Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 52(1): 2756-2770, 2020 07.
Article in English | MEDLINE | ID: mdl-32243028

ABSTRACT

Including sex is of paramount importance in preclinical and clinical stroke researches, and molecular studies dealing in depth with sex differences in stroke pathophysiology are needed. To gain insight into the molecular sex dimorphism of ischaemic stroke in rat cerebral cortex, male and female adult rats were subjected to transient middle cerebral artery occlusion. The expression of neuroglobin (Ngb) and other functionally related molecules involved in sex steroid signalling (oestrogen and androgen receptors), steroidogenesis (StAR, TSPO and aromatase) and autophagic activity (LC3B-II/LC3B-I ratio, UCP2 and HIF-1α) was assessed in the ipsilateral ischaemic and contralateral non-ischaemic hemispheres. An increased expression of Ngb was detected in the injured female cerebral cortex. In contrast, increased expression of oestrogen receptor α, GPER, StAR, TSPO and UCP2, and decreased androgen receptor expression were detected in the injured male cortex. In both sexes, the ischaemic insult induced an upregulation of LC3B-II/-I ratio, indicative of increased autophagy. Therefore, the cerebral cortex activates both sex-specific and common molecular responses with neuroprotective potential after ischaemia-reperfusion, which globally results in similar stroke outcome in both sexes. Nonetheless, these different potential molecular targets should be taken into account when neuroprotective drugs aiming to reduce brain damage in ischaemic stroke are investigated.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Animals , Autophagy , Cerebral Cortex , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery , Male , Neuroglobin , Rats , Rats, Sprague-Dawley , Sex Characteristics , Steroids
2.
J Neuroendocrinol ; 32(1): e12774, 2020 01.
Article in English | MEDLINE | ID: mdl-31323169

ABSTRACT

Cardiolipin (CL) is a phospholipid that is almost exclusively located in the inner mitochondrial membrane of eukaryotic cells. As a result of its unique structure and distribution, CL establishes non-covalent bonds with a long list of proteins involved in ATP production, mitochondria biogenesis, mitophagy and apoptosis. Thus, the amount of CL, as well as its fatty acid composition and location, strongly impacts upon mitochondrial-dependent functions and therefore the metabolic homeostasis of different tissues. The brain is particularly sensitive to mitochondrial dysfunction as a result of its high metabolic demand. Several mitochondrial related-neurodegenerative disorders, as well as physiological ageing, show altered CL metabolism. Furthermore, mice lacking enzymes involved in CL synthesis show cognitive impairments. CL content and metabolism are regulated by gonadal hormones in the developing and adult brain. In neuronal cultures, oestradiol increases CL content, whereas adult ovariectomy decreases CL content and alters CL metabolism in the hippocampal mitochondria. Transient sex differences in brain CL metabolism have been detected during development. At birth, brain CL has a higher proportion of unsaturated fatty acids in the brain of male mice than in the brain of females. In addition, the expression of enzymes involved in CL de novo and recycling synthetic pathways is higher in males. Most of these sex differences are abolished by the neonatal androgenisation of females, suggesting a role for testosterone in the generation of sex differences in brain CL. The regulation of brain CL by gonadal hormones may be linked to their homeostatic and protective actions in neural cells, as well as the manifestation of sex differences in neurodegenerative disorders.


Subject(s)
Brain/metabolism , Cardiolipins/metabolism , Gonadal Steroid Hormones/metabolism , Neurons/metabolism , Animals , Female , Humans , Male , Mitochondria/metabolism , Sex Characteristics
3.
Cell Death Discov ; 5: 85, 2019.
Article in English | MEDLINE | ID: mdl-30962951

ABSTRACT

In the nervous system, Notch pathway has a prominent role in the control of neuronal morphology and in the determination of the astrocyte fate. However, the role of Notch in morphological astrocyte plasticity is unknown. Here, we have explored the role of Notch activity on the morphological reactivity of primary astrocytes in response to LPS, an inflammatory stimulus. We found that LPS induces reactive astrocyte morphology by the inhibition of Notch signaling via NFκB activation and Jagged upregulation. In contrast, IGF-1, an anti-inflammatory molecule, inhibits LPS-induced reactive astrocyte morphological phenotype by enhancing Notch signaling through the inhibition of NFκB and the activation of MAPK. Therefore, Notch signaling pathway emerges as a mediator of the regulation of astrocyte morphology by inflammatory and anti-inflammatory stimuli.

4.
Mol Neurobiol ; 56(3): 1665-1680, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29916142

ABSTRACT

Obesity is associated with an increase in the brain levels of saturated free fatty acids, such as palmitic acid (PA). Previous studies have shown that PA exerts proinflammatory actions and reduces cell viability in astrocyte cultures. In this study, we have assessed whether an alteration in autophagy is involved in the effects of PA on astrocytes. Primary astrocytes were obtained from the cerebral cortex of male and female CD1 mouse pups and were incubated for 4.5 or 24 h with 250-500 µM PA. PA increased the levels of LC3-II, an autophagosome marker, and reduced LC3-II flux in astrocytes, suggesting a blockade of autophagy. This effect was observed both after 4.5 and 24 h of treatment with PA. PA had additional effects after treatment for 24 h, increasing the expression of proinflammatory cytokines, decreasing cell viability, and increasing the levels of an endoplasmic reticulum stress marker. In addition, PA decreased the expression of estrogen receptors, but only in female astrocytes. However, the treatment with estradiol, estrogen receptor agonists, or inhibitor of estradiol synthesis did not counteract the action of PA on cell viability. Rapamycin, an autophagy inducer, was unable to prevent the effect of PA on cell viability. In addition, hydroxychloroquine, an autophagy blocker, did not cause per se astrocyte death. These findings suggest that the effect of PA on autophagy is not sufficient to induce astrocyte loss, which is only observed when prolonged PA treatment causes other alterations in astrocytes, such as increased inflammation and endoplasmic reticulum stress.


Subject(s)
Astrocytes/drug effects , Autophagy/drug effects , Cerebral Cortex/drug effects , Palmitic Acid/pharmacology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Endoplasmic Reticulum Stress/drug effects , Estradiol/pharmacology , Female , Male , Mice , Sex Characteristics , Sirolimus/pharmacology
5.
Sci Rep ; 7: 43878, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262723

ABSTRACT

Cardiolipin (CL) is a mitochondrial-specific phospholipid. CL content and acyl chain composition are crucial for energy production. Given that estradiol induces CL synthesis in neurons, we aimed to assess CL metabolism in the cerebral cortex (CC) of male and female mice during early postnatal life, when sex steroids induce sex-dimorphic maturation of the brain. Despite the fact that total amount of CL was similar, its fatty acid composition differed between males and females at birth. In males, CL was more mature (lower saturation ratio) and the expression of the enzymes involved in synthetic and remodeling pathways was higher, compared to females. Importantly, the sex differences found in CL metabolism were due to the testosterone peak that male mice experience perinatally. These changes were associated with a higher expression of UCP-2 and its activators in the CC of males. Overall, our results suggest that the perinatal testosterone surge in male mice regulates CL biosynthesis and remodeling in the CC, inducing a sex-dimorphic fatty acid composition. In male's CC, CL is more susceptible to peroxidation, likely explaining the testosterone-dependent induction of neuroprotective molecules such as UCP-2. These differences may account for the sex-dependent mitochondrial susceptibility after perinatal hypoxia/ischemia.


Subject(s)
Cardiolipins/metabolism , Cerebral Cortex/metabolism , Mitochondria/metabolism , Testosterone/metabolism , Animals , Animals, Newborn , Cerebral Cortex/growth & development , Fatty Acids/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice, Inbred C57BL , Sex Factors , Time Factors , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism
6.
Endocrinology ; 157(10): 3983-3993, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27547849

ABSTRACT

The incidence of traumatic brain injuries (TBIs) in humans has rapidly increased in the last ten years. The most common causes are falls and car accidents. Approximately 80 000-90 000 persons per year will suffer some permanent disability as a result of the lesion, and one of the most common symptoms is the decline of hormone levels, also known as post-TBI hormonal deficiency syndrome. This issue has become more and more important, and many studies have focused on shedding some light on it. The hormonal decline affects not only gonadal steroid hormones but also neuroactive steroids, which play an important role in TBI recovery by neuroprotective and neurotrophic actions. The present work used an adolescent close-head murine model to analyze brain and plasma neurosteroid level changes after TBI and to establish correlations with edema and neurological impairments, 2 of the hallmarks of TBI. Our results showed changes in brain pregnenolone, testosterone, dihydrotestosterone (DHT), and 3α-diol levels whereas in plasma, the changes were present in progesterone, DHT, 3α-diol, and 3ß-diol. Within them, pregnenolone, progesterone, DHT, and 3α-diol levels positively correlated with edema formation and neurological score, whereas testosterone inversely correlated with these 2 variables. These findings suggest that changes in the brain levels of some neuroactive steroids may contribute to the alterations in brain function caused by the lesion and that plasma levels of some neuroactive steroids could be good candidates of blood markers to predict TBI outcome.


Subject(s)
Brain Edema/metabolism , Brain Injuries, Traumatic/blood , Brain/metabolism , Neurotransmitter Agents/blood , Androstane-3,17-diol/analogs & derivatives , Androstane-3,17-diol/blood , Animals , Brain Edema/etiology , Brain Injuries, Traumatic/complications , Dihydrotestosterone/blood , Male , Mice , Pregnenolone/metabolism , Progesterone/blood , Random Allocation , Testosterone/blood , Weight Loss
7.
Prog Neurobiol ; 144: 5-26, 2016 09.
Article in English | MEDLINE | ID: mdl-27283249

ABSTRACT

In the last years there has been a considerable advance in the knowledge on the regulation of astrocytes by sex steroids under physiological and pathological conditions. By the activation of a variety of nuclear and membrane receptors, sex steroid hormones regulate the functions of astrocytes and their communication with other cell types in the central nervous system. Under physiological conditions astrocytes participate in the neuroendocrine and behavioral actions of gonadal steroids, as well as in the hormonal control of brain tissue homeostasis. Under pathological conditions astrocytes mediate, at least partially, the neuroprotective effects of gonadal steroid hormones; given that sex steroids modulate reactive astrogliosis and reduce the release of pro-inflammatory molecules by these cells. Given the side effects that sex steroids may have when administered systemically, a number of synthetic agonists of the receptors for gonadal steroid hormones in the nervous system have been developed, and may be considered for clinical use after brain injury as potential enhancers of the neuroprotective astrocytic functions.


Subject(s)
Astrocytes/metabolism , Brain Injuries/drug therapy , Brain Injuries/metabolism , Gonadal Steroid Hormones/metabolism , Gonadal Steroid Hormones/therapeutic use , Neuroprotection/physiology , Animals , Gonadal Steroid Hormones/agonists , Humans , Neuroprotection/drug effects
8.
Exp Neurol ; 279: 274-282, 2016 May.
Article in English | MEDLINE | ID: mdl-27006282

ABSTRACT

BACKGROUND AND PURPOSE: The rates for traumatic brain injury (TBI) have risen in the last decade. Studies in animal models and clinical trials have not yet resulted in an effective treatment for TBI. Leptin, a 16kDa peptidic hormone is mainly known as a regulator of energy balance and has been shown to exert neuroprotective effects in different models of brain pathology. In this study, we have assessed whether leptin exerts protective actions in a TBI mouse model. In addition, the possible implication of CB2 cannabinoid receptor in leptin actions has been explored, since it is known that the endocannabinoid system interacts with leptin and actively participates in brain recovery after lesions. METHODS: Swiss (CD1) male mice were subjected to weigh-drop model for TBI. Prior to the lesion, mice were injected with an antagonist of CB2 receptor (AM630) or the vehicle and immediately after TBI, they received leptin or vehicle treatment. Data were analyzed using a two-way ANOVA or the non-parametric test Kruskal-Wallis when appropriate. For correlation analyses, Spearman's rho test, followed by linear regression test, was used. RESULTS: TBI induced a neurological deficit, which was improved by leptin treatment. Leptin recovered several parameters affected by TBI, including the expression of cannabinoid receptors, axonal injury marker and neuroinflammatory components. The effects of leptin were prevented or reduced when it was administered in combination with the CB2 receptor antagonist, AM630. CONCLUSIONS AND IMPLICATIONS: Since some of the beneficial effects of leptin were not evident in the presence of AM630, our results suggest that CB2 receptor might be involved in the full expression of the neuroprotective effects of the hormone. These findings open new avenues for the study of leptin as a therapeutic treatment for TBI and enhance the importance of CB2 receptor in TBI pathophysiology and recovery.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Injuries/drug therapy , Leptin/pharmacology , Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB2/drug effects , Animals , Axons/pathology , Behavior, Animal/drug effects , Body Weight/drug effects , Brain Injuries/pathology , Brain Injuries/psychology , Cannabinoid Receptor Antagonists/pharmacology , Cytokines/metabolism , Indoles/pharmacology , Male , Mice , Nervous System Diseases/etiology , Nervous System Diseases/psychology , Neuritis/pathology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/antagonists & inhibitors
9.
Endocrinology ; 156(10): 3706-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26200092

ABSTRACT

The selective estrogen receptor modulator raloxifene reduces blood pressure in hypertensive postmenopausal women. In the present study we have explored whether raloxifene regulates gene expression of arginine vasopressin (AVP), which is involved in the pathogenesis of hypertension. The effect of raloxifene was assessed in human female SH-SY5Y neuroblastoma cells, which have been recently identified as a suitable cellular model to study the estrogenic regulation of AVP. Raloxifene, within a concentration ranging from 10(-10) M to 10(-6) M, decreased the mRNA levels of AVP in SH-SY5Y cells with maximal effect at 10(-7) M. This effect of raloxifene was imitated by an agonist (±)-1-[(3aR*,4S*,9bS*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone of G protein-coupled estrogen receptor-1 (GPER) and blocked by an antagonist (3aS*,4R*,9bR*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-3H-cyclopenta[c]quinoline of GPER and by GPER silencing. Raloxifene induced a time-dependent increase in the level of phosphorylated ERK1 and ERK2, by a mechanism blocked by the GPER antagonist. The treatment of SH-SY5Y cells with either a MAPK/ERK kinase 1/2-specific inhibitor (1,4-diamino-2, 3-dicyano-1,4-bis(2-aminophenylthio)butadine) or a protein kinase C inhibitor (sotrastaurin) blocked the effects of raloxifene on the phosphorylation of ERK1/2 and the regulation of AVP mRNA levels. These results reveal a mechanism mediating the regulation of AVP expression by raloxifene, involving the activation of GPER, which in turn activates protein kinase C, MAPK/ERK kinase, and ERK. The regulation of AVP by raloxifene and GPER may have implications for the treatment of blood hypertension(.).


Subject(s)
Arginine Vasopressin/genetics , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Signaling System/drug effects , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Blotting, Western , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyrroles/pharmacology , Quinazolines/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Selective Estrogen Receptor Modulators/pharmacology
10.
PLoS One ; 10(6): e0128782, 2015.
Article in English | MEDLINE | ID: mdl-26039099

ABSTRACT

Traumatic brain injury (TBI) incidence rises during adolescence because during this critical neurodevelopmental period some risky behaviors increase. The purpose of this study was to assess the contribution of cannabinoid receptors (CB1 and CB2), blood brain barrier proteins (AQP4) and astrogliosis markers (vimentin) to neurological deficit and brain edema formation in a TBI weight drop model in adolescent male mice. These molecules were selected since they are known to change shortly after lesion. Here we extended their study in three different timepoints after TBI, including short (24h), early mid-term (72h) and late mid-term (two weeks). Our results showed that TBI induced an increase in brain edema up to 72 h after lesion that was directly associated with neurological deficit. Neurological deficit appeared 24 h after TBI and was completely recovered two weeks after trauma. CB1 receptor expression decreased after TBI and was negatively correlated with edema formation and behavioral impairments. CB2 receptor increased after injury and was associated with high neurological deficit whereas no correlation with edema was found. AQP4 increased after TBI and was positively correlated with edema and neurological impairments as occurred with vimentin expression in the same manner. The results suggest that CB1 and CB2 differ in the mechanisms to resolve TBI and also that some of their neuroprotective effects related to the control of reactive astrogliosis may be due to the regulation of AQP4 expression on the end-feet of astrocytes.


Subject(s)
Aquaporin 4/genetics , Brain Edema/genetics , Brain Injuries/genetics , Cognition Disorders/genetics , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Vimentin/genetics , Animals , Aquaporin 4/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Brain Edema/complications , Brain Edema/metabolism , Brain Edema/pathology , Brain Injuries/complications , Brain Injuries/metabolism , Brain Injuries/pathology , Cognition Disorders/complications , Cognition Disorders/metabolism , Cognition Disorders/pathology , Exploratory Behavior , Gene Expression Regulation , Male , Mice , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction , Stereotyped Behavior , Vimentin/metabolism
11.
Glia ; 63(11): 1966-1981, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26037411

ABSTRACT

Several brain disorders associated with neuroinflammation show sex differences in their incidence, onset, progression and/or outcome. The different regulation of the neuroinflammatory response in males and females could underlie these sex differences. In this study, we have explored whether reactive gliosis after a penetrating cortical injury exhibits sex differences. Males presented a higher density of Iba1 immunoreactive cells in the proximity of the wound (0-220 µm) than females. This sex difference was due to a higher number of Iba1 immunoreactive cells with nonreactive morphology. In addition microglia/macrophages in that region expressed arginase-1, marker of alternatively activated microglia, and the neuroprotective protein Neuroglobin, in a greater proportion in males than in females. No sex differences were found in the number of astrocytes around the lesion. However, the percentage of astrocytes expressing chemokine (C-C motif) ligand 2 (CCL2), involved in recruitment of immune cells and gliosis regulation, was higher in males. Males also presented a significantly higher density of neurons in the lesion edge than females. These findings indicate that male and female mice have different neuroinflammatory responses after a cortical stab wound injury and suggest that sex differences in reactive gliosis may contribute to sex differences in neuroinflammatory diseases. GLIA 2015;63:1966-1981.

12.
Psychoneuroendocrinology ; 56: 1-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25770855

ABSTRACT

Traumatic brain injury (TBI) is an important cause of disability in humans. Neuroactive steroids, such as progesterone and dehydroepiandrosterone (DHEA), are neuroprotective in TBI models. However in order to design potential neuroprotective strategies based on neuroactive steroids it is important to determine whether its brain levels are altered by TBI. In this study we have used a weight-drop model of TBI in young adult female mice to determine the levels of neuroactive steroids in the brain and plasma at 24h, 72 h and 2 weeks after injury. We have also analyzed whether the levels of neuroactive steroids after TBI correlated with the neurological score of the animals. TBI caused neurological deficit detectable at 24 and 72 h, which recovered by 2 weeks after injury. Brain levels of progesterone, tetrahydroprogesterone (THP), isopregnanolone and 17ß-estradiol were decreased 24h, 72 h and 2 weeks after TBI. DHEA and brain testosterone levels presented a transient decrease at 24h after lesion. Brain levels of progesterone and DHEA showed a positive correlation with neurological recovery. Plasma analyses showed that progesterone was decreased 72 h after lesion but, in contrast with brain progesterone, its levels did not correlate with neurological deficit. These findings indicate that TBI alters the levels of neuroactive steroids in the brain with independence of its plasma levels and suggest that the pharmacological increase in the brain of the levels of progesterone and DHEA may result in the improvement of neurological recovery after TBI.


Subject(s)
Brain Chemistry , Brain Injuries/metabolism , Brain Injuries/pathology , Dehydroepiandrosterone/metabolism , Progesterone/metabolism , Recovery of Function , Animals , Behavior, Animal , Body Weight/physiology , Brain Edema/etiology , Brain Edema/pathology , Brain Injuries/psychology , Dehydroepiandrosterone/blood , Estrous Cycle/physiology , Female , Mice , Neurotransmitter Agents/analysis , Neurotransmitter Agents/metabolism , Progesterone/blood
13.
Mol Cell Endocrinol ; 389(1-2): 48-57, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24444786

ABSTRACT

Neuroprotective actions of 17ß-estradiol (estradiol) are in part mediated by direct actions on neurons. Astrocytes, which play an essential role in the maintenance of the homeostasis of neural tissue, express estrogen receptors and are also involved in the neuroprotective actions of estradiol in the brain. Estradiol controls gliosis and regulates neuroinflammation, edema and glutamate transport acting on astrocytes. In addition, the hormone regulates the release of neurotrophic factors and other neuroprotective molecules by astrocytes. In addition, reactive astrocytes are a local source of neuroprotective estradiol for the injured brain. Since estradiol therapy is not free from peripheral risks, alternatives for the hormone have been explored. Some selective estrogen receptor modulators (SERMs), which are already in use in clinical practice for the treatment of breast cancer, osteoporosis or menopausal symptoms, exert similar actions to estradiol on astrocytes. Therefore, SERMs represent therapeutic alternatives to estradiol for the activation of astroglia-mediated neuroprotective mechanisms.


Subject(s)
Astrocytes/metabolism , Estradiol/metabolism , Neuroprotective Agents/metabolism , Selective Estrogen Receptor Modulators/metabolism , Animals , Brain/metabolism , Estrogens/metabolism , Humans , Neurons/metabolism , Receptors, Estrogen/metabolism
14.
Neurotox Res ; 25(3): 271-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23943137

ABSTRACT

A low dose of the organophosphorus insecticide dimethoate (DMT) produces oxidation of lipids and proteins and impairs mitochondrial function in the brain of male rats, together with a reduction of gonadal hormones in plasma. Here, we have assessed whether DMT affected the expression of inflammatory molecules, the production of reactive oxygen species (ROS), and the expression of steroidogenic proteins and estrogen receptors in cortical astrocyte-enriched cultures obtained separately from male and female CD1 mice pups. Furthermore, we have analyzed whether estradiol may counteract the effects of DMT. A dose of DMT (2 µg/mL) did not affect cell viability, increased interleukin (IL) 6, IL1ß, tumor necrosis factor (TNF)α, interferon-γ-inducible protein 10 (IP10), ERß, steroidogenic acute regulatory protein, and aromatase mRNA levels and ERα protein levels in male but not in female cultures. Estradiol decreased the mRNA levels of IL6, IP10, TNFα, and IL1ß in male but not in female cultures treated with DMT. The effect of estradiol was prevented by the ER antagonist ICI 182,780, fully imitated by an ERß agonist and partially imitated by an ERα agonist. Furthermore, DMT increased the production of ROS in male astrocytes while estradiol reduced ROS production to control levels. These findings indicate that a sublethal dose of DMT alters astrocyte function. The selective action of estradiol on male astrocytes and the sexually dimorphic action of DMT suggest that the pesticide may have different neurological outcomes in males and females.


Subject(s)
Astrocytes/drug effects , Astrocytes/physiology , Dimethoate/toxicity , Estradiol/pharmacology , Estrogens/pharmacology , Insecticides/toxicity , Animals , Astrocytes/immunology , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Cerebral Cortex/physiology , Female , Male , Mice , Mice, Inbred Strains , Reactive Oxygen Species/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Sex Factors
15.
Endocrinology ; 154(6): 2092-100, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584859

ABSTRACT

The expression of arginine-vasopressin (AVP) is regulated by estradiol and testosterone (T) in different neuronal populations by mechanisms that are not yet fully understood. Estrogen receptors (ERs) have been shown to participate in the regulation of AVP neurons by estradiol. In addition, there is evidence of the participation of ERß in the regulation of AVP expression exerted by T via its metabolite 5α-dihydrotestosterone (5α-DHT) and its further conversion in the androgen metabolite and ERß ligand 3ß-diol. In this study we have explored the role of ERs in the regulation exerted by estradiol and T on AVP expression, using the human neuroblastoma cell line SH-SY5Y. Estradiol treatment increased AVP mRNA levels in SH-SY5Y cells in comparison with cells treated with vehicle. The stimulatory effect of estradiol on AVP expression was imitated by the ERα agonist 4,4',4',-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol and blocked by the ER antagonist, ICI 182,780, and the ERα antagonist 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1hpyrazoledihydrochloride. In contrast, the ERß agonist 2,3-bis(4-hydroxyphenyl)-propionitrile reduced AVP expression, whereas the ERß antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl) pyrazolo[1,5-a]pyrimidin-3-yl]phenol enhanced the action of estradiol on AVP expression. T increased AVP expression in SH-SY5Y cells by a mechanism that was dependent on aromatase but not on 5α-reductase activity. The T effect was not affected by blocking the androgen receptor, was not imitated by the T metabolite 5α-DHT, and was blocked by the ERα antagonist 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1hpyrazoledihydrochloride. In contrast, 5α-DHT had a similar effect as the ERß agonists 2,3-bis(4-hydroxyphenyl)-propionitrile and 3ß-diol, reducing AVP expression. These findings suggest that estradiol and T regulate AVP expression in SH-SY5Y cells through ERs, exerting a stimulatory action via ERα and an inhibitory action via ERß.


Subject(s)
Arginine Vasopressin/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic/drug effects , Testosterone/pharmacology , Cell Line, Tumor , Estradiol/analogs & derivatives , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Female , Fulvestrant , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Nitriles/pharmacology , Phenols/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS One ; 8(1): e55237, 2013.
Article in English | MEDLINE | ID: mdl-23383123

ABSTRACT

Neurogenin 3 (Ngn3), a proneural gene, regulates dendritogenesis and synaptogenesis in mouse hippocampal neurons. Ngn3 is transiently exported from the cell nucleus to the cytoplasm when neuronal polarity is initiated, suggesting that the nucleo-cytoplasmic transport of the protein is important for its action on neuronal development. In this study, we identified for the first time a functional nuclear export sequence (NES2; ¹³¹YIWALTQTLRIA¹4²) in Ngn3. The green fluorescent protein (EGFP)-NES2 fusion protein was localized in the cytoplasm and its nucleo-cytoplasmic shuttling was blocked by the CRM1 specific export inhibitor leptomycin B. Mutation of a leucine residue to alanine (L135A) in the NES2 motif resulted in both cytoplasmic and nuclear localization of the EGFP-NES2 fusion protein and in the nuclear accumulation of ectopic full-length myc-Ngn3. In addition, point mutation of the leucine 135 counteracted the effects of Ngn3 on neuronal morphology and synaptic inputs indicating that the cytoplasmic localization of Ngn3 is important for neuronal development. Pharmacological perturbation of the cytoskeleton revealed that cytoplasmic Ngn3 is associated with microtubules.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Karyopherins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Export Signals/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Blotting, Western , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Immunohistochemistry , Immunoprecipitation , Mice , Microscopy, Fluorescence , Microtubules/metabolism , Mutagenesis , Oligonucleotides/genetics , Transfection , Exportin 1 Protein
17.
Horm Behav ; 63(2): 216-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22401743

ABSTRACT

Astrocytes and microglia respond to central nervous system (CNS) injury with changes in morphology, proliferation, migration and expression of inflammatory regulators. This phenomenon is known as reactive gliosis. Activation of astrocytes and microglia after acute neural insults, such as stroke or traumatic CNS injury, is considered to be an adaptive response that contributes to minimize neuronal damage. However, reactive gliosis may amplify CNS damage under chronic neurodegenerative conditions. Progesterone, estradiol and testosterone have been shown to control reactive gliosis in different models of CNS injury, modifying the number of reactive astrocytes and reactive microglia and the expression of anti-inflammatory and proinflammatory mediators. The actions of gonadal hormones on reactive gliosis involve different mechanisms, including the modulation of the activity of steroid receptors, such as estrogen receptors α and ß, the regulation of nuclear factor-κB mediated transcription of inflammatory molecules and the recruitment of the transcriptional corepressor c-terminal binding protein to proinflammatory promoters. In addition, the Parkinson's disease related gene parkin and the endocannabinoid system also participate in the regulation of reactive gliosis by estradiol. The control exerted by gonadal hormones on reactive gliosis may affect the response of neural tissue to trauma and neurodegeneration and may contribute to sex differences in the manifestation of neurodegenerative diseases. However, the precise functional consequences of the regulation of reactive gliosis by gonadal hormones under acute and chronic neurodegenerative conditions are still not fully clarified.


Subject(s)
Brain Injuries/complications , Gliosis/etiology , Gliosis/prevention & control , Gonadal Hormones/physiology , Animals , Brain Injuries/physiopathology , Gliosis/physiopathology , Gonadal Hormones/metabolism , Gonadal Hormones/pharmacology , Humans , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
18.
J Steroid Biochem Mol Biol ; 131(1-2): 52-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21971420

ABSTRACT

This review analyzes the signaling mechanisms activated by estradiol to regulate neuritogenesis in several neuronal populations. Estradiol regulates axogenesis by the activation of the mitogen activated protein kinase (MAPK) cascade through estrogen receptor α located in the plasma membrane. In addition, estradiol regulates MAPK signaling via the activation of protein kinase C and by increasing the expression of brain derived neurotrophic factor and tyrosine kinase receptor B. Estradiol also interacts with the signaling of insulin-like growth factor-I receptor through estrogen receptor α, modulating the phosphoinositide-3 kinase signaling pathway, which contributes to the stabilization of microtubules. Finally, estradiol modulates dendritogenesis by the inhibition of Notch signaling, by a mechanism that, at least in hippocampal neurons, is mediated by G-protein coupled receptor 30. This article is part of a Special Issue entitled 'Neurosteroids'.


Subject(s)
Estradiol/physiology , Neurites/physiology , Neurons/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Protein Kinase C/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Notch/physiology
19.
Biol Sex Differ ; 2: 7, 2011 Jul 11.
Article in English | MEDLINE | ID: mdl-21745355

ABSTRACT

BACKGROUND: Numerous neurological and psychiatric disorders show sex differences in incidence, age of onset, symptomatology or outcome. Astrocytes, one of the glial cell types of the brain, show sex differences in number, differentiation and function. Since astrocytes are involved in the response of neural tissue to injury and inflammation, these cells may participate in the generation of sex differences in the response of the brain to pathological insults. To explore this hypothesis, we have examined whether male and female astrocytes show a different response to an inflammatory challenge and whether perinatal testosterone influences this response. METHODS: Cortical astrocyte cultures were prepared from postnatal day 1 (one day after birth) male or female CD1 mice pups. In addition, cortical astrocyte cultures were also prepared from female pups that were injected at birth with 100 µg of testosterone propionate or vehicle. Cultures were treated for 5 hours with medium containing lipopolysaccharide (LPS) or with control medium. The mRNA levels of IL6, interferon-inducible protein 10 (IP10), TNFα, IL1ß, Toll-like receptor 4 (TLR4), steroidogenic acute regulatory protein and translocator protein were assessed by quantitative real-time polymerase chain reaction. Statistical significance was assessed by unpaired t-test or by one-way analysis of variance followed by the Tukey post hoc test. RESULTS: The mRNA levels of IL6, TNFα and IL1ß after LPS treatment were significantly higher in astrocytes derived from male or androgenized females compared to astrocytes derived from control or vehicle-injected females. In contrast, IP10 mRNA levels after LPS treatment were higher in astrocytes derived from control or vehicle-injected females than in those obtained from males or androgenized females. The different response of male and female astrocytes to LPS was due neither to differences in the basal expression of the inflammatory molecules nor to differences in the expression of the LPS receptor TLR4. In contrast, the different inflammatory response was associated with increased mRNA levels of translocator protein, a key steroidogenic regulator, in female astrocytes that were treated with LPS. CONCLUSIONS: Male and female cortical astrocytes respond differentially to an inflammatory challenge and this may be predetermined by perinatal testosterone exposure.

20.
Article in English | MEDLINE | ID: mdl-22654797

ABSTRACT

The transmembrane receptor Notch, a master developmental regulator, controls gliogenesis, neurogenesis, and neurite development in the nervous system. Estradiol, acting as a hormonal signal or as a neurosteroid, also regulates these developmental processes. Here we review recent evidence indicating that estradiol and Notch signaling interact in developing hippocampal neurons by a mechanism involving the putative membrane receptor G protein-coupled receptor 30. This interaction is relevant for the control of neuronal differentiation, since the downregulation of Notch signaling by estradiol results in the upregulation of neurogenin 3, which in turn promotes dendritogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL
...