Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters










Publication year range
1.
Front Neuroendocrinol ; 65: 100974, 2022 04.
Article in English | MEDLINE | ID: mdl-34995643

ABSTRACT

Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.


Subject(s)
Neuropeptides , Paraventricular Hypothalamic Nucleus , Estradiol/metabolism , Female , Humans , Male , Neurons/metabolism , Neuropeptides/metabolism , Neurosecretory Systems/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism
4.
Neuroscience ; 346: 203-215, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28131623

ABSTRACT

Genistein (GEN) is a natural xenoestrogen (isoflavonoid) that may interfere with the development of estrogen-sensitive neural circuits. Due to the large and increasing use of soy-based formulas for babies (characterized by a high content of GEN), there are some concerns that this could result in an impairment of some estrogen-sensitive neural circuits and behaviors. In a previous study, we demonstrated that its oral administration to female mice during late pregnancy and early lactation induced a significant decrease of nitric oxide synthase-positive cells in the amygdala of their male offspring. In the present study, we have used a different experimental protocol mimicking, in mice, the direct precocious exposure to GEN. Mice pups of both sexes were fed either with oil, estradiol or GEN from birth to postnatal day 8. Nitric oxide synthase and vasopressin neural systems were analyzed in adult mice. Interestingly, we observed that GEN effect was time specific (when compared to our previous study), sex specific, and not always comparable to the effects of estradiol. This last observation suggests that GEN may act through different intracellular pathways. Present results indicate that the effect of natural xenoestrogens on the development of the brain may be highly variable: a plethora of neuronal circuits may be affected depending on sex, time of exposure, intracellular pathway involved, and target cells. This raises concern on the possible long-term effects of the use of soy-based formulas for babies, which may be currently underestimated.


Subject(s)
Brain/drug effects , Brain/metabolism , Genistein/administration & dosage , Nitric Oxide Synthase Type I/metabolism , Phytoestrogens/administration & dosage , Sex Characteristics , Vasopressins/metabolism , Animals , Estradiol/administration & dosage , Female , Male , Mice
6.
J Neuroendocrinol ; 28(2): 12351, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26681259

ABSTRACT

Neuroactive steroids are endogenous neuromodulators synthesised in the brain that rapidly alter neuronal excitability by binding to membrane receptors, in addition to the regulation of gene expression via intracellular steroid receptors. Neuroactive steroids induce potent anxiolytic, antidepressant, anticonvulsant, sedative, analgesic and amnesic effects, mainly through interaction with the GABAA receptor. They also exert neuroprotective, neurotrophic and antiapoptotic effects in several animal models of neurodegenerative diseases. Neuroactive steroids regulate many physiological functions, such as the stress response, puberty, the ovarian cycle, pregnancy and reward. Their levels are altered in several neuropsychiatric and neurological diseases and both preclinical and clinical studies emphasise a therapeutic potential of neuroactive steroids for these diseases, whereby symptomatology ameliorates upon restoration of neuroactive steroid concentrations. However, direct administration of neuroactive steroids has several challenges, including pharmacokinetics, low bioavailability, addiction potential, safety and tolerability, which limit its therapeutic use. Therefore, modulation of neurosteroidogenesis to restore the altered endogenous neuroactive steroid tone may represent a better therapeutic approach. This review summarises recent approaches that target the neuroactive steroid biosynthetic pathway at different levels aiming to promote neurosteroidogenesis. These include modulation of neurosteroidogenesis through ligands of the translocator protein 18 kDa and the pregnane xenobiotic receptor, as well as targeting of specific neurosteroidogenic enzymes such as 17ß-hydroxysteroid dehydrogenase type 10 or P450 side chain cleavage. Enhanced neurosteroidogenesis through these targets may be beneficial not only for neurodegenerative diseases, such as Alzheimer's disease and age-related dementia, but also for neuropsychiatric diseases, including alcohol use disorders.


Subject(s)
Neurotransmitter Agents/biosynthesis , Neurotransmitter Agents/therapeutic use , Translational Research, Biomedical , 17-Hydroxysteroid Dehydrogenases/metabolism , Alcoholism/drug therapy , Alzheimer Disease/drug therapy , Humans , Pregnane X Receptor , Receptors, GABA/metabolism , Receptors, Steroid/metabolism
7.
Neuroscience ; 286: 162-70, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25481234

ABSTRACT

Steroid hormones are important players to regulate adult neurogenesis in the dentate gyrus of the hippocampus, but their involvement in the regulation of the same phenomenon in the subventricular zone (SVZ) of the lateral ventricles is not completely understood. Here, in male rats, we tested the existence of activational effects of testosterone (T) on cell proliferation in the adult SVZ. To this aim, three groups of male rats: castrated, castrated and treated with T, and controls were treated with 5-bromo-2'-deoxyuridine (BrdU) and killed after 24h. The density of BrdU-labeled cells was significantly lower in castrated animals in comparison to the other two groups, thus supporting a direct correlation between SVZ proliferation and levels of circulating T. To clarify whether this effect is purely androgen-dependent, or mediated by the T metabolites, estradiol (E2) and dihydrotestosterone (DHT), we evaluated SVZ proliferation in castrated males treated with E2, DHT and E2+DHT, in comparison to T- and vehicle-treated animals, and sham-operated controls. The stereological analysis demonstrated that E2 and T, but not DHT, increase proliferation in the SVZ of adult male rats. Quantitative evaluation of cells expressing the endogenous marker of cell proliferation phosphorylated form of Histone H3 (PHH3), or the marker of highly dividing SVZ progenitors Mash1, indicated the effect of T/E2 is mostly restricted to SVZ proliferating progenitors. The same experimental protocol was repeated on ovariectomized female rats treated with E2 or T. In this case, no statistically significant difference was found among groups. Overall, our results clearly show that the gonadal hormones T and E2 represent important mediators of cell proliferation in the adult SVZ. Moreover, we show that such an effect is restricted to males, supporting adult neurogenesis in rats is a process differentially modulated in the two sexes.


Subject(s)
Cell Proliferation , Estradiol/physiology , Lateral Ventricles/physiology , Neurogenesis , Testosterone/physiology , Animals , Bromodeoxyuridine/analysis , Castration , Estradiol/pharmacology , Female , Lateral Ventricles/drug effects , Male , Rats , Rats, Wistar , Testosterone/metabolism , Testosterone/pharmacology
8.
Brain Res ; 1588: 55-62, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25218558

ABSTRACT

Arginine-Vasopressin (AVP) may regulate the hypothalamic-pituitary-adrenal axis (HPA) and its effects on depressive responses. In a recent study, we demonstrated that Chronic Unpredictable Stress (CUS) depressive effects are enhanced by long-term ovariectomy (a model of post-menopause). In the present study, we investigated the effects of long-term ovariectomy and CUS on AVP expression in different subdivision of the paraventricular nucleus (PVN) of female mice. Both long-term ovariectomy and CUS affect AVP immunoreactivity in some of the PVN subnuclei of adult female mice. In particular, significant changes on AVP immunoreactivity were observed in magnocellular subdivisions, the paraventricular lateral magnocellular (PaLM) and the paraventricular medial magnocellular (PaMM), the 2 subnuclei projecting to the neurohypophysis for the hormonal regulation of body homeostasis. AVP immunoreactivity was decreased in the PaLM by both the long-term deprivation of ovarian hormones and the CUS. In contrast, AVP immunoreactivity was increased in the PaMM by CUS, whereas it was decreased by ovariectomy. Therefore, present results suggest morphological and functional differences among the PVN's subnuclei and complex interactions among CUS, gonadal hormones and AVP immunoreactivity.


Subject(s)
Arginine Vasopressin/metabolism , Gonadal Hormones/deficiency , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Stress, Psychological/metabolism , Stress, Psychological/pathology , Animals , Chronic Disease , Corticosterone/blood , Female , Immunohistochemistry , Mice, Inbred C57BL , Ovariectomy , Photomicrography , Uncertainty
9.
Neuroscience ; 253: 67-77, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-23999123

ABSTRACT

In rodents as well as in many other mammalian and non-mammalian species, the arginine-vasopressin (AVP) system includes a parvocellular sexually dimorphic portion located within the bed nucleus of the stria terminalis (BST), the medial amygdaloid nucleus (MeA) and the lateral septum. In this system, males have more cells and denser projections than females, neurons show androgen and estrogen receptors, and gonadal hormones are required for the activation. However, the role of these hormones for the differentiation of the system is not clear. Previous studies performed on aromatase knockout mice suggested that estradiol is not necessary for the differentiation of the system, but it is important for its activation in adulthood. To elucidate the role of androgens on differentiation and functioning of AVP parvocellular system, we compared male and female rats with a non-functional mutation of androgen receptor (Tfm, testicular feminization mutation) to their control littermates. Our data show that the lack of a functional androgen receptor significantly decreases the expression of AVP immunoreactivity within the BST and MeA of male Tfm. Thus supporting the hypothesis that androgens, through the action of their receptor, should have a relevant role in the organization and modulation of the AVP parvocellular sexually dimorphic system.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Arginine Vasopressin/metabolism , Brain/metabolism , Receptors, Androgen/deficiency , Sex Characteristics , Analysis of Variance , Androgen-Insensitivity Syndrome/pathology , Animals , Animals, Newborn , Brain/pathology , Disease Models, Animal , Female , Male , Mutation/genetics , Neurons/pathology , Rats
10.
Neuroscience ; 248: 127-35, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23707802

ABSTRACT

Estrogen receptors (ERs) α and ß are involved in the regulation of the nitrergic system in the supraoptic (SON) and paraventricular (PVN) nuclei under basal conditions. In this study we have assessed whether ERs are also involved in the modulation of the nitrergic system in the SON and PVN under acute systemic hypertonic conditions. Adult ovariectomized rats received a single injection of either estradiol, a selective ERα agonist, a selective ERß agonist, a selective ERα antagonist, a selective ERß antagonist or vehicle. Twenty-four hours later, animals received one i.p. injection of 1.5M NaCl to induce osmotic stress and were sacrificed after two additional hours. The number of NADPH-diaphorase-positive cells in the SON and PVN was determined. Their number in the SON was not affected by NaCl administration, whereas in the four PVN subdivisions it was decreased after NaCl administration. Estradiol and the ERα agonist prevented the action of NaCl in the four subdivisions of the PVN. In contrast, the inhibition of ERα enhanced the effect of NaCl, inducing a further decrease in the number of NADPH-diaphorase-positive cells. Moreover, the ERß agonist enhanced and the ERß antagonist blocked the effect of NaCl on the number of NADPH-diaphorase-positive neurons in the SON and in the medial magnocellular subdivision of the PVN. These findings indicate that estradiol regulates the nitrergic system in the SON and PVN under acute osmotic stress conditions, but the effects specifically depend on the anatomical subregions and different ERs.


Subject(s)
Estradiol/physiology , NADPH Dehydrogenase/metabolism , Neurons/enzymology , Osmotic Pressure , Paraventricular Hypothalamic Nucleus/enzymology , Stress, Physiological , Supraoptic Nucleus/enzymology , Animals , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Female , Ovariectomy , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Wistar , Supraoptic Nucleus/drug effects
12.
Behav Brain Res ; 229(2): 333-9, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22289198

ABSTRACT

The impact of stress is widely recognized in the etiology of multiple disorders. In particular, psychological stress may increase the risk of cardiovascular, metabolic, immune, and mood disorders. Several genes are considered potential candidates to account for the deleterious consequences of stress and recent data point to role of Vgf. VGF mRNA is abundantly expressed in the hypothalamus, where it has been involved in metabolism and energy homeostasis; more recently a link between VGF-derived peptides and mood disorders has been highlighted. The following experiments were performed to address the contribution of the VGF-system to stress induced changes in mice: the distribution of VGF immuno-reactivity in hypothalamic nuclei and its modulation by social stress; the role of VGF-derived peptide TLQP-21 in plasma catecholamine release induced by acute restraint stress (RS); the efficacy of chronic TLQP-21 in a mouse model of chronic subordination stress (CSS). VGF fibers were found in high density in arcuate, dorsomedial, and suprachiasmatic and, at lower density, in lateral, paraventricular, and ventromedial hypothalamic nuclei. Central administration of either 2 or 4 mM TLQP-21 acutely altered the biphasic serum epinephrine release and decreased norepinephrine serum levels in response to RS. Finally, 28-day of 40 µg/day TLQP-21 treatment increased CSS-induced social avoidance of an unfamiliar conspecific. Overall these data support a role for TLQP-21 in stress responses providing a promising starting point to further elucidate its role as a player in stress-related human pathologies.


Subject(s)
Hypothalamus/metabolism , Neuropeptides/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Animals , Avoidance Learning/drug effects , Catecholamines/blood , Disease Models, Animal , Hypothalamus/drug effects , Infusions, Subcutaneous , Injections, Intraventricular , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Nerve Growth Factors , Peptide Fragments/administration & dosage , Social Behavior , Stress, Psychological/blood
13.
J Neuroendocrinol ; 24(1): 144-59, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21951193

ABSTRACT

Some environmental contaminants interact with hormones and may exert adverse consequences as a result of their actions as endocrine disrupting chemicals (EDCs). Exposure in people is typically a result of contamination of the food chain, inhalation of contaminated house dust or occupational exposure. EDCs include pesticides and herbicides (such as dichlorodiphenyl trichloroethane or its metabolites), methoxychlor, biocides, heat stabilisers and chemical catalysts (such as tributyltin), plastic contaminants (e.g. bisphenol A), pharmaceuticals (i.e. diethylstilbestrol; 17α-ethinylestradiol) or dietary components (such as phytoestrogens). The goal of this review is to address the sources, effects and actions of EDCs, with an emphasis on topics discussed at the International Congress on Steroids and the Nervous System. EDCs may alter reproductively-relevant or nonreproductive, sexually-dimorphic behaviours. In addition, EDCs may have significant effects on neurodevelopmental processes, influencing the morphology of sexually-dimorphic cerebral circuits. Exposure to EDCs is more dangerous if it occurs during specific 'critical periods' of life, such as intrauterine, perinatal, juvenile or puberty periods, when organisms are more sensitive to hormonal disruption, compared to other periods. However, exposure to EDCs in adulthood can also alter physiology. Several EDCs are xenoestrogens, which can alter serum lipid concentrations or metabolism enzymes that are necessary for converting cholesterol to steroid hormones. This can ultimately alter the production of oestradiol and/or other steroids. Finally, many EDCs may have actions via (or independent of) classic actions at cognate steroid receptors. EDCs may have effects through numerous other substrates, such as the aryl hydrocarbon receptor, the peroxisome proliferator-activated receptor and the retinoid X receptor, signal transduction pathways, calcium influx and/or neurotransmitter receptors. Thus, EDCs, from varied sources, may have organisational effects during development and/or activational effects in adulthood that influence sexually-dimorphic, reproductively-relevant processes or other functions, by mimicking, antagonising or altering steroidal actions.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Endocrine Disruptors/pharmacology , Reproduction/drug effects , Animals , Benzhydryl Compounds , Environmental Pollutants/pharmacology , Humans , Phenols/pharmacology , Phytoestrogens/pharmacology , Puberty/drug effects
14.
J Neuroendocrinol ; 24(1): 1-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22188420

ABSTRACT

During the last 10 years, the conference on 'Steroids and Nervous System' held in Torino (Italy) has been an important international point of discussion for scientists involved in this exciting and expanding research field. The present review aims to recapitulate the main topics that have been presented through the various meetings. Two broad areas have been explored: the impact of gonadal hormones on brain circuits and behaviour, as well as the mechanism of action of neuroactive steroids. Relationships among steroids, brain and behaviour, the sexual differentiation of the brain and the impact of gonadal hormones, the interactions of exogenous steroidal molecules (endocrine disrupters) with neural circuits and behaviour, and how gonadal steroids modulate the behaviour of gonadotrophin-releasing hormone neurones, have been the topics of several lectures and symposia during this series of meetings. At the same time, many contributions have been dedicated to the biosynthetic pathways, the physiopathological relevance of neurosteroids, the demonstration of the cellular localisation of different enzymes involved in neurosteroidogenesis, the mechanisms by which steroids may exert some of their effects, both the classical and nonclassical actions of different steroids, the role of neuroactive steroids on neurodegeneration, neuroprotection, and the response of the neural tissue to injury. In these 10 years, this field has significantly advanced and neuroactive steroids have emerged as new potential therapeutic tools to counteract neurodegenerative events.


Subject(s)
Brain/physiology , Congresses as Topic/history , Neurobiology/history , Neurotransmitter Agents/physiology , Animals , History, 21st Century , Humans , Neurology/history , Research , Steroids/biosynthesis , Translational Research, Biomedical
15.
J Toxicol Environ Health B Crit Rev ; 14(5-7): 449-72, 2011.
Article in English | MEDLINE | ID: mdl-21790321

ABSTRACT

Endocrine-disrupting chemicals (EDC) are molecules that interfere with endocrine signaling pathways and produce adverse consequences on animal and human physiology, such as infertility or behavioral alterations. Some EDC act through binding to androgen or/and estrogen receptors primarily operating through a genomic mechanism regulating gene expression. This mechanism of action may induce profound developmental adverse effects, and the major targets of the EDC action are the gene products, i.e., mRNAs inducing the synthesis of various peptidic molecules, which include neuropeptides and enzymes related to neurotransmitters syntheses. Available immunohistochemical data on some of the systems that are affected by EDC in lower and higher vertebrates are detailed in this review.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Animals , Enzymes/drug effects , Enzymes/metabolism , Gene Expression Regulation/drug effects , Humans , Neuropeptides/drug effects , Neuropeptides/metabolism , Protein Binding , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism
16.
Peptides ; 32(6): 1330-4, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21514339

ABSTRACT

In the present study we used a transgenic mouse model, carrying the neuropeptide Y (NPY) Y1 receptor gene promoter linked to the LacZ reporter gene (Y1R/LacZ mice) to test the hypothesis of its up-regulation by gonadal hormones. Y1 receptor gene expression was detected by means of histochemical procedures and quantitative image analysis in the paraventricular nucleus, arcuate nucleus, medial preoptic nucleus, ventromedial nucleus and bed nucleus of stria terminalis of two-month-old female mice at different stages of estrous cycle. Qualitative and quantitative analyses showed that Y1R/LacZ transgene expression was higher in the paraventricular, arcuate, and ventromedial nuclei of proestrus mice as compared to mice in the other stages of the estrous cycle. In addition, we performed a comparison with a group of sexually active males. In this comparison a significant difference (less in males) was observed between males and proestrus females in the same nuclei. In conclusion, these data indicate that fluctuations in circulating levels of gonadal hormones, depending by estrous cycle, are paralleled by changes in the expression of NPY Y1 receptor in the hypothalamic nuclei involved in the control of both energy balance and reproduction.


Subject(s)
Energy Metabolism/physiology , Estrous Cycle/physiology , Hypothalamus , Receptors, Neuropeptide Y , Reproduction/physiology , Animals , Female , Gene Expression , Genes, Reporter , Histocytochemistry , Hypothalamus/cytology , Hypothalamus/physiology , Lac Operon , Male , Mice , Mice, Transgenic , Molecular Imaging , Neuropeptide Y/metabolism , Organ Specificity , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Sex Factors , Sexual Behavior, Animal , Transgenes
17.
Cell Death Dis ; 2: e130, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21412276

ABSTRACT

Signalling by receptor tyrosine kinases (RTKs) coordinates basic cellular processes during development and in adulthood. Whereas aberrant RTK signalling can lead to cancer, reactivation of RTKs is often found following stress or cell damage. This has led to the common belief that RTKs can counteract degenerative processes and so strategies to exploit them for therapy have been extensively explored. An understanding of how RTK stimuli act at cellular levels is needed, however, to evaluate their mechanism of therapeutic action. In this study, we genetically explored the biological and functional significance of enhanced signalling by the Met RTK in neurons, in the context of a neurodegenerative disease. Conditional met-transgenic mice, namely Rosa26(LacZ-stop-Met), have been engineered to trigger increased Met signalling in a temporal and tissue-specific regulated manner. Enhancing Met levels in neurons does not affect either motor neuron (MN) development or maintenance. In contrast, increased neuronal Met in amyotrophic lateral sclerosis (ALS) mice prolongs life span, retards MN loss, and ameliorates motor performance, by selectively delaying disease onset. Thus, our studies highlight the properties of RTKs to counteract toxic signals in a disease characterized by dysfunction of multiple cell types by acting in MNs. Moreover, they emphasize the relevance of genetically assessing the effectiveness of agents targeting neurons during ALS evolution.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/pathology , Proto-Oncogene Proteins c-met/genetics
18.
Neurotoxicology ; 32(2): 277-80, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21185327

ABSTRACT

Tributyltin (TBT) is a largely diffused environmental pollutant, banned from paints in the European Union from 2003. However, the level of TBT (and other organotins) in food, particularly fish and shellfish, remains still high. Several studies demonstrated that TBT is involved in the development of obesity, via peripheral action, but currently, there are only a few data illustrating effects of TBT on the nervous system. In the present study, we tested the hypothesis that acute exposure to TBT may directly activate brain cells in particular, in those hypothalamic nuclei regulating the food intake. To this purpose, TBT was orally administered at a single dose (10 mg/kg/body weight) to two groups of adult male mice: regularly fed or fasted for 24 h. Mice were sacrificed 90 min after the TBT administration and perfused by 4% paraformaldehyde. Brains were quickly dissected, frozen and sectioned for immunocytochemical detection of c-fos, a common marker of cell activation. In both, fed or fasted mice, exposure to TBT induced a significant increase of c-fos expression in the arcuate nucleus in comparison to control mice. The other nuclei involved in the control of feeding behavior did not show any significant increase. These data are the first in vivo demonstration that TBT has not only peripheral effects, but also may activate elements in the brain, in particular in a crucial region for the regulation of food intake like the arcuate nucleus.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Trialkyltin Compounds/administration & dosage , Animals , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Trialkyltin Compounds/toxicity
19.
Ann N Y Acad Sci ; 1163: 271-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19456348

ABSTRACT

Several environmental chemicals have the capability of impacting endocrine function (endocrine disrupting chemicals [EDCs]), and therefore they may have long-term consequences, especially if exposure occurs during embryonic development. In this study we present data relative to two widely used animal models: the Japanese quail and the mouse. These two species have been used to understand neural, neuroendocrine, and behavioral components of reproduction and are optimal models to understand how these components are altered by precocious exposure to EDCs. In particular, we discuss the effects of embryonic exposure to diethylstilbestrol, genistein, or ethylene,1,1-dichloro-2,2-bis(p-chlorophenyl) on the sexually dimorphic parvocellular vasotocin system and male copulatory behavior in quail and the effects of bisphenol A on the nitrinergic and kisspeptin systems and their behavioral impact in the mouse. In both models the exposure to EDCs during the critical period (early embryonic period in birds, perinatal period in rodents) alters the differentiation of relevant sexually dimorphic pathways, often inducing the appearance of a sex-reversed neurochemical phenotype that is the most probable cause of the final alteration of sexually differentiated behaviors in the adult animal. In conclusion, the data presented here should stimulate a critical reanalysis of the way to determine the "safe" exposure levels to EDCs for wild species and humans, considering behavior and related neural circuits among the factors to be analyzed.


Subject(s)
Behavior, Animal/physiology , Estrogens/metabolism , Neurons/metabolism , Vertebrates/physiology , Animals , Behavior, Animal/drug effects , Benzhydryl Compounds , Endocrine Disruptors/pharmacology , Humans , Neurons/drug effects , Phenols/pharmacology
20.
Eur J Neurosci ; 29(4): 679-92, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19200078

ABSTRACT

Newborn neurons generated by proliferative progenitors in the adult subventricular zone (SVZ) integrate into the olfactory bulb circuitry of mammals. Survival of these newly-formed cells is regulated by the olfactory input. The presence of new neurons in the accessory olfactory bulb (AOB) has already been demonstrated in some mammalian species, albeit their neurochemical profile and functional integration into AOB circuits are still to be investigated. To unravel whether the mouse AOB represents a site of adult constitutive neurogenesis and whether this process can be modulated by extrinsic factors, we have used multiple in vivo approaches. These included fate mapping of bromodeoxyuridine-labelled cells, lineage tracing of SVZ-derived enhanced green fluorescent protein-positive engrafted cells and neurogenesis quantification in the AOB, in both sexes, as well as in females alone after exposure to male-soiled bedding or its derived volatiles. Here, we show that a subpopulation of SVZ-derived neuroblasts acquires proper neurochemical profiles of mature AOB interneurons. Moreover, 3D reconstruction of long-term survived engrafted neuroblasts in the AOB confirms these cells show features of fully integrated neurons. Finally, exposure to male-soiled bedding, but not to its volatile compounds, significantly increases the number of new neurons in the AOB, but not in the main olfactory bulb of female mice. These data show SVZ-derived neuroblasts differentiate into new functionally integrated neurons in the AOB of young and adult mice. Survival of these cells seems to be regulated by an experience-specific mechanism mediated by pheromones.


Subject(s)
Neurons/physiology , Olfactory Bulb/physiology , Smell/physiology , Animals , Bromodeoxyuridine , Cell Survival , Cerebral Ventricles/physiology , Female , Imaging, Three-Dimensional , Male , Mice , Neurogenesis , Neurons/cytology , Olfactory Bulb/anatomy & histology , Organ Size , Pheromones , Physical Stimulation , Prosencephalon/cytology , Prosencephalon/physiology , Proto-Oncogene Proteins c-fos/metabolism , Sex Characteristics , Time
SELECTION OF CITATIONS
SEARCH DETAIL
...