; cort/DHEAS: r=-0.331, p=0.19). However, Group (MDDs vs controls) did not have a significant effect on the relationship between cortisol, DHEA(S), and their ratios with HCV (p>0.475 in all analyses). Although the exact relationship between serum and central steroid concentrations as well as their effects on the human hippocampus remains not known, these preliminary results suggest that the ratio of cortisol to DHEA(S), compared to serum cortisol alone, may convey additional information about "net steroid activity" with relation to HCV.
Collapse
Affiliation(s)
- Rowen O Jin
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Sara Mason
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Synthia H Mellon
- Department of Obstetrics-Gynecology and Reproductive Sciences, University of California, San Francisco, School of Medicine California, 94143, United States
| | - Elissa S Epel
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Victor I Reus
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Laura Mahan
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Rebecca L Rosser
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Christina M Hough
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Heather M Burke
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States
| | - Susanne G Mueller
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, School of Medicine California, 94143, United States
| | - Owen M Wolkowitz
- Department of Psychiatry, University of California, San Francisco, School of Medicine, CA, 94143, United States.
| |
Collapse
|
137
|
Selvaraj V, Tu LN. Current status and future perspectives: TSPO in steroid neuroendocrinology. J Endocrinol 2016; 231:R1-R30. [PMID: 27422254 DOI: 10.1530/joe-16-0241] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022]
Abstract
The mitochondrial translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor (PBR), has received significant attention both as a diagnostic biomarker and as a therapeutic target for different neuronal disease pathologies. Recently, its functional basis believed to be mediating mitochondrial cholesterol import for steroid hormone production has been refuted by studies examining both in vivo and in vitro genetic Tspo-deficient models. As a result, there now exists a fundamental gap in the understanding of TSPO function in the nervous system, and its putative pharmacology in neurosteroid production. In this review, we discuss several recent findings in steroidogenic cells that are in direct contradiction to previous studies, and necessitate a re-examination of the purported role for TSPO in de novo neurosteroid biosynthesis. We critically examine the pharmacological effects of different TSPO-binding drugs with particular focus on studies that measure neurosteroid levels. We highlight the basis of key misconceptions regarding TSPO that continue to pervade the literature, and the need for interpretation with caution to avoid negative impacts. We also summarize the emerging perspectives that point to new directions that need to be investigated for understanding the molecular function of TSPO, only after which the true potential of this therapeutic target in medicine may be realized.
Collapse
Affiliation(s)
- Vimal Selvaraj
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Lan N Tu
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| |
Collapse
|
138
|
Black KL, Witty CF, Daniel JM. Previous Midlife Oestradiol Treatment Results in Long-Term Maintenance of Hippocampal Oestrogen Receptor α Levels in Ovariectomised Rats: Mechanisms and Implications for Memory. J Neuroendocrinol 2016; 28:10.1111/jne.12429. [PMID: 27603028 PMCID: PMC5527336 DOI: 10.1111/jne.12429] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 01/28/2023]
Abstract
Ovariectomised rats that have received previous administration of oestradiol in midlife display enhanced cognition and increased hippocampal levels of oestrogen receptor (ER)α months after oestradiol treatment ended compared to ovariectomised controls. The present study aimed to investigate the mechanisms by which ERα levels are maintained following midlife oestradiol exposure and the role of ERα in memory in ageing females in the absence of circulating oestrogens. Unliganded ERα has increased interaction with the ubiquitin ligase, C-terminus of Hsc-70 interacting protein (CHIP), leading to increased degradation of the receptor. In our first experiment, we tested the hypothesis that midlife oestradiol exposure in ovariectomised rats results in decreased interaction between CHIP and hippocampal ERα, leading to increased levels of ERα. Middle-aged rats were ovariectomised and received oestradiol or vehicle implants. After 40 days, implants were removed. One month later, rats were killed and hippocampi were processed for whole protein western blotting and co-immunoprecipitation, in which ERα was immunoprecipitated from lysate. As expected, ERα protein expression was increased in rats previously treated with oestradiol compared to vehicle-treated rats. In rats treated with oestradiol, there was a decrease in CHIP-ERα interaction, suggesting that previous oestradiol treatment reduces interaction, slowing the degradation of ERα. In a second experiment, we determined the impact on memory of antagonism of ER in the absence of circulating oestrogens. Rats were ovariectomised and implanted with oestradiol capsules. Capsules were removed after 40 days. Rats received chronic i.c.v. infusion of ER antagonist, ICI 182 780, or artificial cerebrospinal fluid vehicle and were tested on a spatial memory radial-maze task. Rats treated with ICI 182 780 had significantly worse performance (more errors). These experiments provide evidence that previous midlife oestradiol treatment maintains hippocampal ERα by decreasing its interaction with CHIP and that activation of these receptors provides cognitive benefits in the absence of circulating oestrogens.
Collapse
Affiliation(s)
- K L Black
- Program in Neuroscience, Tulane University, New Orleans, LA, USA
- Brain Institute, Tulane University, New Orleans, LA, USA
| | - C F Witty
- Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | - J M Daniel
- Program in Neuroscience, Tulane University, New Orleans, LA, USA.
- Brain Institute, Tulane University, New Orleans, LA, USA.
- Department of Psychology, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
139
|
Ago Y, Hasebe S, Hiramatsu N, Mori K, Watabe Y, Onaka Y, Hashimoto H, Takuma K, Matsuda T. Involvement of GABAA receptors in 5-HT1A and σ1 receptor synergism on prefrontal dopaminergic transmission under circulating neurosteroid deficiency. Psychopharmacology (Berl) 2016; 233:3125-34. [PMID: 27339616 DOI: 10.1007/s00213-016-4353-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/09/2016] [Indexed: 12/29/2022]
Abstract
RATIONALE We previously reported that the fluvoxamine-induced increase in prefrontal dopamine levels is enhanced by adrenalectomy/castration (which results in circulating neurosteroid deficiency), via combined activation of serotonin1A (5-HT1A) and σ1 receptors. However, the mechanistic details of the interaction between 5-HT1A and σ1 receptors are unknown. OBJECTIVES Because most neurosteroids have affinity for γ-aminobutyric acid (GABA)A receptors, in the present study, we examined the involvement of GABAA receptors in this process. RESULTS Adrenalectomy/castration decreased pentobarbital-induced sleeping time in mice, suggesting that it reduced GABAA receptor function. The GABAA receptor antagonist picrotoxin (1 mg/kg) enhanced the fluvoxamine-induced increase in prefrontal dopamine, but not noradrenaline or serotonin, levels in mice, suggesting that picrotoxin mimicked the effect of adrenalectomy/castration. Picrotoxin also potentiated the increase in prefrontal dopamine levels mediated by co-administration of the 5-HT1A receptor agonist osemozotan and the σ1 receptor agonist (+)-SKF-10,047, while it did not affect the co-administration-induced changes in noradrenaline and serotonin levels. Conversely, the GABAA receptor agonist diazepam (1 mg/kg) blocked the effect of adrenalectomy/castration on the fluvoxamine-induced increase in prefrontal dopamine levels. Co-administration of osemozotan and (+)-SKF-10,047 did not affect the expression of the neuronal activity marker c-Fos in the prefrontal cortex, ventral tegmental area, and nucleus accumbens in control mice, while it increased the c-Fos expression only in the prefrontal cortex and ventral tegmental area in picrotoxin-treated mice. CONCLUSIONS These results suggest that the GABAA receptor plays a key role in mediating the synergistic effects of 5-HT1A and σ1 receptor activation on prefrontal dopamine neurotransmission.
Collapse
Affiliation(s)
- Yukio Ago
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Hasebe
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Naoki Hiramatsu
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kazuya Mori
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yuji Watabe
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Onaka
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Toshio Matsuda
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
140
|
Nguyen TV, Gower P, Albaugh MD, Botteron KN, Hudziak JJ, Fonov VS, Collins L, Ducharme S, McCracken JT. The developmental relationship between DHEA and visual attention is mediated by structural plasticity of cortico-amygdalar networks. Psychoneuroendocrinology 2016; 70:122-33. [PMID: 27236606 PMCID: PMC4907862 DOI: 10.1016/j.psyneuen.2016.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/25/2016] [Accepted: 05/04/2016] [Indexed: 01/18/2023]
Abstract
Humans and the great apes are the only species demonstrated to exhibit adrenarche, a key developmental event leading to increased production of dehydroepiandrosterone (DHEA), suggesting that this hormone may play an important evolutionary role. Similarly, visual attention networks have been shown to evolve in a human-specific manner, with some anatomical connections and elements of cortical organization exclusive to our species. Existing studies of human brain development support the notion that DHEA shows significant uptake in cortical structures and the amygdala, and as such, could be involved in the bottom-up regulation of visual attention. Here we examined associations between DHEA, structural covariance of the amygdala with whole-brain cortical thickness, and tests of visual attention, in a longitudinal sample of typically developing children and adolescents 6-22 years of age. We found that DHEA predicted covariance between amygdalar volume and the left occipital pole, right somatosensory parietal cortex and right anterior cingulate cortex. Amygdala-occipital covariance predicted visual awareness; amygdala-parietal covariance predicted visuo-motor dexterity and processing speed; amygdala-prefrontal covariance predicted global attentional impairment. Further, effects of DHEA were above and beyond those of age and sex, as well as distinct from those of pubertal stage, estradiol and testosterone. These findings support the notion that DHEA may play a unique role in shaping amygdala-dependent cortical plasticity and in regulating 'bottom-up' visual attention processes from childhood to young adulthood.
Collapse
Affiliation(s)
- Tuong-Vi Nguyen
- Department of Psychiatry and Department of Obstetrics-Gynecology, McGill University Health Center (Royal Victoria Hospital at the Glen site), McGill University, Montreal, QC H4A 3J1, Canada.
| | - Patricia Gower
- Department of Psychology, McGill University, Montreal, QC, Canada, H4A 3J1
| | - Matthew D Albaugh
- University of Vermont, College of Medicine, Burlington, VT, USA, 05405
| | - Kelly N Botteron
- Washington University School of Medicine, St. Louis, MO, USA, 63110,Brain Development Cooperative Group
| | - James J Hudziak
- University of Vermont, College of Medicine, Burlington, VT, USA, 05405,Brain Development Cooperative Group
| | - Vladimir S Fonov
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC Canada H3A 2B4
| | - Louis Collins
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC Canada H3A 2B4
| | - Simon Ducharme
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC Canada H3A 2B4,McGill University Health Centre, Department of Psychiatry and Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada, H3A 1A1
| | - James T McCracken
- Brain Development Cooperative Group,Department of Child and Adolescent Psychiatry, University of California in Los Angeles, Los Angeles, CA, USA, 90024
| |
Collapse
|
141
|
Rossetti MF, Varayoud J, Lazzarino GP, Luque EH, Ramos JG. Pregnancy and lactation differentially modify the transcriptional regulation of steroidogenic enzymes through DNA methylation mechanisms in the hippocampus of aged rats. Mol Cell Endocrinol 2016; 429:73-83. [PMID: 27040308 DOI: 10.1016/j.mce.2016.03.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 01/10/2023]
Abstract
In the present study, we examined the mRNA expression and DNA methylation state of steroidogenic enzymes in the hippocampus of young adult (90-days-old) and middle-aged (450-days-old) nulliparous rats, and middle-aged multiparous rats subjected to three pregnancies with and without lactation. Aging decreased the mRNA levels of steroidogenic-related genes, while pregnancy and lactation significantly reduced the effect of aging, maintaining high expression levels of cytochrome P450 side-chain cleavage (P450scc), steroid 5α-reductase-1 (5αR-1), cytochrome P450arom (P450arom) and aldosterone synthase (P450(11β)-2). In addition, pregnancy and lactation diminished the methylation state of the 5αR-1 promoter and increased the transcription of brain-derived neurotrophic factor, synaptophysin and spinophilin. Pregnancy without lactation increased P450scc and 5αR-1 gene expression and decreased the methylation of their promoters. We concluded that the age-related decrease in the mRNA expression of steroidogenic enzymes is differentially attenuated by pregnancy and lactation in the rat hippocampus and that differential methylation mechanisms could be involved.
Collapse
Affiliation(s)
- María F Rossetti
- Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Gisela P Lazzarino
- Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Jorge G Ramos
- Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| |
Collapse
|
142
|
Rossetti MF, Cambiasso MJ, Holschbach MA, Cabrera R. Oestrogens and Progestagens: Synthesis and Action in the Brain. J Neuroendocrinol 2016; 28. [PMID: 27306650 DOI: 10.1111/jne.12402] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/14/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022]
Abstract
When steroids, such as pregnenolone, progesterone and oestrogen, are synthesised de novo in neural tissues, they are more specifically referred to as neurosteroids. These neurosteroids bind specific receptors to promote essential brain functions. Pregnenolone supports cognition and protects mouse hippocampal cells against glutamate and amyloid peptide-induced cell death. Progesterone promotes myelination, spinogenesis, synaptogenesis, neuronal survival and dendritic growth. Allopregnanolone increases hippocampal neurogenesis, neuronal survival and cognitive functions. Oestrogens, such as oestradiol, regulate synaptic plasticity, reproductive behaviour, aggressive behaviour and learning. In addition, neurosteroids are neuroprotective in animal models of Alzheimer's disease, Parkinson's disease, brain injury and ageing. Using in situ hybridisation and/or immunohistochemistry, steroidogenic enzymes, including cytochrome P450 side-chain cleavage, 3β-hydroxysteroid dehydrogenase/Δ5-Δ4 isomerase, cytochrome P450arom, steroid 5α-reductase and 3α-hydroxysteroid dehydrogenase, have been detected in numerous brain regions, including the hippocampus, hypothalamus and cerebral cortex. In the present review, we summarise some of the studies related to the synthesis and function of oestrogens and progestagens in the central nervous system.
Collapse
Affiliation(s)
- M F Rossetti
- Departamento de Bioquímica Clínica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Instituto de Salud y Ambiente del Litoral, CONICET-Universidad Nacional del Litoral, Santa Fe, Argentina
| | - M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M A Holschbach
- Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - R Cabrera
- Instituto de Investigaciones Biomédicas, INBIOMED-IMBECU-CONICET, Universidad de Mendoza, Mendoza, Argentina
| |
Collapse
|
143
|
Zhang M, Liu J, Zhou MM, Wu H, Hou Y, Li YF, Yin Y, Zheng L, Liu FY, Yi M, Wan Y. Elevated Neurosteroids in the Lateral Thalamus Relieve Neuropathic Pain in Rats with Spared Nerve Injury. Neurosci Bull 2016; 32:311-22. [PMID: 27325509 DOI: 10.1007/s12264-016-0044-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/11/2016] [Indexed: 01/01/2023] Open
Abstract
Neurosteroids are synthesized in the nervous system from cholesterol or steroidal precursors imported from peripheral sources. These compounds are important allosteric modulators of γ-aminobutyric acid A receptors (GABAARs), which play a vital role in pain modulation in the lateral thalamus, a main gate where somatosensory information enters the cerebral cortex. Using high-performance liquid chromatography/tandem mass spectrometry, we found increased levels of neurosteroids (pregnenolone, progesterone, deoxycorticosterone, allopregnanolone, and tetrahydrodeoxycorticosterone) in the chronic stage of neuropathic pain (28 days after spared nerve injury) in rats. The expression of the translocator protein TSPO, the upstream steroidogenesis rate-limiting enzyme, increased at the same time. In vivo stereotaxic microinjection of neurosteroids or the TSPO activator AC-5216 into the lateral thalamus (AP -3.0 mm, ML ±3.0 mm, DV 6.0 mm) alleviated the mechanical allodynia in neuropathic pain, while the TSPO inhibitor PK 11195 exacerbated it. The analgesic effects of AC-5216 and neurosteroids were significantly attenuated by the GABAAR antagonist bicuculline. These results suggested that elevated neurosteroids in the lateral thalamus play a protective role in the chronic stage of neuropathic pain.
Collapse
Affiliation(s)
- Meng Zhang
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
| | - Jia Liu
- Institute of Systems Biomedicine, Peking University, Beijing, 100191, China
| | - Meng-Meng Zhou
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
| | - Honghai Wu
- Department of Pharmacy, Bethune International Peace Hospital Shijiazhuang, Shijiazhuang, 050082, China
| | - Yanning Hou
- Department of Pharmacy, Bethune International Peace Hospital Shijiazhuang, Shijiazhuang, 050082, China
| | - Yun-Feng Li
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing, 100007, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Peking University, Beijing, 100191, China
| | - Lemin Zheng
- Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, and Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Peking University, Beijing, 100191, China
| | - Feng-Yu Liu
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
| | - Ming Yi
- Neuroscience Research Institute, Peking University, Beijing, 100191, China.
| | - You Wan
- Neuroscience Research Institute, Peking University, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
144
|
Quinn TA, Ratnayake U, Dickinson H, Castillo-Melendez M, Walker DW. The feto-placental unit, and potential roles of dehydroepiandrosterone (DHEA) in prenatal and postnatal brain development: A re-examination using the spiny mouse. J Steroid Biochem Mol Biol 2016; 160:204-13. [PMID: 26485665 DOI: 10.1016/j.jsbmb.2015.09.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 12/12/2022]
Abstract
Synthesis of dehydroepiandrosterone (DHEA) by the fetal adrenal gland is important for placental oestrogen production, and may also be important for modulating the effects of glucocorticoids on the developing brain. We have preciously shown that the enzymes and accessory proteins needed for DHEA synthesis-cytochrome P450 enzyme 17α-hydroxylase/17,20 lyase (P450c17), cytochrome-b5 (Cytb5), 3β-hydroxysteroid dehydrogenase (3βHSD)-are expressed in the adrenal gland from 30 days gestation, and DHEA, cortisol and aldosterone are present in fetal plasma from this time. Explant culture of fetal adrenal tissue showed that the spiny mouse adrenal gland, can synthesize and secrete DHEA from at least 0.75 of gestation, and suggest that DHEA may have an important role(s) in placental biosynthesis of oestrogens and in modulating the actions of glucocorticoids in the developing brain in this species. Post-natally, increased immuno-expression of P450c17 and Cytb5 expression in the zona reticularis of the adrenal gland and a significant increase in the synthesis and secretion of DHEA in plasma from 8 to 20 days of age in the spiny mouse, are representative of a period of high adrenal androgen production consistent with the human phenomenon of adrenarche. The studies summarised in this review also show that DHEA is produced de novo in the developing brain of the spiny mouse. These results showed that the spiny mouse brain can indeed produce DHEA from pregnenolone in a time-dependant manner, and coupled with the identification of P450c17 and Cytb5 protein in several regions of the brain, support the idea that DHEA is an endogenous neuro-active steroid in this species. Together, the studies outlined in this review indicate that the androgen DHEA is an important hormone of adrenal and Central Nervous System (CNS) origin in the fetal and postnatal spiny mouse. Disturbance of the development of these fetal tissues, and/or of the relationship between the fetal adrenal gland and placenta during pregnancy, may have significant consequences for fetal development, placental function, and maturation of the brain. It is proposed that such disturbances of normal adrenal function could account for some of the neuropathologies that arise in juvenile and adult offspring following illness and stress experienced by the mother during pregnancy.
Collapse
Affiliation(s)
- Tracey A Quinn
- The Ritchie Centre, Hudson Institute of Medical Research, Australia; Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Clayton 3168, Australia
| | - Udani Ratnayake
- The Ritchie Centre, Hudson Institute of Medical Research, Australia; The Florey Institute of Neuroscience and Mental Health, Melbourne University, Australia
| | - Hayley Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research, Australia; Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Clayton 3168, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Australia; Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Clayton 3168, Australia; The Florey Institute of Neuroscience and Mental Health, Melbourne University, Australia
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research, Australia; Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Clayton 3168, Australia.
| |
Collapse
|
145
|
Hirst JJ, Cumberland AL, Shaw JC, Bennett GA, Kelleher MA, Walker DW, Palliser HK. Loss of neurosteroid-mediated protection following stress during fetal life. J Steroid Biochem Mol Biol 2016; 160:181-8. [PMID: 26365557 DOI: 10.1016/j.jsbmb.2015.09.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/01/2015] [Accepted: 09/08/2015] [Indexed: 11/22/2022]
Abstract
Elevated levels of neurosteroids during late gestation protect the fetal brain from hypoxia/ischaemia and promote neurodevelopment. Suppression of allopregnanolone production during pregnancy leads to the onset of seizure-like activity and potentiates hypoxia-induced brain injury. Markers of myelination are reduced and astrocyte activation is increased. The placenta has a key role in maintaining allopregnanolone concentrations in the fetal circulation and brain during gestation and levels decline markedly after both normal and preterm birth. This leads to the preterm neonate developing in a neurosteroid deficient environment between delivery and term equivalence. The expression of 5α-reductases is also lower in the fetus prior to term. These deficiencies in neurosteroid exposure may contribute to the increase in incidence of the adverse patterns of behaviour seen in children that are born preterm. Repeated exposure to glucocorticoid stimulation suppresses 5α-reductase expression and allopregnanolone levels in the fetus and results in reduced myelination. Both fetal growth restriction and prenatal maternal stress lead to increased cortisol concentrations in the maternal and fetal circulation. Prenatal stress results in reduced expression of key GABAA receptor subunits that normally heighten neurosteroid sensitivity. These stressors also result in altered placental allopregnanolone metabolism pathways. These findings suggest that reduced neurosteroid production and action in the perinatal period may contribute to some of the adverse neurodevelopmental and behavioural outcomes that result from these pregnancy compromises. Studies examining perinatal steroid supplementation therapy with non-metabolisable neurosteroid analogues to improve these outcomes are warranted.
Collapse
Affiliation(s)
- Jonathan J Hirst
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Angela L Cumberland
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Julia C Shaw
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Greer A Bennett
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | - David W Walker
- Ritchie Centre for Baby Health Research, Department of Obstetrics and Gynaecology, Monash University, VIC 3800, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
146
|
Vallée M. Neurosteroids and potential therapeutics: Focus on pregnenolone. J Steroid Biochem Mol Biol 2016; 160:78-87. [PMID: 26433186 DOI: 10.1016/j.jsbmb.2015.09.030] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/18/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022]
Abstract
Considerable evidence from preclinical and clinical studies shows that steroids and in particular neurosteroids are important endogenous modulators of several brain-related functions. In this context, it remains to be elucidated whether neurosteroids may serve as biomarkers in the diagnosis of disorders and might have therapeutic potential for the treatment of these disorders. Pregnenolone (PREG) is the main steroid synthesized from cholesterol in mammals and invertebrates. PREG has three main sources of synthesis, the gonads, adrenal glands and brain and is submitted to various metabolizing pathways which are modulated depending on various factors including species, steroidogenic tissues and steroidogenic enzymes. Looking at the whole picture of steroids, PREG is often known as the precursor to other steroids and not as an active steroid per se. Actually, physiological and brain functions have been studied mainly for steroids that are very active either binding to specific intracellular receptors, or modulating with high affinity the abundant membrane receptors, GABAA or NMDA receptors. However, when high sensitive and specific methodological approaches were available to analyze low concentrations of steroids and then match endogenous levels of different steroid metabolomes, several studies have reported more significant alterations in PREG than in other steroids in extraphysiological or pathological conditions, suggesting that PREG could play a functional role as well. Additionally, several molecular targets of PREG were revealed in the mammalian brain and beneficial effects of PREG have been demonstrated in preclinical and clinical studies. On this basis, this review will be divided into three parts. The first provides a brief overview of the molecular targets of PREG and the pharmacological effects observed in animal and human studies. The second will focus on the possible functional role of PREG with an outline of the modulation of PREG levels in animal and in human research. Finally, the review will highlight the possible therapeutic uses of PREG that point towards the development of pregnenolone-like molecules.
Collapse
Affiliation(s)
- Monique Vallée
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux F33077, France; Université de Bordeaux, Bordeaux F33077, France.
| |
Collapse
|
147
|
Odermatt A, Strajhar P, Engeli RT. Disruption of steroidogenesis: Cell models for mechanistic investigations and as screening tools. J Steroid Biochem Mol Biol 2016; 158:9-21. [PMID: 26807866 DOI: 10.1016/j.jsbmb.2016.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/31/2015] [Accepted: 01/20/2016] [Indexed: 02/03/2023]
Abstract
In the modern world, humans are exposed during their whole life to a large number of synthetic chemicals. Some of these chemicals have the potential to disrupt endocrine functions and contribute to the development and/or progression of major diseases. Every year approximately 1000 novel chemicals, used in industrial production, agriculture, consumer products or as pharmaceuticals, are reaching the market, often with limited safety assessment regarding potential endocrine activities. Steroids are essential endocrine hormones, and the importance of the steroidogenesis pathway as a target for endocrine disrupting chemicals (EDCs) has been recognized by leading scientists and authorities. Cell lines have a prominent role in the initial stages of toxicity assessment, i.e. for mechanistic investigations and for the medium to high throughput analysis of chemicals for potential steroidogenesis disrupting activities. Nevertheless, the users have to be aware of the limitations of the existing cell models in order to apply them properly, and there is a great demand for improved cell-based testing systems and protocols. This review intends to provide an overview of the available cell lines for studying effects of chemicals on gonadal and adrenal steroidogenesis, their use and limitations, as well as the need for future improvements of cell-based testing systems and protocols.
Collapse
Affiliation(s)
- Alex Odermatt
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Petra Strajhar
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Roger T Engeli
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
148
|
Ogura Y, Haraguchi S, Nagino K, Ishikawa K, Fukahori Y, Tsutsui K. 7α-Hydroxypregnenolone regulates diurnal changes in sexual behavior of male quail. Gen Comp Endocrinol 2016; 227:130-5. [PMID: 26608258 DOI: 10.1016/j.ygcen.2015.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 11/14/2015] [Accepted: 11/17/2015] [Indexed: 11/16/2022]
Abstract
In the Japanese quail, 7α-hydroxypregnenolone, a previously undescribed avian neurosteroid, is actively produced in the brain. 7α-Hydroxypregnenolone acts as a novel neuronal activator to stimulate locomotor activity of quail. Therefore, in this study, we determined whether 7α-hydroxypregnenolone changes the expression of sexual behavior in Japanese quail. We first measured diurnal changes in sexual behavior of male quail exposed to a long-day photoperiod. We found that sexual behavior of male quail was high in the morning when endogenous 7α-hydroxypregnenolone level is high. Subsequently, we centrally administered 7α-hydroxypregnenolone in the evening when endogenous 7α-hydroxypregnenolone level is low. In the 30 min after intracerebroventricular (ICV) injection, 7α-hydroxypregnenolone dose dependently increased the frequency of sexual behavior of male quail. However, 7β-hydroxypregnenolone, a stereoisomer of 7α-hydroxypregnenolone, did not effect on the frequency of sexual behavior of male quail. In addition, to confirm the action of 7α-hydroxypregnenolone on sexual behavior, male birds received an ICV injection of ketoconazole, an inhibitor of cytochrome P450s, and behavioral experiments were performed in the morning. Ketoconazole significantly decreased the frequency of sexual behavior of male quail, whereas administration of 7α-hydroxypregnenolone to ketoconazole-treated males increased the frequency of their sexual behavior. These results indicate that 7α-hydroxypregnenolone regulates diurnal changes in sexual behavior of male quail.
Collapse
Affiliation(s)
- Yuki Ogura
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan
| | - Shogo Haraguchi
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan.
| | - Koki Nagino
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan
| | - Kei Ishikawa
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan
| | - Yoko Fukahori
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan.
| |
Collapse
|
149
|
Tsutsui K. How to contribute to the progress of neuroendocrinology: New insights from discovering novel neuropeptides and neurosteroids regulating pituitary and brain functions. Gen Comp Endocrinol 2016; 227:3-15. [PMID: 26145291 DOI: 10.1016/j.ygcen.2015.05.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/28/2015] [Accepted: 05/07/2015] [Indexed: 12/29/2022]
Abstract
Obtaining new insights by discovering novel neuropeptides and neurosteroids regulating pituitary and brain functions is essential for the progress of neuroendocrinology. At the beginning of 1970s, gonadotropin-releasing hormone (GnRH) was discovered in mammals. Since then, it was generally accepted that GnRH is the only hypothalamic neuropeptide regulating gonadotropin release in vertebrates. In 2000, however, gonadotropin-inhibitory hormone (GnIH), a novel hypothalamic neuropeptide that actively inhibits gonadotropin release, was discovered in quail. The follow-up studies demonstrated that GnIH acts as a new key player for regulation of reproduction across vertebrates. It now appears that GnIH acts on the pituitary and the brain to serve a number of behavioral and physiological functions. On the other hand, a new concept has been established that the brain synthesizes steroids, called neurosteroids. The formation of neurosteroids in the brain was originally demonstrated in mammals and subsequently in other vertebrates. Recently, 7α-hydroxypregnenolone was discovered as a novel bioactive neurosteroid inducing locomotor behavior of vertebrates, indicating that neurosteroidogenesis in the brain is still incompletely elucidated in vertebrates. At the beginning of 2010s, it was further found that the pineal gland actively produces neurosteroids. Pineal neurosteroids act on the brain to regulate locomotor rhythms and neuronal survival. Furthermore, the interaction of neuropeptides and neurosteroids is becoming clear. GnIH decreases aggressive behavior by regulating neuroestrogen synthesis in the brain. This review summarizes these new insights by discovering novel neuropeptides and neurosteroids in the field of neuroendocrinology.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo 162-8480, Japan.
| |
Collapse
|
150
|
do Rego JL, Vaudry H. Comparative aspects of neurosteroidogenesis: From fish to mammals. Gen Comp Endocrinol 2016; 227:120-9. [PMID: 26079790 DOI: 10.1016/j.ygcen.2015.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
It is now clearly established that the central and peripheral nervous systems have the ability to synthesize de novo steroids referred to as neurosteroids. The major evidence for biosynthesis of neuroactive steroids by nervous tissues is based on the expression of enzymes implicated in the formation of steroids in neural cells. The aim of the present review is to summarize the current knowledge regarding the presence of steroidogenic enzymes in the brain of vertebrates and to highlight the very considerable contribution of Professor Kazuyoshi Tsutsui in this domain. The data indicate that expression of steroid-producing enzymes in the brain appeared early during vertebrate evolution and has been preserved from fish to mammals.
Collapse
Affiliation(s)
- Jean Luc do Rego
- Institute for Research and Innovation in Biomedicine (IRIB), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Rouen, 76821 Mont-Saint-Aignan, France
| | - Hubert Vaudry
- Institute for Research and Innovation in Biomedicine (IRIB), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Rouen, 76821 Mont-Saint-Aignan, France; Neurotrophic Factors and Neuronal Differentiation Team, Inserm U982, University of Rouen, 76821 Mont-Saint-Aignan, France.
| |
Collapse
|