51
|
Abstract
Anesthesiologists are frequently confronted with patients who are at risk for neurological complications due to perioperative stroke or prior traumatic brain injury. In this review, we address the growing and fascinating body of data that suggests gender and sex steroids influence the pathophysiology of injury and outcome for these patients. Cerebral ischemia, traumatic brain injury, and epilepsy are reviewed in the context of potential sex differences in mechanisms and outcomes of brain injury and the role of estrogen, progesterone, and androgens in shaping these processes. Lastly, implications for current and future perioperative and intensive care are identified.
Collapse
Affiliation(s)
- Kamila Vagnerova
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | | | |
Collapse
|
52
|
Lee AW, Pfaff DW. Hormone effects on specific and global brain functions. J Physiol Sci 2008; 58:213-20. [PMID: 18505601 DOI: 10.2170/physiolsci.rv007008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Accepted: 05/24/2008] [Indexed: 11/05/2022]
Abstract
The first demonstration of how biochemical changes in neurons in specific parts of the brain direct a complete mammalian behavior derived from the effects of estrogens in hypothalamic neurons that facilitate lordosis behavior, the primary reproductive behavior of female quadrupeds (Pfaff. Estrogens and Brain Function. 1980; Pfaff. Drive: Neurobiological and Molecular Mechanisms of Sexual Motivation. 1999). Sex behaviors depend on sexual arousal that in turn depends on a primitive function: generalized CNS arousal (Pfaff. Brain Arousal and Information Theory. 2006). Here we summarize one of the ways in which a generalized arousal transmitter, norepinephrine, can influence the electrical excitability of ventromedial hypothalamic cells in a way that will foster female sex behavior.
Collapse
Affiliation(s)
- A W Lee
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
53
|
Fatehi M, Fatehi-Hassanabad Z. Effects of 17beta-estradiol on neuronal cell excitability and neurotransmission in the suprachiasmatic nucleus of rat. Neuropsychopharmacology 2008; 33:1354-64. [PMID: 17687263 DOI: 10.1038/sj.npp.1301523] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
17beta-Estradiol receptors have been found in several brain nuclei including the suprachiasmatic nucleus (SCN) of mammalian species. The SCN is believed to act as brain clock regulating circadian and circannual biological rhythms, such as body temperature, sleep, and mood. Here, we examined whether 17beta-estradiol (E2) could affect cell excitability and synaptic transmission in the SCN. Bath application of E2 (0.03-3 microM) increased the spontaneous firing frequency and depolarized cell membrane of the SCN neurons significantly. Furthermore, E2 (0.03-3 microM) increased (by about 25-150% of control) frequency of the miniature excitatory postsynaptic currents. Amplitude of the evoked excitatory postsynaptic currents was enhanced (by about 32% of control) after exposure to 1 microM E2. The paired-pulse ratio was reduced by E2. These effects were prevented by the estrogen receptor antagonist, ICI 182780. Exposure to the biologically inactive 17alpha-estradiol did not cause any significant changes in the parameters mentioned above. These findings are in favor of an implication of estrogen in modulation of neuronal activity in SCN and possibly regulating circadian rhythms.
Collapse
Affiliation(s)
- Mohammad Fatehi
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, Canada.
| | | |
Collapse
|
54
|
Vasudevan N, Pfaff DW. Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front Neuroendocrinol 2008; 29:238-57. [PMID: 18083219 DOI: 10.1016/j.yfrne.2007.08.003] [Citation(s) in RCA: 262] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 08/14/2007] [Indexed: 12/16/2022]
Abstract
Ligands for the nuclear receptor superfamily have at least two mechanisms of action: (a) classical transcriptional regulation of target genes (genomic mechanisms); and (b) non-genomic actions, which are initiated at the cell membrane, which could also impact transcription. Though transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. This has led to considerable debate over the physiological relevance of membrane-initiated actions of hormones versus genomic actions of hormones, with genomic actions predominating in the endocrine field. There is good evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium and that these are linked to physiologically relevant scenarios in the brain. We show evidence in this review, that membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription in both the central nervous system and in non-neuronal cell lines. We present a theoretical scenario which can be used to understand this phenomenon. These signaling cascades may occur in parallel or in series but subsequently, converge at the modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other non-cognate hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription, though the relevance of this is less clear. The idea that coupling between membrane-initiated and genomic actions of hormones is a novel idea in neuroendocrinology and provides us with a unified view of hormone action in the central nervous system.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Cell and Molecular Biology Department, Tulane University, LA 70118, USA.
| | | |
Collapse
|
55
|
Jefremov V, Rakitin A, Mahlapuu R, Zilmer K, Bogdanovic N, Zilmer M, Karelson E. 17beta-Oestradiol stimulation of G-proteins in aged and Alzheimer's human brain: comparison with phytoestrogens. J Neuroendocrinol 2008; 20:587-96. [PMID: 18363809 DOI: 10.1111/j.1365-2826.2008.01696.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The neuroprotective action of oestrogens and oestrogen-like compounds is in the focus of basic and clinical research. Although such action has been shown to be associated with neuronal plasma membranes, the implication of G-proteins remains to be elucidated. This study revealed that micromolar concentrations (microM) of 17beta-oestradiol and phytoestrogens, genistein and daidzein, significantly (P < 0.05) stimulate G-proteins ([(35)S]GTP gamma S binding) in the post-mortem hippocampal membranes of age-matched control women with the respective maximum effects of 28, 20 and 15% at 10 microM. In the frontocortical membranes, the stimulation of G-proteins did not differ significantly from that in hippocampal membranes. Although in the hippocampus and frontal cortex of the Alzheimer's disease (AD) women's brain, 10 microM 17beta-oestradiol produced significantly (P < 0.05) lower stimulation of G-proteins than in the control regions, stimulation by phytoestrogens revealed no remarkable decline. 17beta-Oestradiol, genistein and daidzein revealed a selective effect on various G-proteins (G(alphas), G(alpha o), G(alpha i1) or G(alpha 11) plus G(beta 1 gamma 2)) expressed in Sf9 cells. At a concentration of 10 microM, 17beta-oestradiol suppressed the H(2)O(2) and homocysteine stimulated G-proteins in the frontocortical membranes of control women to a greater extent than phytoestrogens. In AD, the suppressing effect of each compound was lower than in the controls. In the cell-free systems, micromolar concentrations of phytoestrogens scavenged OH(*) and the 2.2-diphenyl-1-picrylhydrazyl free radical (DPPH(*)) more than 17beta-oestradiol did. In the frontocortical membranes of control women, the 20 microM 17beta-oestradiol stimulated adenylate cyclase with 20% maximal effect, whereas, in AD, the effect was insignificant. Genistein did not stimulate enzyme either in control or AD frontocortical membranes. Our data confirm that the agents stimulate G-proteins in control and AD women's brains, although 17beta-oestradiol and phytoestrogens have similarities and differences in this respect. We suggest that, besides the ER-dependent one, the ER-independent antioxidant mechanism is responsible for the oestrogen stimulation of G-proteins in the brain membranes. Both of these mechanisms could be involved in the neuroprotective signalling of oestrogens that contributes to their preventive/therapeutic action against postmenopausal neurological disorders.
Collapse
Affiliation(s)
- V Jefremov
- Department of Biochemistry, Tartu University, Tartu, Estonia
| | | | | | | | | | | | | |
Collapse
|
56
|
Loyd DR, Morgan MM, Murphy AZ. Sexually dimorphic activation of the periaqueductal gray-rostral ventromedial medullary circuit during the development of tolerance to morphine in the rat. Eur J Neurosci 2008; 27:1517-24. [PMID: 18364026 PMCID: PMC2821209 DOI: 10.1111/j.1460-9568.2008.06100.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The midbrain periaqueductal gray (PAG) and its descending projections to the rostral ventromedial medulla (RVM) provides an essential neural circuit for the antinociceptive effects of opiates, and has been implicated in the development of tolerance to morphine. Systemic morphine activates a greater proportion of PAG-RVM neurons in male vs female rats, and induces tolerance to a greater degree in males. The present studies tested the hypothesis that if the PAG-RVM pathway is essential for the development of tolerance, then: (i) morphine activation of the PAG-RVM pathway should decline as tolerance develops; and (ii) sex differences in the development of tolerance to morphine should be reflected as a greater decline in the activation of this pathway in males. These hypotheses were tested in male and female rats using behavioral testing (hot-plate) and immunohistochemistry to map the activation of the PAG-RVM pathway following repeated morphine administration (5 mg/kg; s.c.). In males, morphine potency decreased from 3.0 to 6.3 mg/kg, indicating tolerance, and this was paralleled by a steady decline in the percentage of PAG-RVM output neurons activated by morphine. In contrast, in females the shift in morphine potency was significantly attenuated (D(50) 6-8.3 mg/kg), and no significant difference in the activity of PAG-RVM output neurons was noted. These results demonstrate that the greater development of tolerance to morphine administration in male rats corresponds with a significant reduction in the activation of the PAG-RVM circuit and suggest a central role for the PAG in the development of tolerance to morphine.
Collapse
Affiliation(s)
- Dayna R Loyd
- Department of Biology, Center for Behavioral Neuroscience, Georgia State University, PO Box 4010, Atlanta, GA 30302-4010, USA
| | | | | |
Collapse
|
57
|
Raz L, Khan MM, Mahesh VB, Vadlamudi RK, Brann DW. Rapid Estrogen Signaling in the Brain. Neurosignals 2008; 16:140-53. [DOI: 10.1159/000111559] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
58
|
Lee AW, Kyrozis A, Chevaleyre V, Kow LM, Zhou J, Devidze N, Zhang Q, Etgen AM, Pfaff DW. Voltage-dependent calcium channels in ventromedial hypothalamic neurones of postnatal rats: modulation by oestradiol and phenylephrine. J Neuroendocrinol 2008; 20:188-98. [PMID: 18088362 DOI: 10.1111/j.1365-2826.2007.01637.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Oestradiol actions in the hypothalamus play an important role in reproductive behaviour. Oestradiol treatment in vivo induces alpha(1b)-adrenoceptor mRNA and increases the density of alpha(1B)-adrenoceptor binding in the hypothalamus. Oestradiol is also known to modulate neuronal excitability, in some cases by modulating calcium channels. We assessed the effects of phenylephrine, an alpha(1)-adrenergic agonist, on low-voltage-activated (LVA) and high-voltage-activated (HVA) calcium channels in ventromedial hypothalamic (VMN) neurones from vehicle- and oestradiol-treated female rats. Whole-cell and gramicidin perforated-patch recordings were obtained, with barium as the charge carrier. In the absence of phenylephrine, oestradiol treatment increased the magnitude of LVA currents compared to controls, but had no effect on HVA currents. Phenylephrine enhanced HVA currents in a significantly greater proportion of neurones from oestradiol-treated rats (76%) than from vehicle-treated (41%) rats. The L-channel blocker nifedipine abolished this oestradiol effect on phenylephrine-enhanced HVA currents. Preincubating slices with the N-type channel blocker omega-conotoxin GVIA completely blocked the phenylephrine response, suggesting that the N-type channel is essential. Phenylephrine also stimulated LVA currents in approximately two-thirds of neurones in slices from both vehicle- and oestradiol-treated rats. Our data show that oestradiol increases LVA currents in the VMN. Oestradiol also amplifies alpha(1)-adrenergic signalling by increasing the proportion of neurones showing phenylephrine-stimulated HVA currents mediated by N- and L-type calcium channels. In this way, oestradiol may increase excitatory responses to arousing adrenergic inputs to VMN neurones governing oestradiol-dependent reproductive behaviour.
Collapse
Affiliation(s)
- A W Lee
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Chang E, O'Donnell ME, Barakat AI. Shear stress and 17β-estradiol modulate cerebral microvascular endothelial Na-K-Cl cotransporter and Na/H exchanger protein levels. Am J Physiol Cell Physiol 2008; 294:C363-71. [DOI: 10.1152/ajpcell.00045.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ion transporters of blood-brain barrier (BBB) endothelial cells play an important role in regulating the movement of ions between the blood and brain. During ischemic stroke, reduction in cerebral blood flow is accompanied by transport of Na and Cl from the blood into the brain, with consequent brain edema formation. We have shown previously that a BBB Na-K-Cl cotransporter (NKCC) participates in ischemia-induced brain Na and water uptake and that a BBB Na/H exchanger (NHE) may also participate. While the abrupt reduction of blood flow is a prominent component of ischemia, the effects of flow on BBB NKCC and NHE are not known. In the present study, we examined the effects of changes in shear stress on NKCC and NHE protein levels in cerebral microvascular endothelial cells (CMECs). We have shown previously that estradiol attenuates both ischemia-induced cerebral edema and CMEC NKCC activity. Thus, in the present study, we also examined the effects of estradiol on NKCC and NHE protein levels in CMECs. Exposing CMECs to steady shear stress (19 dyn/cm2) increased the abundance of both NKCC and NHE. Estradiol abolished the shear stress-induced increase in NHE but not NKCC. Abrupt reduction of shear stress did not alter NKCC or NHE abundance in the absence of estradiol, but it decreased NKCC abundance in estradiol-treated cells. Our results indicate that changes in shear stress modulate BBB NKCC and NHE protein levels. They also support the hypothesis that estradiol attenuates edema formation in ischemic stroke in part by reducing the abundance of BBB NKCC protein.
Collapse
|
60
|
McCullough LD, Koerner IP, Hurn PD. Effects of gender and sex steroids on ischemic injury. HANDBOOK OF CLINICAL NEUROLOGY 2008; 92:149-69. [PMID: 18790274 DOI: 10.1016/s0072-9752(08)01908-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
61
|
Iqbal J, Latchoumanin O, Clarke IJ. Rapid in vivo effects of estradiol-17beta in ovine pituitary gonadotropes are displayed by phosphorylation of extracellularly regulated kinase, serine/threonine kinase, and 3',5'-cyclic adenosine 5'-monophosphate-responsive element-binding protein. Endocrinology 2007; 148:5794-802. [PMID: 17823264 DOI: 10.1210/en.2007-0986] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have determined the time course of phosphorylation of MAPK/ERK, cAMP-responsive element-binding protein (CREB), and serine/threonine kinase (Akt) in ovine pituitary gonadotropes after in vivo injection (iv) of either 25 mug estradiol-17beta (E17beta) or vehicle. In ovariectomized ewes, E17beta increased the number of gonadotropes expressing phosphorylated (p)ERK-1/2 and pCREB immunoreactivity (-IR) within 90 min, as assessed by immunohistochemistry. By Western blot, we also showed that pERK-1/2, pCREB, and pAkt (ser 473) proteins were up-regulated by E17beta. In ovariectomized, hypothalamo-pituitary-disconnected animals, gonadotrope function was restored with hourly GnRH pulses (iv), and E17beta injection (iv) reduced LH response within 1 h. Immunohistochemistry showed that E17beta increased pERK-1/2-IR in gonadotropes within 15 min and peak response at 60 min. The number of cells immunoreactive for pCREB was greater in E17beta-treated animals than in vehicle-injected controls at 60 and 90 min. Western blot revealed a pERK-1/2 response within 15 min and pCREB response at 30 min. Up-regulation of pAkt occurred within 60 min of E17beta treatment. Thus, rapid effects of E17beta on gonadotropes involve phosphorylation of second messenger proteins with a time course that may relate to the rapid negative feedback effect to reduce responsiveness to GnRH.
Collapse
Affiliation(s)
- Javed Iqbal
- Department of Physiology, Monash University, Clayton, Victoria 3880, Australia
| | | | | |
Collapse
|
62
|
Palmer ML, Schiller KR, O'Grady SM. Apical SK potassium channels and Ca2+-dependent anion secretion in endometrial epithelial cells. J Physiol 2007; 586:717-26. [PMID: 18048454 DOI: 10.1113/jphysiol.2007.142877] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Apical uridine triphosphate (UTP) stimulation was shown to increase short circuit current (I(sc)) in immortalized porcine endometrial gland epithelial monolayers. Pretreatment with the bee venom toxin apamin enhanced this response. Voltage-clamp experiments using amphotericin B-permeablized monolayers revealed that the apamin-sensitive current increased immediately after UTP stimulation and was K(+) dependent. The current-voltage relationship was slightly inwardly rectifying with a reversal potential of -52 +/- 2 mV, and the P(K)/P(Na) ratio was 14, indicating high selectivity for K(+). Concentration-response relationships for apamin and dequalinium had IC(50) values of 0.5 nm and 1.8 microm, respectively, consistent with data previously reported for SK3 channels in excitable cells and hepatocytes. Treatment of monolayers with 50 microm BAPTA-AM completely blocked the effects of UTP on K(+) channel activation, indicating that the apamin-sensitive current was also Ca(2+) dependent. Moreover, channel activation was blocked by calmidazolium (IC(50) = 5 microm), suggesting a role for calmodulin in Ca(2+)-dependent regulation of channel activity. RT-PCR experiments demonstrated expression of mRNA for the SK1 and SK3 channels, but not SK2 channels. Treatment of monolayers with 20 nm oestradiol-17beta produced a 2-fold increase in SK3 mRNA, a 2-fold decrease in SK1 mRNA, but no change in GAPDH mRNA expression. This result correlated with a 2.5-fold increase in apamin-sensitive K(+) channel activity in the apical membrane. We speculate that SK channels provide a mechanism for rapidly sensing changes in intracellular Ca(2+) near the apical membrane, evoking immediate hyperpolarization necessary for increasing the driving force for anion efflux following P2Y receptor activation.
Collapse
Affiliation(s)
- Melissa L Palmer
- Department of Physiology, 495 Animal Science/Veterinary Medicine Bldg, University of Minnesota, 1988 Fitch Ave., St Paul, MN 55108, USA
| | | | | |
Collapse
|
63
|
Greenspan JD, Craft RM, LeResche L, Arendt-Nielsen L, Berkley KJ, Fillingim RB, Gold MS, Holdcroft A, Lautenbacher S, Mayer EA, Mogil JS, Murphy AZ, Traub RJ. Studying sex and gender differences in pain and analgesia: a consensus report. Pain 2007; 132 Suppl 1:S26-S45. [PMID: 17964077 DOI: 10.1016/j.pain.2007.10.014] [Citation(s) in RCA: 713] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Accepted: 10/09/2007] [Indexed: 12/21/2022]
Abstract
In September 2006, members of the Sex, Gender and Pain Special Interest Group of the International Association for the Study of Pain met to discuss the following: (1) what is known about sex and gender differences in pain and analgesia; (2) what are the "best practice" guidelines for pain research with respect to sex and gender; and (3) what are the crucial questions to address in the near future? The resulting consensus presented herein includes input from basic science, clinical and psychosocial pain researchers, as well as from recognized experts in sexual differentiation and reproductive endocrinology. We intend this document to serve as a utilitarian and thought-provoking guide for future research on sex and gender differences in pain and analgesia, both for those currently working in this field as well as those still wondering, "Do I really need to study females?"
Collapse
Affiliation(s)
- Joel D Greenspan
- Department of Biomedical Sciences, University of Maryland Dental School, University of Maryland, Baltimore, MD 21201-1510, USA Research Center for Neuroendocrine Influences on Pain, Baltimore, MD 21201-1510, USA Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA Department of Oral Medicine, University of Washington, Seattle, WA 98195-6370, USA Laboratory for Experimental Pain Research, Department of Health Science and Technology, Center for Sensory-Motor Interaction, Aalborg University, Fredrik Bajers Vej 7, DK-9220 Aalborg, Denmark Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA Department of Community Dentistry and Behavioral Science, University of Florida College of Dentistry, Gainesville, FL 32610-3628, USA Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA Division of Surgery, Oncology, Reproductive Biology and Anaesthetics, Chelsea and Westminster Hospital, Imperial College, London SW10 9NH, UK Department of Physiological Psychology, University of Bamberg, Bamberg 96045, Germany Center for Neurovisceral Sciences and Women's Health, and Departments of Medicine, Psychiatry and Biobehavioral Sciences, and Physiology, UCLA School of Medicine, Los Angeles, CA 900095-1792, USA Department of Psychology, McGill University, Montreal, Canada PQ H3A 1B1 Department of Biology, Georgia State University, Atlanta, GA 30303-0389, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Quesada A, Romeo HE, Micevych P. Distribution and localization patterns of estrogen receptor-beta and insulin-like growth factor-1 receptors in neurons and glial cells of the female rat substantia nigra: localization of ERbeta and IGF-1R in substantia nigra. J Comp Neurol 2007; 503:198-208. [PMID: 17480015 PMCID: PMC2907103 DOI: 10.1002/cne.21358] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although several studies have focused on the neuroprotective effects of estrogen (E2) on stroke, there have been tantalizing reports on the potential neuroprotective role of E2 in degenerative neuronal diseases such as Alzheimer's and Parkinson's (PD). In animal models of PD, E2 protects the nigrostriatal dopaminergic (DA) system against neurotoxins. However, little is known about the cellular and molecular mechanism(s) involved by which E2 elicits its neuroprotective effects on the nigrostriatal DA system. A preferred mechanism for neuroprotection is the interaction of E2 with specific neuroprotective growth factors and receptors. One such neuroprotective factor/receptor system is insulin-like growth factor-1 (IGF-1). E2 neuroprotective effects in the substantia nigra (SN) DA system have been shown to be dependent on IGF-1. To determine whether E2 also interacts with the IGF-1 receptor (IGF-1R) and to determine the cellular localization of estrogen receptor (ER) and IGF-1R, we compared the distribution of ER and IGF-1R in the SN. Stereological measurements revealed that 40% of the subpopulation of tyrosine hydroxylase-immunoreactive (TH-ir) SN pars compacta (SNpc) DA neurons are immunoreactive for estrogen receptor-beta (ERbeta). No immunolabeling for ERalpha was observed. In situ hybridization and immunocytochemistry studies confirmed the expression of IGF-1R mRNA and revealed that almost all TH-ir SNpc DA neurons were immunoreactive for IGF-1R, respectively. Moreover, one-third of glial fibrillary acidic protein (GFAP-ir) cells in the SN were ERbeta-ir, and 67% of GFAP-ir cells expressed IGF-1R-ir. Therefore, the localization of ERbeta and IGF-1R on SNpc DA neurons and astrocytes suggests a modulatory role of E2 on IGF-1R, and this modulation may affect neuroprotection.
Collapse
Affiliation(s)
- Arnulfo Quesada
- Department of Neurobiology, Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095-1763, USA.
| | | | | |
Collapse
|
65
|
Fricke O, Kow LM, Bogun M, Pfaff DW. Estrogen evokes a rapid effect on intracellular calcium in neurons characterized by calcium oscillations in the arcuate nucleus. Endocrine 2007; 31:279-88. [PMID: 17906376 DOI: 10.1007/s12020-007-0034-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 06/11/2007] [Accepted: 06/13/2007] [Indexed: 10/23/2022]
Abstract
Rapid estrogen effects became an interesting topic to explain estrogen effects not associated with the classical nuclear pathway. The rapid estrogen effect on intracellular calcium oscillations was characterized in neurons of the arcuate nucleus. Ratiometric calcium imaging (fura-2AM) was used to measure intracellular calcium in brain slices of female Swiss Webster mice (median of age 27 days p.n.). Calcium oscillations were dependent on intracellular calcium and also on calcium influx from the extracellular space. The perfusion of slices with calcium-free solution inhibited spontaneous calcium oscillations. The metabotropic glutamate receptor agonist t-ACPD (5 microM) and low concentrated ryanodine (100 nM) induced intracellular calcium release when slices were perfused with calcium-free solution. 17beta-estradiol (10 nM) also induced intracellular calcium release in calcium-free ACSF. This effect was inhibited by the preceding administration of thapsigargin (2 microM) indicating the association of the rapid estrogen effect with intracellular calcium stores. The administration of the non-selective phospholipase C-inhibitor ET-18 (30 microM), but not U73122 (10 microM), and the inhibition of protein kinase A by H-89 (0.25 microM) suppressed the rapid estrogen effect. Analyses indicated a qualitative, but not quantitatively significant effect of 17beta-estradiol on calcium oscillations.
Collapse
Affiliation(s)
- Oliver Fricke
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
66
|
Loyd DR, Morgan MM, Murphy AZ. Morphine preferentially activates the periaqueductal gray-rostral ventromedial medullary pathway in the male rat: a potential mechanism for sex differences in antinociception. Neuroscience 2007; 147:456-68. [PMID: 17540508 PMCID: PMC1949345 DOI: 10.1016/j.neuroscience.2007.03.053] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Revised: 03/16/2007] [Accepted: 03/20/2007] [Indexed: 01/06/2023]
Abstract
The midbrain periaqueductal gray (PAG), and its descending projections to the rostral ventromedial medulla (RVM), provide an essential neural circuit for opioid-produced antinociception. Recent anatomical studies have reported that the projections from the PAG to the RVM are sexually dimorphic and that systemic administration of morphine significantly suppresses pain-induced activation of the PAG in male but not female rats. Given that morphine antinociception is produced in part by disinhibition of PAG output neurons, it is hypothesized that a differential activation of PAG output neurons mediates the sexually dimorphic actions of morphine. The present study examined systemic morphine-induced activation of PAG-RVM neurons in the absence of pain. The retrograde tracer Fluorogold (FG) was injected into the RVM to label PAG-RVM output neurons. Activation of PAG neurons was determined by quantifying the number of Fos-positive neurons 1 h following systemic morphine administration (4.5 mg/kg). Morphine produced comparable activation of the PAG in both male and female rats, with no significant differences in either the quantitative or qualitative distribution of Fos. While microinjection of FG into the RVM labeled significantly more PAG output neurons in female rats than male rats, very few of these neurons (20%) were activated by systemic morphine administration in comparison to males (50%). The absolute number of PAG-RVM neurons activated by morphine was also greater in males. These data demonstrate widespread disinhibition of PAG neurons following morphine administration. The greater morphine-induced activation of PAG output neurons in male compared with female rats is consistent with the greater morphine-induced antinociception observed in males.
Collapse
Affiliation(s)
- Dayna R. Loyd
- Department of Biology, Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302-4010
| | - Michael M. Morgan
- Department of Psychology, Washington State University, Vancouver, Washington 98686-9600
| | - Anne Z. Murphy
- Department of Biology, Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302-4010
| |
Collapse
|
67
|
Barhoumi R, Burghardt RC, Qian Y, Tiffany-Castiglioni E. Effects of propofol on intracellular Ca2+ homeostasis in human astrocytoma cells. Brain Res 2007; 1145:11-8. [PMID: 17328872 DOI: 10.1016/j.brainres.2007.01.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 01/22/2007] [Accepted: 01/26/2007] [Indexed: 11/28/2022]
Abstract
The effects of propofol, a short-acting general anesthetic, upon cell growth and Ca(2+) signaling in a human astrocytic cell line were examined. Exposure of cells to graded concentrations of propofol resulted in a dose-dependent decrease in cell number with an inhibitory concentration of cell viability (IC50) of 31.7+/-1.2 microM. To evaluate the changes in intracellular Ca(2+) homeostasis induced by propofol, cytoplasmic and mitochondrial Ca(2+) were measured by fluorescence imaging. Mitochondrial Ca(2+) increased while cytoplasmic Ca(2+) decreased significantly at a propofol concentration lower than the IC50 (10 microM for 24 h, 1 microM for 72 h). In addition, propofol diminished the Ca(2+) response induced by fetal bovine serum (FBS). To determine the source of Ca(2+) alterations induced by propofol, pharmacologic agents targeting intracellular Ca(2+) homeostasis mechanisms were used. Nifedipine, an L-type Ca(2+) channel blocker, decreased FBS-induced Ca(2+) response of control cells to a level similar to propofol treated cells. However, diazoxide (a K(+)-ATP channel opener) administered 1 h before FBS addition restored the FBS response in propofol treated cells to a level similar to control. In addition, diazoxide increased mitochondrial Ca(2+) in control cells to a level comparable to propofol treated cells suggesting activation of these channels by propofol treatment. Addition of 1 muM RU-360 (a selective blocker of the mitochondrial Ca(2+) uniporter) for 30 min prior to propofol treatment restored mitochondrial and cytoplasmic Ca(2+) to control levels. These data suggest that voltage operated Ca(2+) channels, mitochondrial Ca(2+) and K(+)-ATP channels may be targets of propofol action in astrocytes.
Collapse
Affiliation(s)
- Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA.
| | | | | | | |
Collapse
|
68
|
Glidewell-Kenney C, Hurley LA, Pfaff L, Weiss J, Levine JE, Jameson JL. Nonclassical estrogen receptor alpha signaling mediates negative feedback in the female mouse reproductive axis. Proc Natl Acad Sci U S A 2007; 104:8173-7. [PMID: 17470805 PMCID: PMC1876590 DOI: 10.1073/pnas.0611514104] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Ovarian estrogen exerts both positive and negative feedback control over luteinizing hormone (LH) secretion during the ovulatory cycle. Estrogen receptor (ER) alpha but not ERbeta knockout mice lack estrogen feedback. Thus, estrogen feedback appears to be primarily mediated by ERalpha. However, it is now recognized that, in addition to binding to estrogen response elements (EREs) in DNA to alter target gene transcription, ERalpha signals through ERE-independent or nonclassical pathways, and the relative contributions of these pathways in conveying estrogen feedback remain unknown. Previously we created a knockin mouse model expressing a mutant form of ERalpha (AA) with ablated ERE-dependent but intact ERE-independent activity. Breeding this allele onto the ERalpha-null (-/-) background, we examine the ability of ERE-independent ERalpha signaling pathways to convey estrogen feedback regulation of the female hypothalamic-pituitary axis in vivo. ERalpha-/AA exhibited 69.9% lower serum LH levels compared with ERalpha-/- mice. Additionally, like wild type, ERalpha-/AA mice exhibited elevated LH after ovariectomy (OVX). Furthermore, the post-OVX rise in serum LH was significantly suppressed by estrogen treatment in OVX ERalpha-/AA mice. However, unlike wild type, both ERalpha-/AA and ERalpha-/- mice failed to exhibit estrous cyclicity, spontaneous ovulation, or an afternoon LH surge response to estrogen. These results indicate that ERE-independent ERalpha signaling is sufficient to convey a major portion of estrogen's negative feedback actions, whereas positive feedback and spontaneous ovulatory cyclicity require ERE-dependent ERalpha signaling.
Collapse
Affiliation(s)
- C. Glidewell-Kenney
- *Division of Endocrinology, Metabolism, and Molecular Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008; and
| | - L. A. Hurley
- *Division of Endocrinology, Metabolism, and Molecular Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008; and
| | - L. Pfaff
- *Division of Endocrinology, Metabolism, and Molecular Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008; and
| | - J. Weiss
- *Division of Endocrinology, Metabolism, and Molecular Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008; and
| | - J. E. Levine
- Department of Neurobiology and Physiology, Northwestern University, Evanston, IL 60208
| | - J. L. Jameson
- *Division of Endocrinology, Metabolism, and Molecular Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008; and
- To whom correspondence should be addressed at:
251 East Huron Street, Galter Pavilion Suite 3-150, Chicago, IL 60611. E-mail:
| |
Collapse
|
69
|
Abstract
Hormonal ligands for the nuclear receptor superfamily have at least two interacting mechanisms of action: 1) classical transcriptional regulation of target genes (genomic mechanisms); and 2) nongenomic actions that are initiated at the cell membrane, which could impact transcription. Although transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. Historically, this has led to a considerable divergence of thought in the molecular endocrine field. We have attempted to uncover principles of hormone action that are relevant to membrane-initiated actions of estrogens. There is evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium. Membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription. These signaling cascades may occur in parallel or in series but subsequently converge at the level of modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription. The idea of synergistic coupling between membrane-initiated and genomic actions of hormones fundamentally revises the paradigms of cell signaling in neuroendocrinology.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | |
Collapse
|
70
|
Loucif AJC, Bonnavion P, Macri B, Golmard JL, Boni C, Melfort M, Leonard G, Lesch KP, Adrien J, Jacquin TD. Gender-dependent regulation of G-protein-gated inwardly rectifying potassium current in dorsal raphe neurons in knock-out mice devoid of the 5-hydroxytryptamine transporter. ACTA ACUST UNITED AC 2007; 66:1475-88. [PMID: 17013926 DOI: 10.1002/neu.20321] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Agonists at G-protein-coupled receptors in neurons of the dorsal raphe nucleus (DRN) of knock-out mice devoid of the serotonin transporter (5-HTT(-/-)) exhibit lower efficacy to inhibit cellular discharge than in wild-type counterparts. Using patch-clamp whole-cell recordings, we found that a G-protein-gated inwardly rectifying potassium (GIRK) current is involved in the inhibition of spike discharge induced by 5-HT1A agonists (5-carboxamidotryptamine (5-CT) and (+/-)-2-dipropylamino-8-hydroxy-1,2,3,4-tetrahydronaphthalene hydrobromide (8-OH-DPAT); 50 nM-30 microM) in both wild-type and 5-HTT(-/-) female and male mice. These effects were mimicked by 5'-guanylyl-imido-diphosphate (Gpp(NH)p; 400 microM) dialysis into cells with differences between genders. The 5-HTT(-/-) knock-out mutation reduced the current density induced by Gpp(NH)p in females but not in males. These data suggest that the decreased response of 5-HT1A receptors to agonists in 5-HTT(-/-) mutants reflects notably alteration in the coupling between G-proteins and GIRK channels in females but not in males. Accordingly, gender differences in central 5-HT neurotransmission appear to depend-at least in part-on sex-related variations in corresponding receptor-G protein signaling mechanisms.
Collapse
Affiliation(s)
- Alexandre Julien Châu Loucif
- UMR 677, INSERM/UPMC, NeuroPsychoPharmacologie, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, 91 Boulevard de l'Hôpital, 75634 Paris Cedex 13, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
|
72
|
Huddleston GG, Paisley JC, Graham S, Grober MS, Clancy AN. Implants of estradiol conjugated to bovine serum albumin in the male rat medial preoptic area promote copulatory behavior. Neuroendocrinology 2007; 86:249-59. [PMID: 17726305 DOI: 10.1159/000107695] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Accepted: 07/06/2007] [Indexed: 12/26/2022]
Abstract
The expression of mating behavior in male rats is dependent on estrogen-responsive neurons in the medial preoptic area (MPO). Previous reports showed that mating is attenuated if the aromatization of testosterone to estradiol (E2) is blocked in the MPO and that mating is maintained by MPO E2 implants. However, the mechanisms by which E2 exerts its action are not fully understood. It had been thought that E2 acted exclusively by binding to nuclear estrogen receptors to exert it effects; however, recent reports suggest that E2 also binds to membrane-associated receptors activating downstream intracellular cascade responses. In this study, we aimed to determine if an action of E2 at the cell surface is sufficient to support mating behavior. Therefore, either vehicle, E2, or E2 conjugated to bovine serum albumin (BSA-E2: a complex of E2 and a large protein that will not cross the plasma membrane, thereby restricting the action of E2 to cell surface signaling) was chronically administered bilaterally to the MPO of castrated, dihydrotestosterone-treated male rats. Mating behavior was supported by MPO BSA-E2 implants, suggesting that E2 operates in the MPO via a cell surface mechanism to facilitate male rat mating behavior.
Collapse
Affiliation(s)
- Gloria G Huddleston
- Department of Biology, Georgia State University, Atlanta, GA 30302-4010, USA
| | | | | | | | | |
Collapse
|
73
|
Fatehi M, Zidichouski JA, Kombian SB, Saleh TM. 17beta-estradiol attenuates excitatory neurotransmission and enhances the excitability of rat parabrachial neurons in vitro. J Neurosci Res 2006; 84:666-74. [PMID: 16773648 DOI: 10.1002/jnr.20959] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The steroid hormone 17beta-estradiol and its respective receptors have been found in several cardiovascular nuclei in the central nervous system including the parabrachial nucleus. In a previous study, we provided evidence that 17beta-estradiol attenuated an outward potassium conductance in parabrachial neurons of male rats, using an in vitro slice preparation. In this study we sought to enhance the comprehensive information provided previously on estradiol's postsynaptic effects in the parabrachial nucleus by directly examining whether 17beta-estradiol application will modulate excitatory synaptic neurotransmission. Using a pontine slice preparation and whole-cell patch-clamp recording, bath application of either 17beta-estradiol (20-100 muM) or BSA-17beta-estradiol (50 muM) decreased the amplitude of evoked excitatory postsynaptic currents (from 30-60% of control) recorded from neurons in the parabrachial nucleus. The paired pulse ratio was not significantly affected and suggests a post-synaptic site of action. The inhibitory effect on the synaptic current was relatively long-lasting (non-reversible) and was blocked by the selective estrogen receptor antagonist, ICI 182,780. Furthermore, 17beta-estradiol reduced the maximum current elicited by a ramp protocol, increased the input resistance measured between resting membrane potential and action potential threshold and caused an increase in the firing frequency of the cells under current-clamp. In summary, 17beta-estradiol caused 3 effects: first, a depolarization; second, a reduction in evoked excitatory postsynaptic potentials; and third, an enhancement of action potential firing frequency in neurons of the parabrachial nucleus. These observations are consistent with our previous findings and support a role for estrogen in modulating neurotransmission in this nucleus.
Collapse
Affiliation(s)
- Mohammad Fatehi
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | | | | | | |
Collapse
|
74
|
Pozzi S, Benedusi V, Maggi A, Vegeto E. Estrogen Action in Neuroprotection and Brain Inflammation. Ann N Y Acad Sci 2006; 1089:302-23. [PMID: 17261778 DOI: 10.1196/annals.1386.035] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The fertile period of women's life compared to menopause is associated with a lower incidence of degenerative inflammatory diseases. In brain, estrogens ameliorate brain performance and have positive effects on selected neural pathologies characterized by a strong inflammatory component. We thus hypothesized that the inflammatory response is a target of estrogen action; several studies including ours provided strong evidence to support this prediction. Microglia, the brain's inflammatory cells, and circulating monocytes express the estrogen receptors ER-alpha and ER-beta and their responsiveness in vivo and in vitro to pro-inflammatory agents, such as lipopolysaccharide (LPS), is controlled by 17beta-estradiol (E(2)). Susceptibility of central nervous system (CNS) macrophage cells to E(2) is also preserved in animal models of neuroinflammatory diseases, in which ER-alpha seems to be specifically involved. At the molecular level, induction of inflammatory gene expression is blocked by E(2). We recently observed that, differently from conventional anti-inflammatory drugs, E(2) stimulates a nongenomic event that interferes with the LPS signal transduction from the plasma membrane to cytoskeleton and intracellular effectors, which results in the inhibition of the nuclear translocation of NF-kappaB, a transcription factor of inflammatory genes. Interference with NF-kappaB intracellular trafficking is selectively mediated by ER-alpha. In summary, evidence from basic research strongly indicates that the use of estrogenic drugs that can mimic the anti-inflammatory activity of E(2) might trigger beneficial effects against neurodegeneration in addition to carrying out their specific therapeutic function.
Collapse
Affiliation(s)
- Silvia Pozzi
- Center of Excellence on Neurodegenerative Diseases, Department of Pharmacological Sciences, University of Milan, Via Balzaretti, 9, 20133 Milan, Italy
| | | | | | | |
Collapse
|
75
|
Amin Z, Mason GF, Cavus I, Krystal JH, Rothman DL, Epperson CN. The interaction of neuroactive steroids and GABA in the development of neuropsychiatric disorders in women. Pharmacol Biochem Behav 2006; 84:635-43. [PMID: 16860856 DOI: 10.1016/j.pbb.2006.06.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 06/01/2006] [Accepted: 06/12/2006] [Indexed: 11/25/2022]
Abstract
A growing literature suggests that hormonal fluctuations occurring across the menstrual cycle, during and after pregnancy, and during the menopausal transition are associated with onset of affective disorders or exacerbation of existing disorders. This influence of the neuroendocrine system on psychiatric disorders is thought to be mediated by an abnormality in central nervous system response to neuroactive steroids such as estradiol, progesterone, and the progesterone derivative allopregnanolone (ALLO). This interplay is considerably complex as neuroactive steroids modulate the function of multiple neurotransmitter systems throughout various stages of development. While one could choose to study any number of steroid-neurotransmitter interactions, our group in addition to others has focused our investigative efforts on unraveling the contribution of neuroactive steroids to psychiatric syndromes and disorders via their modulation of gamma aminobutyric acid (GABA), the brain's major inhibitory neurotransmitter. The goal of this article is two-fold: to synthesize the clinical and preclinical research focusing on the interplay between neuroactive steroids and GABA as they relate to neuropsychiatric and substance use disorders in women and to integrate data from our laboratory using proton magnetic resonance spectroscopy into this context.
Collapse
|
76
|
Schlichter R, Keller AF, De Roo M, Breton JD, Inquimbert P, Poisbeau P. Fast nongenomic effects of steroids on synaptic transmission and role of endogenous neurosteroids in spinal pain pathways. J Mol Neurosci 2006; 28:33-51. [PMID: 16632874 DOI: 10.1385/jmn:28:1:33] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Revised: 11/30/1999] [Accepted: 06/28/2005] [Indexed: 11/11/2022]
Abstract
Steroids exert long-term modulatory effects on numerous physiological functions by acting at intracellular/nuclear receptors influencing gene transcription. Steroids and neurosteroids can also rapidly modulate membrane excitability and synaptic transmission by interacting with ion channels, that is, ionotropic neurotransmitter receptors or voltage-dependent Ca2+ or K+ channels. More recently, the cloning of a plasma membrane-located G protein-coupled receptor for progestins in various species has suggested that steroids/neurosteroids could also influence second-messenger pathways by directly interacting with specific membrane receptors. Here we review the experimental evidence implicating steroids/neurosteroids in the modulation of synaptic transmission and the evidence for a role of endogenously produced neurosteroids in such modulatory effects. We present some of our recent results concerning inhibitory synaptic transmission in lamina II of the spinal cord and show that endogenous 5alpha-reduced neurosteroids are produced locally in lamina II and modulate synaptic gamma-aminobutyric acid A(GABAA) receptor function during development, as well as during inflammatory pain. The production of 5alpha-reduced neurosteroids is controlled by the endogenous activation of the peripheral benzodiazepine receptor (PBR), which initiates the first step of neurosteroidogenesis by stimulating the translocation of cholesterol across the inner mitochondrial membrane. Tonic neurosteroidogenesis observed in immature animals was decreased during postnatal development, resulting in an acceleration of GABAA receptor-mediated miniature inhibitory postsynaptic current (mIPSC) kinetics observed in the adult. Stimulation of the PBR resulted in a prolongation of GABAergic mIPSCs at all ages and was observed during inflammatory pain. Neurosteroidogenesis might play an important role in the control of nociception at least at the spinal cord level.
Collapse
Affiliation(s)
- Rémy Schlichter
- Institut des Neurosciences Cellulaires et Intégratives-Centre National de la Recherche Scientifique (CNRS), Université Louis Pasteur, 67084 Strasbourg Cedex, France.
| | | | | | | | | | | |
Collapse
|
77
|
Miller JE, Levine RB. Steroid hormone activation of wandering in the isolated nervous system of Manduca sexta. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2006; 192:1049-62. [PMID: 16788816 DOI: 10.1007/s00359-006-0143-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Revised: 05/17/2006] [Accepted: 05/21/2006] [Indexed: 02/02/2023]
Abstract
Steroid hormones modulate motor circuits in both vertebrates and invertebrates. The insect Manduca sexta, with its well-characterized developmental and endocrinological history, is a useful model system in which to study these effects. Wandering is a stage-specific locomotor behavior triggered by the steroid hormone 20-hydroxyecdysone (20E), consisting of crawling and burrowing movements as the animal searches for a pupation site. This study was undertaken to determine whether the wandering motor pattern is activated by direct action of 20E on the CNS. 20E acts on the isolated larval nervous system to induce a fictive motor pattern showing features of crawling and burrowing. The latency of the response to 20E is long, suggestive of a genomic mechanism of action. The abdominal motoneurons or segmental pattern generating circuits are unlikely to be the primary targets of 20E action in inducing fictive wandering. Exposure of the segmental ganglia alone to hormone did not evoke fictive wandering. Therefore, as suggested by an earlier study, the likely site of 20E action is within the brain.
Collapse
Affiliation(s)
- Julie E Miller
- Division of Neurobiology, University of Arizona, Tucson, AZ 85721, USA.
| | | |
Collapse
|
78
|
Rifkind AB. CYP1A in TCDD toxicity and in physiology-with particular reference to CYP dependent arachidonic acid metabolism and other endogenous substrates. Drug Metab Rev 2006; 38:291-335. [PMID: 16684662 DOI: 10.1080/03602530600570107] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Toxicologic and physiologic roles of CYP1A enzyme induction, the major biochemical effect of aryl hydrocarbon receptor activation by TCDD and other receptor ligands, are unknown. Evidence is presented that CYP1A exerts biologic effects via metabolism of endogenous substrates (i.e., arachidonic acid, other eicosanoids, estrogens, bilirubin, and melatonin), production of reactive oxygen, and effects on K(+) and Ca(2+) channels. These interrelated pathways may connect CYP1A induction to TCDD toxicities, including cardiotoxicity, vascular dysfunction, and wasting. They may also underlie homeostatic roles for CYP1A, especially when transiently induced by common chemical exposures and environmental conditions (i.e., tryptophan photoproducts, dietary indoles, and changes in oxygen tension).
Collapse
Affiliation(s)
- Arleen B Rifkind
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
79
|
Loyd DR, Murphy AZ. Sex differences in the anatomical and functional organization of the periaqueductal gray-rostral ventromedial medullary pathway in the rat: a potential circuit mediating the sexually dimorphic actions of morphine. J Comp Neurol 2006; 496:723-38. [PMID: 16615128 PMCID: PMC2823481 DOI: 10.1002/cne.20962] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies have demonstrated that morphine, administered systemically or directly into the periaqueductal gray (PAG), produces a significantly greater degree of antinociception in males in comparison with females. Because the midbrain PAG and its descending projections to the rostral ventromedial medulla (RVM) constitute an essential neural circuit for opioid-based analgesia, the present studies were conducted to determine whether sex differences in the anatomical organization of the PAG-RVM pathway, and its activation during persistent inflammatory pain, could account for sex-based differences in opioid analgesia. In the rat, retrograde tracing was combined with Fos immunocytochemistry to investigate sexual dimorphism in the organization of the PAG-RVM circuit and its activation by persistent inflammatory pain induced by intraplantar injection of complete Freund's adjuvant (CFA). The ability of morphine to suppress the activation of the PAG-RVM circuit was also examined. Sexually dimorphic retrograde labeling was observed within the dorsomedial and lateral/ventrolateral PAG at all rostrocaudal levels, with females having significantly more PAG-RVM output neurons in comparison with males. While no sex differences were noted in the activation of the PAG by persistent inflammatory pain, significantly more PAG-RVM cells were activated in males in comparison with females. Systemic administration of morphine significantly suppressed CFA-induced Fos in the PAG in males only. The results of these studies demonstrate that both the anatomical organization and the functional activation of the PAG-RVM circuit are sexually dimorphic and may provide the anatomical substrate for sex-based differences in morphine analgesia.
Collapse
Affiliation(s)
- Dayna R Loyd
- Department of Biology, Center for Behavioral Neuroscience, Georgia State University, Atlanta, 30302-4010, USA
| | | |
Collapse
|
80
|
Beckett EAH, McCloskey C, O'Kane N, Sanders KM, Koh SD. Effects of female steroid hormones on A-type K+ currents in murine colon. J Physiol 2006; 573:453-68. [PMID: 16581861 PMCID: PMC1779718 DOI: 10.1113/jphysiol.2006.107375] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Idiopathic constipation is higher in women of reproductive age than postmenopausal women or men, suggesting that female steroid hormones influence gastrointestinal motility. How female hormones affect motility is unclear. Colonic motility is regulated by ion channels in colonic myocytes. Voltage-dependent K(+) channels serve to set the excitability of colonic muscles. We investigated regulation of Kv 4.3 channel expression in response to acute or chronic changes in female hormones. Patch clamp experiments and quantitative PCR were used to compare outward currents and transcript expression in colonic myocytes from male, non-pregnant, pregnant and ovariectomized mice. Groups of ovariectomized mice received injections of oestrogen or progesterone to investigate the effects of hormone replacement. The capacitance of colonic myocytes from non-pregnant females was larger than in males. Net outward current density in male and ovariectomized mice was higher than in non-pregnant females and oestrogen-treated ovariectomized mice. Current densities in late pregnancy were lower than in female controls. Progesterone had no effect on outward currents. A-type currents were decreased in non-pregnant females compared with ovariectomized mice, and were further decreased by pregnancy or oestrogen replacement. Kv 4.3 transcripts did not differ significantly between groups; however, expression of the potassium channel interacting protein KChIP1 was elevated in ovariectomized mice compared with female controls and oestrogen-treated ovariectomized mice. Delayed rectifier currents were not affected by oestrogen. In the mouse colon, oestrogen suppresses A-type currents, which are important for regulating excitability. These observations suggest a possible link between female hormones and altered colonic motility associated with menses, pregnancy and menopause.
Collapse
Affiliation(s)
- Elizabeth A H Beckett
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, MS 352, Reno, NV 89557, USA.
| | | | | | | | | |
Collapse
|
81
|
Abstract
Gonadal steroids are metabolized in target cells and then interact with specific receptors to exert genomic and nongenomic effects. Complex feedback loops that involve the immune-neuroendocrine axis, limbic system, and gonadal steroids play a vital role in the adaptation to critical illness. Preclinical studies demonstrate adverse physiological effects of androgens on the cardiovascular and immune systems despite its purported anabolic effects. Similar models also demonstrate salutary effects of estrogens on these systems. Thus, during the catabolic phases of acute and chronic critical illness, estrogen, and not androgen, therapy may prove to be a valuable intervention. However, during the post-critical illness recovery phase, when anabolism is critical, androgen therapy may still be useful and safe.
Collapse
Affiliation(s)
- Jeffrey I Mechanick
- Division of Endocrinology, Diabetes and Bone Disease, Mount Sinai School of Medicine, 1192 Park Avenue, New York, NY 10128, USA.
| | | |
Collapse
|
82
|
Wang X, Traub RJ, Murphy AZ. Persistent pain model reveals sex difference in morphine potency. Am J Physiol Regul Integr Comp Physiol 2006; 291:R300-6. [PMID: 16497818 PMCID: PMC2856616 DOI: 10.1152/ajpregu.00022.2006] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Central or systemic administration of agonists directed at the mu or delta opiate receptors generally produce a greater degree of analgesia in males than in females. To date, most studies examining sex-based differences in opioid analgesia have used acute noxious stimuli (i.e., tail-flick and hot plate test); thus the potential dimorphic response of centrally acting opiates in the alleviation of persistent inflammatory pain is not well established. In the present study, right hind paw withdrawal latency (PWL) to radiant thermal stimuli was measured in intact male and cycling female Sprague-Dawley rats before and after unilateral hind paw injection of the inflammatory agent complete Freund's adjuvant (CFA). Control animals received intraplantar injection of saline. Twenty four hours after CFA or saline injection, animals received either saline or morphine bisulfate (0.5-15 mg/kg sc). Separate groups of control or inflamed animals were tested on their responsiveness to morphine at 7, 14, and 21 days post-CFA or saline. No sex differences were noted for baseline PWLs, and females displayed slightly less thermal hyperalgesia at 24 h post-CFA. At all morphine doses administered, both the antihyperalgesic effects of morphine in the inflamed animals and the antinociceptive effects of morphine in control animals were significantly greater in males compared with females. Similarly, in males, the antihyperalgesic effects of morphine increased significantly at 7-21 days post-CFA; no significant shift in morphine potency was noted for females. These studies demonstrate sex-based differences in the effects of morphine on thermal hyperalgesia in a model of persistent inflammatory pain.
Collapse
Affiliation(s)
- Xiaoya Wang
- Department of Biology, Center for Behavioral Neuroscience, Georgia State University, 24 Peachtree Center Ave, 402 Kell Hall, Atlanta, GA 30303-3088
| | - Richard J. Traub
- Department of Biological Sciences, Dental School, University of Maryland, Baltimore, MD 21201
- Research Center for Neuroendocrine Influences on Pain, University of Maryland, Baltimore, MD 21201
| | - Anne Z. Murphy
- Department of Biology, Center for Behavioral Neuroscience, Georgia State University, 24 Peachtree Center Ave, 402 Kell Hall, Atlanta, GA 30303-3088
- Research Center for Neuroendocrine Influences on Pain, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
83
|
Howell N, Dykens J, Moos WH. Alzheimer's disease, estrogens, and clinical trials: a case study in drug development for complex disorders. Drug Dev Res 2006. [DOI: 10.1002/ddr.20046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
84
|
Kushner SA, Guze BH. Treatment of psychomotor agitation and self-injurious behavior with estrogen and progesterone in a patient with Sanfilippo syndrome. Gen Hosp Psychiatry 2005; 27:298-300. [PMID: 15993263 DOI: 10.1016/j.genhosppsych.2005.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Accepted: 02/17/2005] [Indexed: 11/28/2022]
|
85
|
Cone RD. Anatomy and regulation of the central melanocortin system. Nat Neurosci 2005; 8:571-8. [PMID: 15856065 DOI: 10.1038/nn1455] [Citation(s) in RCA: 1120] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2005] [Accepted: 03/15/2005] [Indexed: 12/12/2022]
Abstract
The central melanocortin system is perhaps the best-characterized neuronal pathway involved in the regulation of energy homeostasis. This collection of circuits is unique in having the capability of sensing signals from a staggering array of hormones, nutrients and afferent neural inputs. It is likely to be involved in integrating long-term adipostatic signals from leptin and insulin, primarily received by the hypothalamus, with acute signals regulating hunger and satiety, primarily received by the brainstem. The system is also unique from a regulatory point of view in that it is composed of fibers expressing both agonists and antagonists of melanocortin receptors. Given that the central melanocortin system is an active target for development of drugs for the treatment of obesity, diabetes and cachexia, it is important to understand the system in its full complexity, including the likelihood that the system also regulates the cardiovascular and reproductive systems.
Collapse
Affiliation(s)
- Roger D Cone
- Vollum Institute and the Center for the Study of Weight Regulation, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA.
| |
Collapse
|
86
|
Melcangi RC, Cavarretta ITR, Ballabio M, Leonelli E, Schenone A, Azcoitia I, Miguel Garcia-Segura L, Magnaghi V. Peripheral nerves: a target for the action of neuroactive steroids. ACTA ACUST UNITED AC 2005; 48:328-38. [PMID: 15850671 DOI: 10.1016/j.brainresrev.2004.12.021] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 12/09/2004] [Indexed: 10/25/2022]
Abstract
Peripheral nervous system possesses both classical and non-classical steroid receptors and consequently may represent a target for the action of neuroactive steroids. The present review summarizes the state of art of this intriguing field of research reporting data which indicate that neuroactive steroids, like for instance progesterone, dihydroprogesterone, tetrahydroprogesterone, dihydrotestosterone and 3alpha-diol, stimulate the expression of two important proteins of the myelin of peripheral nerves, the glycoprotein P0 (P0) and the peripheral myelin protein 22 (PMP22). Interestingly, the mechanisms by which neuroactive steroids exert their effects involve classical steroid receptors, like for instance progesterone and androgen receptors, in case of P0 and non-classical steroid receptors, like GABA(A) receptor, in case of PMP22. Moreover, neuroactive steroids not only control the expression of these specific myelin proteins, but also influence the morphology of myelin sheaths and axons suggesting that these molecules may represent an interesting new therapeutic approach to maintain peripheral nerve integrity during neurodegenerative events.
Collapse
|
87
|
The neural basis of puberty and adolescence. Nat Neurosci 2004; 7:1040-7. [PMID: 15452575 DOI: 10.1038/nn1326] [Citation(s) in RCA: 657] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2004] [Accepted: 08/25/2004] [Indexed: 01/01/2023]
Abstract
The pubertal transition to adulthood involves both gonadal and behavioral maturation. A developmental clock, along with permissive signals that provide information on somatic growth, energy balance and season, time the awakening of gonadotropin releasing hormone (GnRH) neurons at the onset of puberty. High-frequency GnRH release results from disinhibition and activation of GnRH neurons at puberty onset, leading to gametogenesis and an increase in gonadal steroid hormone secretion. Steroid hormones, in turn, both remodel and activate neural circuits during adolescent brain development, leading to the development of sexual salience of sensory stimuli, sexual motivation, and expression of copulatory behaviors in specific social contexts. These influences of hormones on reproductive behavior depend in part on changes in the adolescent brain that occur independently of gonadal maturation. Reproductive maturity is therefore the product of developmentally timed, brain-driven and recurrent interactions between steroid hormones and the adolescent nervous system.
Collapse
|
88
|
Plummer HK, Yu Q, Cakir Y, Schuller HM. Expression of inwardly rectifying potassium channels (GIRKs) and beta-adrenergic regulation of breast cancer cell lines. BMC Cancer 2004; 4:93. [PMID: 15603589 PMCID: PMC539303 DOI: 10.1186/1471-2407-4-93] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 12/16/2004] [Indexed: 01/11/2023] Open
Abstract
Background Previous research has indicated that at various organ sites there is a subset of adenocarcinomas that is regulated by beta-adrenergic and arachidonic acid-mediated signal transduction pathways. We wished to determine if this regulation exists in breast adenocarcinomas. Expression of mRNA that encodes a G-protein coupled inwardly rectifying potassium channel (GIRK1) has been shown in tissue samples from approximately 40% of primary human breast cancers. Previously, GIRK channels have been associated with beta-adrenergic signaling. Methods Breast cancer cell lines were screened for GIRK channels by RT-PCR. Cell cultures of breast cancer cells were treated with beta-adrenergic agonists and antagonists, and changes in gene expression were determined by both relative competitive and real time PCR. Potassium flux was determined by flow cytometry and cell signaling was determined by western blotting. Results Breast cancer cell lines MCF-7, MDA-MB-361 MDA-MB 453, and ZR-75-1 expressed mRNA for the GIRK1 channel, while MDA-MB-468 and MDA-MB-435S did not. GIRK4 was expressed in all six breast cancer cell lines, and GIRK2 was expressed in all but ZR-75-1 and MDA-MB-435. Exposure of MDA-MB-453 cells for 6 days to the beta-blocker propranolol (1 μM) increased the GIRK1 mRNA levels and decreased beta2-adrenergic mRNA levels, while treatment for 30 minutes daily for 7 days had no effect. Exposure to a beta-adrenergic agonist and antagonist for 24 hours had no effect on gene expression. The beta adrenergic agonist, formoterol hemifumarate, led to increases in K+ flux into MDA-MB-453 cells, and this increase was inhibited by the GIRK channel inhibitor clozapine. The tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a high affinity agonist for beta-adrenergic receptors stimulated activation of Erk 1/2 in MDA-MB-453 cells. Conclusions Our data suggests β-adrenergic receptors and GIRK channels may play a role in breast cancer.
Collapse
Affiliation(s)
- Howard K Plummer
- Molecular Cancer Analysis Laboratory, Department of Pathobiology, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996-4542, USA
| | - Qiang Yu
- Molecular Cancer Analysis Laboratory, Department of Pathobiology, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996-4542, USA
| | - Yavuz Cakir
- Experimental Oncology Laboratory, Department of Pathobiology, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996-4542, USA
- Current address: Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Hildegard M Schuller
- Experimental Oncology Laboratory, Department of Pathobiology, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996-4542, USA
| |
Collapse
|