Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.565
1.
CNS Neurosci Ther ; 30(5): e14726, 2024 05.
Article En | MEDLINE | ID: mdl-38715251

AIMS: The preoptic area (POA) of the hypothalamus, crucial in thermoregulation, has long been implicated in the pain process. However, whether nociceptive stimulation affects body temperature and its mechanism remains poorly studied. METHODS: We used capsaicin, formalin, and surgery to induce acute nociceptive stimulation and monitored rectal temperature. Optical fiber recording, chemical genetics, confocal imaging, and pharmacology assays were employed to confirm the role and interaction of POA astrocytes and extracellular adenosine. Immunofluorescence was utilized for further validation. RESULTS: Acute nociception could activate POA astrocytes and induce a decrease in body temperature. Manipulation of astrocytes allowed bidirectional control of body temperature. Furthermore, acute nociception and astrocyte activation led to increased extracellular adenosine concentration within the POA. Activation of adenosine A1 or A2A receptors contributed to decreased body temperature, while inhibition of these receptors mitigated the thermo-lowering effect of astrocytes. CONCLUSION: Our results elucidate the interplay between acute nociception and thermoregulation, specifically highlighting POA astrocyte activation. This enriches our understanding of physiological responses to painful stimuli and contributes to the analysis of the anatomical basis involved in the process.


Astrocytes , Hypothermia , Nociception , Preoptic Area , Animals , Preoptic Area/drug effects , Preoptic Area/metabolism , Astrocytes/metabolism , Astrocytes/drug effects , Nociception/physiology , Hypothermia/chemically induced , Male , Mice , Receptors, Purinergic P1/metabolism , Mice, Inbred C57BL , Adenosine/metabolism , Capsaicin/pharmacology , Formaldehyde/toxicity , Formaldehyde/pharmacology
2.
Nature ; 606(7916): 937-944, 2022 06.
Article En | MEDLINE | ID: mdl-35676482

During infection, animals exhibit adaptive changes in physiology and behaviour aimed at increasing survival. Although many causes of infection exist, they trigger similar stereotyped symptoms such as fever, warmth-seeking, loss of appetite and fatigue1,2. Yet exactly how the nervous system alters body temperature and triggers sickness behaviours to coordinate responses to infection remains unknown. Here we identify a previously uncharacterized population of neurons in the ventral medial preoptic area (VMPO) of the hypothalamus that are activated after sickness induced by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid. These neurons are crucial for generating a fever response and other sickness symptoms such as warmth-seeking and loss of appetite. Single-nucleus RNA-sequencing and multiplexed error-robust fluorescence in situ hybridization uncovered the identity and distribution of LPS-activated VMPO (VMPOLPS) neurons and non-neuronal cells. Gene expression and electrophysiological measurements implicate a paracrine mechanism in which the release of immune signals by non-neuronal cells during infection activates nearby VMPOLPS neurons. Finally, we show that VMPOLPS neurons exert a broad influence on the activity of brain areas associated with behavioural and homeostatic functions and are synaptically and functionally connected to circuit nodes controlling body temperature and appetite. Together, these results uncover VMPOLPS neurons as a control hub that integrates immune signals to orchestrate multiple sickness symptoms in response to infection.


Appetite , Fever , Infections , Neurons , Preoptic Area , Animals , Appetite/drug effects , Appetite Depressants/pharmacology , Fever/chemically induced , Fever/physiopathology , In Situ Hybridization, Fluorescence , Infections/chemically induced , Infections/physiopathology , Lipopolysaccharides , Neurons/drug effects , Paracrine Communication , Poly I-C , Preoptic Area/cytology , Preoptic Area/drug effects , Preoptic Area/physiology
3.
Neurosci Lett ; 773: 136518, 2022 03 16.
Article En | MEDLINE | ID: mdl-35150776

In normal hormonal conditions, increased neuronal activity in the ventromedial hypothalamus (VMH) induces lordosis whereas activation of the preoptic area (POA) exerts an opposite effect. In the present work, we explored the effect of bilateral infusion of different doses of the apelin-13 (0.37, 0.75, 1.5, and 15 µg) in both brain areas on the expression of lordosis behavior. Lordosis quotient and lordosis reflex score were performed at 30, 120, and 240 min. Weak lordosis was observed following the 0.37 µg dose of apelin-13 at 30 min in the VMH of EB-primed rats; however, the rest of the doses induced significant lordosis relative to the control group. At 120 min, all doses induced lordosis behavior, while at 240 min, the highest dose of 15 µg did not induce significant differences. Interestingly, only the 0.75 µg infusion of apelin in the POA induced significant lordosis at 120 and 240 min. These results indicate that apelin-13 acts preferably in HVM and slightly in POA to initiate lordosis behavior in estrogen-primed rats.


Intercellular Signaling Peptides and Proteins , Lordosis , Preoptic Area , Animals , Estradiol/pharmacology , Estrogens/pharmacology , Hypothalamus/drug effects , Hypothalamus/pathology , Intercellular Signaling Peptides and Proteins/pharmacology , Lordosis/chemically induced , Preoptic Area/drug effects , Preoptic Area/pathology , Progesterone/pharmacology , Rats , Sexual Behavior, Animal/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology
4.
Biomed Pharmacother ; 146: 112301, 2022 Feb.
Article En | MEDLINE | ID: mdl-34915415

Treatment of sleep disorders promotes the long-term use of commercially available sleep inducers that have several adverse effects, including addiction, systemic fatigue, weakness, loss of concentration, headache, and digestive problems. Therefore, we aimed to limit these adverse effects by investigating a natural product, the extract of the Hibiscus syriacus Linnaeus flower (HSF), as an alternative treatment. In the electric footshock model, we measured anxiety and assessed the degree of sleep improvement after administering HSF extract. In the restraint model, we studied the sleep rate using PiezoSleep, a noninvasive assessment system. In the pentobarbital model, we measured sleep improvement and changes in sleep-related factors. Our first model confirmed the desirable effects of HSF extract and its active constituent, saponarin, on anxiolysis and Wake times. HSF extract also increased REM sleep time. Furthermore, HSF extract and saponarin increased the expression of cortical GABAA receptor α1 (GABAAR α1) and c-Fos in the ventrolateral preoptic nucleus (VLPO). In the second model, HSF extract and saponarin restored the sleep rate and the sleep bout duration. In the third model, HSF extract and saponarin increased sleep maintenance time. Moreover, HSF extract and saponarin increased cortical cholecystokinin (CCK) mRNA levels and the expression of VLPO c-Fos. HSF extract also increased GABAAR α1 mRNA level. Our results suggest that HSF extract and saponarin are effective in maintaining sleep and may be used as a novel treatment for sleep disorder. Eventually, we hope to introduce HSF and saponarin as a clinical treatment for sleep disorders in humans.


Apigenin/therapeutic use , Glucosides/therapeutic use , Hibiscus , Plant Extracts/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep/drug effects , Animals , Apigenin/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Corticosterone/blood , Disease Models, Animal , Electroencephalography , Glucosides/pharmacology , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Pentobarbital , Plant Extracts/pharmacology , Preoptic Area/drug effects , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Sleep Aids, Pharmaceutical , Sleep Wake Disorders/blood , Sleep Wake Disorders/genetics , Sleep Wake Disorders/physiopathology , Stress, Psychological/blood , Stress, Psychological/complications , Stress, Psychological/genetics , Stress, Psychological/physiopathology
5.
Int J Mol Sci ; 22(8)2021 Apr 09.
Article En | MEDLINE | ID: mdl-33918982

Lithium (Li+) salt is widely used as a therapeutic agent for treating neurological and psychiatric disorders. Despite its therapeutic effects on neurological and psychiatric disorders, it can also disturb the neuroendocrine axis in patients under lithium therapy. The hypothalamic area contains GABAergic and glutamatergic neurons and their receptors, which regulate various hypothalamic functions such as the release of neurohormones, control circadian activities. At the neuronal level, several neurotransmitter systems are modulated by lithium exposure. However, the effect of Li+ on hypothalamic neuron excitability and the precise action mechanism involved in such an effect have not been fully understood yet. Therefore, Li+ action on hypothalamic neurons was investigated using a whole-cell patch-clamp technique. In hypothalamic neurons, Li+ increased the GABAergic synaptic activities via action potential independent presynaptic mechanisms. Next, concentration-dependent replacement of Na+ by Li+ in artificial cerebrospinal fluid increased frequencies of GABAergic miniature inhibitory postsynaptic currents without altering their amplitudes. Li+ perfusion induced inward currents in the majority of hypothalamic neurons independent of amino-acids receptor activation. These results suggests that Li+ treatment can directly affect the hypothalamic region of the brain and regulate the release of various neurohormones involved in synchronizing the neuroendocrine axis.


GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Lithium/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synapses/drug effects , Synapses/metabolism , Animals , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Inhibitory Postsynaptic Potentials/drug effects , Patch-Clamp Techniques , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, Amino Acid/metabolism , Synaptic Transmission/drug effects
6.
Sci Rep ; 11(1): 3148, 2021 02 04.
Article En | MEDLINE | ID: mdl-33542258

Delayed emergence from anesthesia was previously reported in a case study of a child with Glycine Encephalopathy. To investigate the neural basis of this delayed emergence, we developed a zebrafish glial glycine transporter (glyt1 - / -) mutant model. We compared locomotor behaviors; dose-response curves for tricaine, ketamine, and 2,6-diisopropylphenol (propofol); time to emergence from these anesthetics; and time to emergence from propofol after craniotomy in glyt1-/- mutants and their siblings. To identify differentially active brain regions in glyt1-/- mutants, we used pERK immunohistochemistry as a proxy for brain-wide neuronal activity. We show that glyt1-/- mutants initiated normal bouts of movement less frequently indicating lethargy-like behaviors. Despite similar anesthesia dose-response curves, glyt1-/- mutants took over twice as long as their siblings to emerge from ketamine or propofol, mimicking findings from the human case study. Reducing glycine levels rescued timely emergence in glyt1-/- mutants, pointing to a causal role for elevated glycine. Brain-wide pERK staining showed elevated activity in hypnotic brain regions in glyt1-/- mutants under baseline conditions and a delay in sensorimotor integration during emergence from anesthesia. Our study links elevated activity in preoptic brain regions and reduced sensorimotor integration to lethargy-like behaviors and delayed emergence from propofol in glyt1-/- mutants.


Delayed Emergence from Anesthesia/genetics , Glycine Plasma Membrane Transport Proteins/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/genetics , Neurons/metabolism , Preoptic Area/metabolism , Zebrafish Proteins/genetics , Aminobenzoates , Anesthesia, General , Anesthetics , Animals , Animals, Genetically Modified , Craniotomy , Delayed Emergence from Anesthesia/metabolism , Delayed Emergence from Anesthesia/physiopathology , Delayed Emergence from Anesthesia/prevention & control , Disease Models, Animal , Gene Expression , Glycine/pharmacology , Glycine Plasma Membrane Transport Proteins/deficiency , Hyperglycinemia, Nonketotic/drug therapy , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/physiopathology , Ketamine , Locomotion/physiology , Neurons/drug effects , Neurons/pathology , Preoptic Area/drug effects , Preoptic Area/pathology , Propofol , Zebrafish , Zebrafish Proteins/deficiency , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
7.
EMBO J ; 40(5): e104267, 2021 03 01.
Article En | MEDLINE | ID: mdl-33491217

Impairments in social relationships and awareness are features observed in autism spectrum disorders (ASDs). However, the underlying mechanisms remain poorly understood. Shank2 is a high-confidence ASD candidate gene and localizes primarily to postsynaptic densities (PSDs) of excitatory synapses in the central nervous system (CNS). We show here that loss of Shank2 in mice leads to a lack of social attachment and bonding behavior towards pubs independent of hormonal, cognitive, or sensitive deficits. Shank2-/- mice display functional changes in nuclei of the social attachment circuit that were most prominent in the medial preoptic area (MPOA) of the hypothalamus. Selective enhancement of MPOA activity by DREADD technology re-established social bonding behavior in Shank2-/- mice, providing evidence that the identified circuit might be crucial for explaining how social deficits in ASD can arise.


Autistic Disorder/drug therapy , Disease Models, Animal , Interpersonal Relations , Maternal Behavior/drug effects , Nerve Tissue Proteins/physiology , Piperazines/pharmacology , Preoptic Area/drug effects , Animals , Autistic Disorder/etiology , Autistic Disorder/metabolism , Autistic Disorder/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Preoptic Area/metabolism , Preoptic Area/pathology , Synapses
8.
J Neuroendocrinol ; 33(1): e12918, 2021 01.
Article En | MEDLINE | ID: mdl-33340384

Gonadal hormones affect neuronal morphology to ultimately regulate behaviour. In female rats, oestradiol mediates spine plasticity in hypothalamic and limbic brain structures, contributing to long-lasting effects on motivated behaviour. Parallel effects of androgens in male rats have not been extensively studied. Here, we investigated the effect of both castration and androgen replacement on spine plasticity in the nucleus accumbens shell and core (NAcSh and NAcC), caudate putamen (CPu), medial amygdala (MeA) and medial preoptic nucleus (MPN). Intact and castrated (gonadectomy [GDX]) male rats were treated with dihydrotestosterone (DHT, 1.5 mg) or vehicle (oil) in three experimental groups: intact-oil, GDX-oil and GDX-DHT. Spine density and morphology, measured 24 hours after injection, were determined through three-dimensional reconstruction of confocal z-stacks of DiI-labelled dendritic segments. We found that GDX decreased spine density in the MPN, which was rescued by DHT treatment. DHT also increased spine density in the MeA in GDX animals compared to intact oil-treated animals. By contrast, DHT decreased spine density in the NAcSh compared to GDX males. No effect on spine density was observed in the NAcC or CPu. Spine length and spine head diameter were unaffected by GDX and DHT in the investigated brain regions. In addition, immunohistochemistry revealed that DHT treatment of GDX animals rapidly increased the number of cell bodies in the NAcSh positive for phosphorylated cAMP response-element binding protein, a downstream messenger of the androgen receptor. These findings indicate that androgen signalling plays a role in the regulation of spine plasticity within neurocircuits involved in motivated behaviours.


Castration , Dihydrotestosterone/pharmacology , Motivation/drug effects , Neuronal Plasticity/drug effects , Reward , Amygdala/drug effects , Amygdala/metabolism , Animals , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Dendrites/drug effects , Dendrites/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats
9.
Curr Biol ; 31(2): 394-405.e4, 2021 01 25.
Article En | MEDLINE | ID: mdl-33188746

Endogenous sleep and general anesthesia are distinct states that share similar traits. Of particular interest to neuroscience is the loss of consciousness that accompanies both states. Multiple lines of evidence demonstrate that general anesthetics can co-opt the neural circuits regulating arousal to produce unconsciousness. However, controversy remains as to whether the neural circuits and, more specifically, the same neurons shaping sleep and wakefulness actually do influence the anesthetic state in vivo. Hypothalamic preoptic area (POA) neurons are intimately involved in modulating spontaneous and anesthetic-induced changes in arousal. Nevertheless, recent work suggests that POA GABAergic or glutamatergic neurons capable of regulating endogenous sleep fail to influence the onset or dissipation of anesthesia. We hypothesized that the POA's broad neuronal diversity could mask convergent roles of a subset of neurons in regulating both arousal and anesthesia. Contrary to a previously published report, we show that chemogenetic activation of POA Tac1 neurons obliterates both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep, strongly consolidating the waking state for hours, even during a period of elevated sleep drive. Moreover, chemogenetic activation of Tac1 POA neurons stabilizes the wake state against both isoflurane- and sevoflurane-induced unconsciousness. Tac1-activated mice display a partial resistance to entering isoflurane anesthesia and a more pronounced ability to exit both isoflurane- and sevoflurane-induced unconscious states. We conclude that POA Tac1 neurons can potently reinforce arousal both against endogenous and drug-induced unconscious states. POA Tac1 neurons thus add causal support for the involvement of arousal-regulating systems in the state of general anesthesia.


Anesthesia, Inhalation , Neurons/metabolism , Preoptic Area/physiology , Sleep/physiology , Wakefulness/physiology , Administration, Inhalation , Animals , Arousal/physiology , Electroencephalography , Female , Isoflurane/administration & dosage , Male , Mice , Mice, Transgenic , Neurons/drug effects , Preoptic Area/cytology , Preoptic Area/drug effects , Sevoflurane/administration & dosage , Sleep/drug effects , Stereotaxic Techniques , Tachykinins/genetics , Tachykinins/metabolism , Unconsciousness/chemically induced , Wakefulness/drug effects
10.
Article En | MEDLINE | ID: mdl-33013709

Numerous chemicals derived from human activity are now disseminated in the environment where their exert estrogenic endocrine disrupting effects, and therefore represent major health concerns. The present study explored whether Methoxychlor (MXC), an insecticide with xenoestrogens activities, given during the perinatal period (from gestational day 11 to postnatal day 8) and at an environmentally dose [20 µg/kg (body weight)/day], would affect reproductive physiology and sexual behavior of the offspring in mice. While MXC exposure did not induce any differences in the weight gain of animals from birth to 4 months of age, a clear difference (although in opposite direction according to the sexes) was observed on the anogenital distance between intact and exposed animals. A similar effect was also observed on preputial separation and vaginal opening, which reflects, respectively, in males and females, puberty occurrence. The advanced puberty observed in females was associated with an enhanced expression of kisspeptin cells in the anteroventral periventricular region of the medial preoptic area. Exposure to MXC did not induce in adult females changes in the estrous cycle or in the weight of the female reproductive tract. By contrast, males showed reduced weight of the epididymis and seminiferous vesicles associated with reduced testosterone levels and seminiferous tubule diameter. We also showed that both males and females showed deficits in mate preference tests. As a whole, our results show that MXC impacts reproductive outcomes.


Endocrine Disruptors/administration & dosage , Insecticides/administration & dosage , Methoxychlor/administration & dosage , Prenatal Exposure Delayed Effects/metabolism , Reproduction/drug effects , Sexual Behavior, Animal/drug effects , Animals , Estrous Cycle/drug effects , Female , Kisspeptins/metabolism , Mice , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Preoptic Area/drug effects , Preoptic Area/metabolism , Sexual Maturation/drug effects
11.
J Neuroendocrinol ; 32(11): e12908, 2020 11.
Article En | MEDLINE | ID: mdl-33034148

In addition to its critical role in lactation, the anterior pituitary hormone prolactin also influences a broad range of other physiological processes. In particular, widespread expression of prolactin receptor (Prlr) in the brain has highlighted pleiotropic roles for prolactin in regulating neuronal function, including maternal behaviour, reproduction and energy balance. Research into the central actions of prolactin has predominately focused on effects on gene transcription via the canonical JAK2/STAT5; however, it is evident that prolactin can exert rapid actions to stimulate activity in specific populations of neurones. We aimed to investigate how widespread these rapid actions of prolactin are in regions of the brain with large populations of prolactin-sensitive neurones, and whether physiological state alters these responses. Using transgenic mice where the Cre-dependent calcium indicator, GCaMP6f, was conditionally expressed in cells expressing the long form of the Prlr, we monitored changes in levels of intracellular calcium ([Ca2+ ]i ) in ex vivo brain slice preparations as a surrogate marker of cellular activity. Here, we surveyed hypothalamic regions implicated in the diverse physiological functions of prolactin such as the arcuate (ARC) and paraventricular nuclei of the hypothalamus (PVN), as well as the medial preoptic area (MPOA). We observed that, in the ARC of males and in both virgin and lactating females, prolactin can exert rapid actions to stimulate neuronal activity in the majority of Prlr-expressing neurones. In the PVN and MPOA, we found a smaller subset of cells that rapidly respond to prolactin. In these brain regions, the effects we detected ranged from rapid or sustained increases in [Ca2+ ]i to inhibitory effects, indicating a heterogeneous nature of these Prlr-expressing populations. These results enhance our understanding of mechanisms by which prolactin acts on hypothalamic neurones and provide insights into how prolactin might influence neuronal circuits in the mouse brain.


Hypothalamus/drug effects , Hypothalamus/metabolism , Neurons/metabolism , Prolactin/pharmacology , Receptors, Prolactin/drug effects , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Calcium Signaling , Female , Humans , Hypothalamus/cytology , Immunohistochemistry , Lactation , Male , Mice , Mice, Transgenic , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, Prolactin/genetics
12.
Neuropeptides ; 84: 102096, 2020 Dec.
Article En | MEDLINE | ID: mdl-33059245

Leptin mediates the interaction between reproductive function and energy balance. However, leptin receptors are not expressed in neurons that produce gonadotropin-releasing hormone (GnRH), likely indicating an indirect action through interneurons. Among likely neurons that modulate the secretion of GnRH are NO (nitric oxide) neurons. We assessed whether estradiol and feeding conditions modulate a possible interaction between leptin and NO in brain areas related to the control of reproductive function. Estradiol-treated and untreated ovariectomized rats were normally fed or fasted for 48 h. Then, saline (control) or leptin (3 µg/1 µl) intracerebroventricular microinjections were administered, and after thirty minutes, the brains collected subsequent to the decapitation or transcardially perfusion. Leptin and estradiol increased NO synthase (nNOS) gene expression (RT-PCR) and content (Western blotting) in the medial preoptic area (MPOA) and medial basal hypothalamus (MBH) only in fasted rats. Leptin increased: 1-phosphorylated-signal transducer and activator of transcription-3(pSTAT3) (immunohistochemistry) in the MPOA and various hypothalamic nuclei [arcuate (ARC); ventromedial (VMH); dorsal/ventral dorsomedial (dDMH/vDMH); premammilar ventral (PMV)], effects potentiated by estradiol/fasting interaction; 2- nNOS/pSTAT3 coexpression in the MPOA only in estradiol-treated, fasted rats; 3- nNOS-immunoreactive cell expression in the VMH, DMH and PMV (areas related to reproductive function control) of estradiol -treated rats. Thus, when leptin is reduced during fasting, leptin replacement effectively increased the expression of nitric oxide, which activated the HPG axis only in the presence of estradiol. Estradiol modulates the nitrergic system, leptin sensitivity and consequently leptin's effects on the nitrergic system in hypothalamus and in particular vDMH and PMV.


Estradiol/metabolism , Gonadotropin-Releasing Hormone/metabolism , Leptin/metabolism , Neurons/metabolism , Animals , Female , Hypothalamus/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism
13.
Endocrinology ; 161(11)2020 11 01.
Article En | MEDLINE | ID: mdl-33095238

Polycystic ovary syndrome (PCOS) is the most common form of infertility in women. The causes of PCOS are not yet understood and both genetics and early-life exposure have been considered as candidates. With regard to the latter, circulating androgens are elevated in mid-late gestation in women with PCOS, potentially exposing offspring to elevated androgens in utero; daughters of women with PCOS are at increased risk for developing this disorder. Consistent with these clinical observations, prenatal androgenization (PNA) of several species recapitulates many phenotypes observed in PCOS. There is increasing evidence that symptoms associated with PCOS, including elevated luteinizing hormone (LH) (and presumably gonadotropin-releasing hormone [GnRH]) pulse frequency emerge during the pubertal transition. We utilized translating ribosome affinity purification coupled with ribonucleic acid (RNA) sequencing to examine GnRH neuron messenger RNAs from prepubertal (3 weeks) and adult female control and PNA mice. Prominent in GnRH neurons were transcripts associated with protein synthesis and cellular energetics, in particular oxidative phosphorylation. The GnRH neuron transcript profile was affected more by the transition from prepuberty to adulthood than by PNA treatment; however, PNA did change the developmental trajectory of GnRH neurons. This included families of transcripts related to both protein synthesis and oxidative phosphorylation, which were more prevalent in adults than in prepubertal mice but were blunted in PNA adults. These findings suggest that prenatal androgen exposure can program alterations in the translatome of GnRH neurons, providing a mechanism independent of changes in the genetic code for altered expression.


Neurogenesis/drug effects , Neurons/drug effects , Prenatal Exposure Delayed Effects , Preoptic Area/drug effects , Virilism , Androgens/adverse effects , Animals , Female , Gene Expression Regulation, Developmental/drug effects , Gonadotropin-Releasing Hormone/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Neurons/metabolism , Neurons/physiology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/physiopathology , Preoptic Area/cytology , Preoptic Area/growth & development , Preoptic Area/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sex Factors , Virilism/chemically induced , Virilism/genetics , Virilism/physiopathology
14.
J Exp Zool A Ecol Integr Physiol ; 333(8): 550-560, 2020 10.
Article En | MEDLINE | ID: mdl-32798281

The developing brain is highly sensitive to the hormonal milieu, with gonadal steroid hormones involved in neurogenesis, neural survival, and brain organization. Limited available evidence suggests that endocrine-disrupting chemicals (EDCs) may perturb these developmental processes. In this study, we tested the hypothesis that prenatal exposure to a mixture of polychlorinated biphenyls (PCBs), Aroclor 1221, would disrupt the normal timing of neurogenesis in two hypothalamic regions: the ventromedial nucleus (VMN) and the preoptic area (POA). These regions were selected because of their important roles in the control of sociosexual behaviors that are perturbed in adulthood by prenatal EDC exposure. Pregnant Sprague-Dawley rats were exposed to PCBs from Embryonic Day 8 (E8) to E18, encompassing the period of neurogenesis of all hypothalamic neurons. To determine the birth dates of neurons, bromo-2-deoxy-5-uridine (BrdU) was administered to dams on E12, E14, or E16. On the day after birth, male and female pups were perfused, brains immunolabeled for BrdU, and numbers of cells counted. In the VMN, exposure to PCBs significantly advanced the timing of neurogenesis compared to vehicle-treated pups, without changing the total number of BrdU+ cells. In the POA, PCBs did not change the timing of neurogenesis nor the total number of cells born. This is the first study to show that PCBs can shift the timing of neurogenesis in the hypothalamus, specifically in the VMN but not the POA. This result has implications for functions controlled by the VMN, especially sociosexual behaviors, as well as for sexual selection more generally.


Endocrine Disruptors/pharmacology , Hypothalamus/drug effects , Neurogenesis/drug effects , Animals , Aroclors/pharmacology , Female , Fetus/drug effects , Neurons/drug effects , Polychlorinated Biphenyls/pharmacology , Pregnancy , Preoptic Area/cytology , Preoptic Area/drug effects , Rats , Rats, Sprague-Dawley , Sexual Behavior/drug effects , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects
15.
Horm Behav ; 125: 104827, 2020 09.
Article En | MEDLINE | ID: mdl-32735801

Estrogens play a key role in the sexual differentiation of the brain and behavior. While early estrogen actions exert masculinizing effects on the brain of male rodents, a diametrically opposite effect is observed in birds where estrogens demasculinize the brain of females. Yet, the two vertebrate classes express similar sex differences in the brain and behavior. Although ERα is thought to play a major role in these processes in rodents, the role of ERß is still controversial. In birds, the identity of the estrogen receptor(s) underlying the demasculinization of the female brain remains unclear. The aim of the present study was thus to determine in Japanese quail the effects of specific agonists of ERα (propylpyrazole triol, PPT) and ERß (diarylpropionitrile, DPN) administered at the beginning of the sensitive period (embryonic day 7, E7) on the sexual differentiation of male sexual behavior and on the density of vasotocin-immunoreactive (VT-ir) fibers, a known marker of the organizational action of estrogens on the quail brain. We demonstrate that estradiol benzoate and the ERß agonist (DPN) demasculinize male sexual behavior and decrease the density of VT-ir fibers in the medial preoptic nucleus and the bed nucleus of the stria terminalis, while PPT has no effect on these measures. These results clearly indicate that ERß, but not ERα, is involved in the estrogen-induced sexual differentiation of brain and sexual behavior in quail.


Brain/anatomy & histology , Coturnix/physiology , Estrogen Receptor beta/physiology , Sexual Behavior, Animal , Animals , Brain/drug effects , Brain/metabolism , Coturnix/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogens/pharmacology , Female , Male , Nitriles/pharmacology , Preoptic Area/drug effects , Preoptic Area/metabolism , Propionates/pharmacology , Sex Characteristics , Sex Differentiation/drug effects , Sexual Behavior, Animal/drug effects , Vasotocin/pharmacology
16.
Horm Behav ; 124: 104775, 2020 08.
Article En | MEDLINE | ID: mdl-32422195

The organizational-activational hypothesis indicates that activation of adult sexual behavior in males depends on organization of the masculine brain during the perinatal sensitive period. In the medial preoptic area such masculinization depends on a neuroendocrine cascade that includes exposure to testosterone, aromatization to estradiol, activation of estrogen receptors, synthesis of cyclooxygenase (COX), increase of prostaglandins, release of glutamate, and activation of AMPA receptors that result in the formation of more dendritic spines. Thus, in the present study we assessed the sexual partner preference (SPP) of adult male rats prenatally treated with acetaminophen (APAP), an analgesic/antipyretic drug that inhibits COX-2 and is commonly used and prescribed during pregnancy. Female rats received either saline (2 ml/kg s.c.) or APAP (50 mg/kg s.c.) every 12 h, during days 16-20 of pregnancy. At postnatal day PD60 half of the male offspring were exposed to sexual experience with receptive females during 5 trials, and the other half remained sexually naïve. At PD90 all them were tested for SPP with one sexually receptive female and one stud male. The results indicated that only APAP-naïve males failed to display SPP. However, APAP-experienced males displayed SPP for females. We discuss the effects of prenatal APAP in the disruption of unconditioned responses towards females (nature mechanisms), and the effects of sexual experience (nurture mechanisms) in the development of conditioned heterosexual preference.


Acetaminophen/pharmacology , Prenatal Exposure Delayed Effects , Sexual Behavior, Animal/drug effects , Animals , Brain/drug effects , Choice Behavior/drug effects , Estradiol/blood , Estradiol/pharmacology , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Preoptic Area/drug effects , Rats , Rats, Wistar , Sex Characteristics , Sexual Behavior, Animal/physiology , Testosterone/blood , Testosterone/pharmacology
17.
Neuropharmacology ; 171: 108069, 2020 07.
Article En | MEDLINE | ID: mdl-32275927

Neurotensin (NTS) is a neuropeptide acting as a neuromodulator in the brain and is a very potent hypothermic agent. However, the cellular mechanisms of actions are not fully understood. Here we report that NTS increases the firing rate of preoptic GABAergic neurons by activating both neurotensin receptor 1 (NTSR1) and neurotensin receptor 2 (NTSR2), expressed by neurons and astrocytes, respectively. Downstream of NTSR1 the neuropeptide activated an inward current, calcium release from intracellular stores and, postsynaptically, increased frequency and amplitude of inhibitory synaptic events. NTSR2 activation in astrocytes resulted in increased excitatory input in preoptic GABAergic neurons, an effect which was dependent upon the activation of P2X4 receptors. We also found that neuromedin N acted as a selective agonist at the NTSR1. Surprisingly, activation of both NTSR1 and NTSR2 in the median preoptic nucleus was required for activating a full hypothermic response.


Hypothermia/chemically induced , Hypothermia/physiopathology , Neurotensin , Preoptic Area/drug effects , Receptors, Neurotensin/agonists , Animals , Astrocytes/physiology , Excitatory Postsynaptic Potentials , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Patch-Clamp Techniques , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Neurotensin/genetics , Receptors, Purinergic P2X4/drug effects , gamma-Aminobutyric Acid/physiology
18.
Curr Biol ; 30(5): 779-787.e4, 2020 03 09.
Article En | MEDLINE | ID: mdl-32084397

The precise mechanism of general anesthesia remains unclear. In the last two decades, there has been considerable focus on the hypothesis that anesthetics co-opt the neural mechanisms regulating sleep. This hypothesis is supported by ample correlative evidence at the level of sleep-promoting nuclei, but causal investigations of potent inhaled anesthetics have not been conducted. Here, we tested the hypothesis that chemogenetic activation of discrete neuronal subpopulations within the median preoptic nucleus (MnPO) and ventrolateral preoptic nucleus (VLPO) of the hypothalamus would modulate sleep/wake states and alter the time to loss and resumption of consciousness associated with isoflurane, a potent halogenated ether in common clinical use. We show that activating MnPO/VLPO GABAergic or glutamatergic neurons does not alter anesthetic induction or recovery time. However, activation of these neuronal subpopulations did alter sleep-wake architecture. Notably, we report the novel finding that stimulation of VLPO glutamatergic neurons causes a strong increase in wakefulness. We conclude that activation of preoptic GABAergic or glutamatergic neurons that increase sleep or wakefulness does not substantively influence anesthetic state transitions. These data indicate that the correlative evidence for a mechanistic overlap of sleep and anesthesia at the level of an individual nucleus might not necessarily have strong causal significance.


Anesthetics, Inhalation/pharmacology , Isoflurane/pharmacology , Neurons/physiology , Preoptic Area/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Female , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Male , Mice , Neurons/drug effects , Preoptic Area/drug effects
19.
Horm Behav ; 121: 104709, 2020 05.
Article En | MEDLINE | ID: mdl-32007517

Territoriality is an adaptive behavioral trait that is important for animal's fitness and there still remains much to learn about the proximate mechanisms underlying the development of territoriality. We speculate that the formation of a conditioned place preference (CPP), an increased time allocation to the environment where a rewarding experience occurred, contributes to territoriality. Testosterone (T) plays an important role in modulating territorial behaviors and T pulses can induce a CPP. We confirmed previous findings in California mice (Peromyscus californicus) that T pulses can induce a CPP in singly-housed, but not group-housed males. Housing singly may be similar enough to dispersal in nature to initiate similar hormonal and neuroanatomical changes needed for the development of territoriality. We further revealed that T pulses interact with the single housing experience and appear to enhance the motivation to be aggressive towards a stimulus male. On a neural level, being singly housed upregulated levels of androgen receptors in the preoptic area, which positively correlated with the strength of the CPP. We speculate that this change in androgen sensitivity in the preoptic area is characteristic of males that have dispersed, making them more sensitive to T pulses. Also, single housing increased markers of synaptic plasticity in the nucleus accumbens, ventral and dorsal hippocampus, neural changes that may be associated with dispersal, reproduction and territory establishment. These behavioral and neural changes may reflect the life history transition from residing in the natal territory to dispersing and establishing a new territory.


Behavior, Animal/drug effects , Models, Biological , Peromyscus/physiology , Territoriality , Testosterone/pharmacology , Aggression/drug effects , Animals , Choice Behavior/drug effects , Conditioning, Psychological/drug effects , Housing, Animal , Male , Motivation/drug effects , Nesting Behavior/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, Androgen/metabolism , Reproduction/drug effects , Reward , Testosterone/physiology
20.
J Neuroendocrinol ; 32(2): e12827, 2020 02.
Article En | MEDLINE | ID: mdl-31917877

A population of neurones in the medial part of the medial preoptic area (mPOA) transiently express melanin-concentrating hormone (MCH) in mid to late lactation in the rat, and this expression disappears on weaning. Prolactin is known to mediate many of the physiological adaptations that occur within the dam associated with lactation and the mPOA is well endowed with prolactin receptors (Prlr); hence, we hypothesised that these transiently MCH-expressing cells may be regulated by prolactin. By in situ hybridisation, we show that approximately 60% of the cells expressing prepro-MCH (Pmch) mRNA in the medial part of the mPOA on day 19 of lactation also express Prlr mRNA. To demonstrate that these transiently MCH-expressing cells can acutely respond to prolactin, dams were treated with bromocriptine on the morning of day 19 of lactation and then given vehicle or prolactin 4 hours later. In the prolactin-treated animals, over 80% of the MCH-immunopositive cells were also immunopositive for phosphorylated signal transducer and activator of transcription 5, an indicator of prolactin receptor activation: double immunopositive cells were rare in vehicle-treated animals. Finally, the effect of manipulating the circulating concentrations of prolactin on days 17, 18 and 19 on the number of MCH-immunopositive cells on day 19 was determined. Reducing circulating concentrations of prolactin over days 17, 18 and 19 of lactation with or without a suckling stimulus resulted in a reduction (P < 0.05) in the number of MCH-immunopositive cells in the medial part of the mPOA on day 19 of lactation. Further research is required to determine the functional role(s) of these prolactin-activated transiently MCH-expressing neurones; however, we suggest the most likely role involves adaptations in maternal metabolism to support the final week of lactation.


Hypothalamic Hormones/metabolism , Lactation/metabolism , Preoptic Area/metabolism , Prolactin/metabolism , Protein Precursors/metabolism , Animals , Female , Preoptic Area/drug effects , Prolactin/administration & dosage , RNA, Messenger/metabolism , Rats, Sprague-Dawley
...