Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 318
Filter
1.
Environ Sci Pollut Res Int ; 29(1): 936-948, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34345985

ABSTRACT

Fipronil (FIP) is a highly effective insecticide that has been used in agriculture and veterinary medicine. Its neurotoxic effect to insects and to non-target organisms, after nonintentional exposure, was reported. Many studies were conducted to evaluate FIP effects on mammals. However, slight is known about its effect on the brain stem and diencephalon. The current study was designed to investigate the ability of FIP to induce oxidative stress as a molecular mechanism of FIP neurotoxicity that resulted in apoptosis and neural tissue reactivity in these regions. Ten adult male rats received 10 mg/kg of FIP technical grade by oral gavage, daily for 45 days. Brain stem and diencephalon were processed to examine oxidative stress-induced macromolecular alteration (MDA, PCC and DNA fragmentation). Also, the histopathological assessment and immunoreactivity for caspase-3 (active form), iNOS and GFAP were performed on the thalamus, hypothalamus and medulla oblongata. Our results revealed that FIP significantly raised MDA, PCC and DNA fragmentation (p ≤ 0.05). In addition, significantly increased immunoreactivity to GFAP, iNOS and caspase-3 (active form) in the FIP-treated group was noticed (p ≤ 0.05). Moreover, alterations in the histoarchitecture of the neural tissue of these regions were observed. We conclude that FIP can induce oxidative stress, leading to apoptosis and tissue reaction in brain stem and diencephalon.


Subject(s)
Apoptosis , Brain Stem/pathology , Diencephalon/pathology , Oxidative Stress , Pyrazoles/toxicity , Animals , Apoptosis/drug effects , Brain Stem/drug effects , Diencephalon/drug effects , Insecticides/toxicity , Male , Oxidative Stress/drug effects , Rats
2.
J Physiol ; 596(16): 3775-3791, 2018 08.
Article in English | MEDLINE | ID: mdl-29874406

ABSTRACT

KEY POINTS: Perioral tactile signals are transmitted via the infraorbital nerve (ION) to trigeminal nuclei. Each cerebellar Purkinje cell (PC) receives this signal as complex spikes (CSs) via a climbing fibre (CF) emerging from the inferior olive (IO). The anatomical pathway from trigeminal nuclei to the IO is not clearly identified. In the present study, we examined candidate anatomical pathways for perioral sensory signalling by analysing CSs recorded from PCs in male mice by single unit recording. CS generation by ION stimulation was inhibited by injection of a GABAA receptor agonist, muscimol, into the contralateral mesodiencephalic junction, which is referred to as the area parafascicularis prerubralis (PfPr). The number of CSs evoked by mechanical whisker stimulation was also decreased by contralateral PfPr inhibition. These results suggest the existence of a sensory signalling pathway to the IO via the PfPr in mice. ABSTRACT: Perioral tactile signals are transmitted via the infraorbital nerve (ION) to trigeminal nuclei. Each cerebellar Purkinje cell receives this signal as complex spikes (CSs) via a climbing fibre emerging from the inferior olive (IO). However, the anatomical pathway from the trigeminal nuclei to the IO is not clearly identified. In the present study, we recorded CSs from Purkinje cells in male mice by single unit recording, and examined the signal transduction pathway. CSs were evoked by electrical stimulation of the ipsilateral or contralateral ION with a latency of 20-70 ms. CS generation by ipsilateral ION stimulation was inhibited by injection of a GABAA receptor agonist, muscimol, into the contralateral mesodiencephalic junction, ranging from around the fasciculus retroflexus to the interstitial nucleus of Cajal, which is referred to as the area parafascicularis prerubralis (PfPr). CSs evoked by contralateral ION stimulation were also suppressed by muscimol injection into the PfPr, although the effective area was more restricted. Furthermore, CSs evoked by mechanical stimulation around the whisker region were suppressed by PfPr inhibition. We also found that the primary motor cortex plays a role to suppress this signalling pathway. These results indicate the existence of an anatomical pathway for conducting perioral sensory signals to the IO via the PfPr.


Subject(s)
Cerebellum/physiology , Diencephalon/physiology , Mesencephalon/physiology , Mouth/physiology , Olivary Nucleus/physiology , Purkinje Cells/physiology , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Diencephalon/cytology , Diencephalon/drug effects , GABA-A Receptor Agonists/pharmacology , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Mice , Mice, Inbred C57BL , Mouth/cytology , Mouth/drug effects , Muscimol/pharmacology , Olivary Nucleus/cytology , Olivary Nucleus/drug effects , Purkinje Cells/cytology , Purkinje Cells/drug effects , Receptors, GABA-A/chemistry , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects
3.
Physiol Behav ; 194: 199-204, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29775631

ABSTRACT

In mammals, anorexia accompanying infection is thought to be mediated via cytokines including interleukins, interferons (IFNs), and tumor necrosis factor (TNF). However, there is a lack of related knowledge on birds. Therefore, the purpose of the present study was to determine if cytokines are associated with reduced food intake in chicks (Gallus gallus). Specifically, we evaluated the effects of TNF-like cytokine 1A (TL1A), a member of the TNF family, interferon-α (IFN-α), and interferon-γ (IFN-γ) on food intake. Additionally, the effect of lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid (poly I:C) on cytokine mRNA expression in the diencephalon and spleen was also measured. Intracerebroventricular (ICV) injection of 0.05 or 0.5 µg TL1A, IFN-α, and IFN-γ had no effect on food intake. However, when 1.0 µg each of these factors was evaluated, TL1A significantly decreased food intake at 180 and 240 min after the injection, but IFN-α and IFN-γ had no effect. When chicks received intraperitoneal (IP) injections of 100 µg LPS or 400 µg poly I:C, their food intake was reduced. Diencephalic mRNA expression of TL1A was significantly decreased following IP injection of LPS or poly I:C. Additionally, diencephalic mRNA expression of IFN-γ mRNA was significantly increased by IP injection of LPS but decreased by IP injection of poly I:C. For the spleen, IP injection of LPS and poly I:C both significantly increased TL1A and IFN-γ mRNA expression. In sum, we have provided evidence that central TL1A but not IFN-α or IFN-γ are related to reduction of food intake in chicks, but the role of these cytokines for mediating anorexia associated with infections may differ from mammals.


Subject(s)
Anorexia/immunology , Eating/immunology , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Animals , Anorexia/etiology , Chickens , Diencephalon/drug effects , Diencephalon/immunology , Eating/drug effects , Illness Behavior/physiology , Interferon-alpha/administration & dosage , Interferon-gamma/administration & dosage , Lipopolysaccharides , Male , Poly I-C , RNA, Messenger/metabolism , Spleen/drug effects , Spleen/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/administration & dosage
4.
Fish Physiol Biochem ; 44(3): 817-828, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29404822

ABSTRACT

Most wrasses are protogynous species that swim to feed, reproduce during the daytime, and bury themselves under the sandy bottom at night. In temperate and subtropical wrasses, low temperature influences emergence from the sandy bottom in the morning, and induces a hibernation-like state in winter. We cloned and characterized the prohormone complementary DNAs (cDNAs) of arginine vasotocin (AVT) and isotocin (IT) in a temperate wrasse (Halichoeres tenuispinis) and examined the effects of day/night and temperature on their expression in the diencephalon, because these neurohypophysial peptides are related to the sex behavior of wrasses. The full-length cDNAs of pro-AVT and pro-IT were 938 base pairs (154 amino acids) and 759 base pairs (156 amino acids) in length, respectively. Both pro-peptides contained a signal sequence followed by the respective hormones and neurophysin connected by a Gly-Lys-Arg bridge. Reverse-transcription polymerase chain reaction (RT-PCR) revealed that pro-AVT mRNA expression was specifically observed in the diencephalon, whereas pro-IT mRNA expression was seen in the whole brain. Quantitative RT-PCR revealed that the mRNA abundance of pro-AVT and pro-IT was higher at midday (zeitgeber time 6; ZT6) than at midnight (ZT18) under 12 h light and 12 h darkness (LD 12:12) conditions, but not under constant light. Intraperitoneal injection of melatonin decreased the mRNA abundance of pro-AVT, but not of pro-IT. When fish were reared under LD 12:12 conditions at 25, 20, and 15 °C, day high and night low mRNA expressions of pro-AVT and pro-IT were maintained. A field survey revealed seasonal variation in the number of swimming fish at observatory sites; many fish emerged from the sandy bottom in summer, but not in winter, suggesting a hibernation-like state under the sandy bottom under low temperature conditions. We conclude that the day-night fluctuation of pro-AVT and pro-IT mRNA abundance in the brain is not affected by temperature and repeated under the sandy bottom in winter.


Subject(s)
Diencephalon/metabolism , Fishes/genetics , Melatonin/pharmacology , Oxytocin/analogs & derivatives , Temperature , Vasotocin/genetics , Animals , Diencephalon/drug effects , Oxytocin/genetics , Periodicity , RNA, Messenger/metabolism
5.
J Neuroendocrinol ; 29(8)2017 08.
Article in English | MEDLINE | ID: mdl-28683170

ABSTRACT

Gonadotrophin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotrophin synthesis and release in birds and mammals. In Japanese quail, GnIH neurones express the noradrenergic receptor and receive noradrenergic innervation. Treatment with noradrenaline (NA) stimulates GnIH release from diencephalic tissue blocks in vitro. However, the effects of NA on hypothalamic GnIH gene expression have not been determined. We investigated noradrenergic regulation of GnIH gene expression in the brain of male quail using the selective noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP-4). We first showed that DSP-4 reduced the number of noradrenergic (dopamine-ß-hydroxylase immunoreactive) cells in the locus coeruleus (LoC) and specifically lowered the NA concentration in the hypothalamus of male quail. Other monoamines, such as dopamine and serotonin, were not affected by drug treatment. DSP-4 did not decrease the numbers of noradrenergic cells of the lateral tegmental cell group, nor the plasma NA concentration. Decreased hypothalamic NA levels after DSP-4 treatment did not change GnIH gene expression in the brains of quail during their interaction with conspecifics. On the other hand, GnIH gene expression increased in the brains of quail socially isolated for 1 hour after DSP-4 treatment. These results suggest that some noradrenergic neurones have inhibitory effects on GnIH gene expression of the hypothalamus in solitary quail.


Subject(s)
Adrenergic Neurons/metabolism , Avian Proteins/genetics , Coturnix , Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Animals , Benzylamines/administration & dosage , Blood Glucose/metabolism , Diencephalon/drug effects , Diencephalon/metabolism , Gene Expression , Hypothalamus/drug effects , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Norepinephrine/blood , RNA, Messenger/metabolism , Social Isolation
6.
Rev Neurol ; 64(2): 70-74, 2017 Jan 16.
Article in Spanish | MEDLINE | ID: mdl-28075000

ABSTRACT

INTRODUCTION: Akinetic mutism is considered as an alteration of the motivational state of the person, which the patient is unable to initiate verbal or motor responses voluntary, even with preserved sensorimotor and surveillance functions. CASE REPORT: A 43 year-old male involved in a cerebellum arteriovenous fistula complicated with hydrocephalus, who responded dramatically to treatment with bromocriptine. CONCLUSION: Typically, akinetic mutism is described as a transient surgeries posterior fossa. However, it can also occur after multiple valvular failure in patients with hydrocephalus.


TITLE: Bromocriptina: podria ser la cura para el mutismo acinetico posquirurgico?Introduccion. El mutismo acinetico se considera una alteracion del estado motivacional de la persona, por el cual el paciente es incapaz de iniciar respuestas verbales o motoras de caracter voluntario, aun teniendo preservadas las funciones sensomotoras y de vigilancia. Caso clinico. Varon de 43 años, intervenido de una fistula arteriovenosa del cerebelo complicada con hidrocefalia, que respondio espectacularmente al tratamiento con bromocriptina. Conclusion. Tipicamente se ha descrito el mutismo acinetico como una complicacion transitoria de las cirugias de la fosa posterior. Sin embargo, tambien puede aparecer tras multiples fallos valvulares en pacientes con hidrocefalia.


Subject(s)
Akinetic Mutism/drug therapy , Bromocriptine/therapeutic use , Dopamine Agonists/therapeutic use , Postoperative Complications/drug therapy , Adult , Akinetic Mutism/diagnostic imaging , Central Nervous System Vascular Malformations/complications , Central Nervous System Vascular Malformations/diagnostic imaging , Central Nervous System Vascular Malformations/surgery , Cerebellum/blood supply , Cerebellum/surgery , Cerebral Angiography , Diencephalon/drug effects , Diencephalon/physiopathology , Emergencies , Equipment Failure , Humans , Hydrocephalus/etiology , Hydrocephalus/surgery , Intracranial Arteriovenous Malformations/complications , Intracranial Arteriovenous Malformations/diagnostic imaging , Intracranial Arteriovenous Malformations/surgery , Male , Mesencephalon/drug effects , Mesencephalon/physiopathology , Tomography, X-Ray Computed
7.
Pharmacol Rep ; 68(5): 990-5, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27391358

ABSTRACT

BACKGROUND: This study aimed to evaluate the effect of extended olanzapine, clozapine and haloperidol administration on NMDA-R subunit immunoexpression in the rat neocortex and diencephalon. METHODS: To explore NR1, NR2A and NR2B subunit protein expression, densytometric analysis of immunohistochemically stained brain slices was performed. RESULTS: Interestingly, all neuroleptics caused a downregulation of NMDA-R subunit expression in the thalamus but increased the level of NR1 in the hypothalamus. Olanzapine upregulated hypothalamic NR2A expression, while clozapine and haloperidol decreased hypothalamic levels. We observed no significant changes in NR2B immunoreactivity. None of the studied medications had significant influence on NMDA-R subunit expression in the neocortex. CONCLUSIONS: Neuroleptic-induced reduction in the expression of thalamic NMDA-R subunits may play an important role in the regulation of glutamatergic transmission disorders in cortico-striato-thalamo-cortical loop in schizophrenia. A decrease in NMDA signaling in this region after long-term neuroleptic administration may also cautiously explain the incomplete effectiveness of these drugs in the therapy of schizophrenia-related cognitive disturbances.


Subject(s)
Antipsychotic Agents/pharmacology , Diencephalon/drug effects , N-Methylaspartate/metabolism , Neocortex/drug effects , Protein Subunits/metabolism , Animals , Benzodiazepines/pharmacology , Clozapine/pharmacology , Diencephalon/metabolism , Down-Regulation/drug effects , Haloperidol/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamus/drug effects , Male , Neocortex/metabolism , Olanzapine , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
8.
Physiol Behav ; 164(Pt A): 268-76, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27317836

ABSTRACT

Recently, it was found that the avian central vasotocin receptor (V1aR) is associated with the regulation of food intake. To identify V1aR-containing brain structures regulating food intake, a selective V1aR antagonist SR-49059 that induced food intake was administrated intracerebroventricularly in male chickens followed by detection of brain structures using FOS immunoreactivity. Particularly, the hypothalamic core region of the paraventricular nucleus, lateral hypothalamic area, dorsomedial hypothalamic nucleus, a subnucleus of the central extended amygdalar complex [dorsolateral bed nucleus of the stria terminalis], medial septal nucleus and caudal brainstem [nucleus of the solitary tract] showed significantly increased FOS-ir cells. On the other hand, the supraoptic nucleus of the preoptic area and the nucleus of the hippocampal commissure of the septum showed suppressed FOS immunoreactivity in the V1aR antagonist treatment group. Further investigation revealed that neuronal activity of arginine vasotocin (AVT-ir) magnocellular neurons in the supraoptic nucleus, preoptic periventricular nucleus, paraventricular nucleus and ventral periventricular hypothalamic nucleus and most likely corticotropin releasing hormone (CRH-ir) neurons in the nucleus of the hippocampal commissure were reduced following the antagonist treatment. Dual immunofluorescence labeling results showed that perikarya of AVT-ir magnocellular neurons in the preoptic area and hypothalamus were colabeled with V1aR. Within the nucleus of the hippocampal commissure, CRH-ir neurons were shown in close contact with V1aR-ir glial cells. Results of the present study suggest that the V1aR plays a role in the regulation of food intake by modulating neurons that synthesize and release anorectic neuropeptides in the avian brain.


Subject(s)
Appetite Regulation/physiology , Avian Proteins/metabolism , Diencephalon/metabolism , Eating/physiology , Receptors, Vasopressin/metabolism , Septum of Brain/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Appetite Regulation/drug effects , Appetitive Behavior/drug effects , Appetitive Behavior/physiology , Avian Proteins/antagonists & inhibitors , Central Nervous System Agents/administration & dosage , Chickens , Diencephalon/cytology , Diencephalon/drug effects , Eating/drug effects , Indoles/pharmacology , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Pyrrolidines/pharmacology , Random Allocation , Septum of Brain/cytology , Septum of Brain/drug effects
9.
Dev Dyn ; 245(6): 667-77, 2016 06.
Article in English | MEDLINE | ID: mdl-27005305

ABSTRACT

BACKGROUND: Antioxidants such as the green tea polyphenol epigallocatechin gallate (EGCG) are neuroprotective under many conditions in mature nervous systems; however, their impact has rarely been explored in developing nervous systems, in which a critical step is the formation of connections between neurons. Axons emerge from newly formed neurons and are led by a dynamic structure found at their tip called a growth cone. Here we explore the impact of EGCG on the development of retinal ganglion cell (RGC) axons, which connect the eye to the brain. RESULTS: EGCG acts directly on RGC axons to increase the number of growth cone filopodia, fingerlike projections that respond to extrinsic signals, in vitro and in vivo. Furthermore, EGCG exposure leads to a dramatic defect in the guided growth of RGC axons where the axons fail to make a key turn in the mid-diencephalon required to reach their target. Intriguingly, at guidance points where RGCs do not show a change in direction, EGCG has no influence on RGC axon behavior. CONCLUSIONS: We propose that EGCG stabilizes filopodia and prevents normal filopodial dynamics required for axons to change their direction of outgrowth at guidance decision points. Developmental Dynamics 245:667-677, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Axons/drug effects , Axons/metabolism , Catechin/analogs & derivatives , Growth Cones/drug effects , Pseudopodia/drug effects , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Animals , Axon Guidance/drug effects , Brain/cytology , Brain/drug effects , Catechin/pharmacology , Chorionic Gonadotropin/pharmacology , Diencephalon/cytology , Diencephalon/drug effects , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Female , Humans , In Situ Hybridization , Neurogenesis/drug effects , Retina/cytology , Retina/drug effects , Xenopus
10.
Mol Cell Endocrinol ; 420: 75-84, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26586207

ABSTRACT

In the frog Pelophylax esculentus, the endocannabinoid anandamide (AEA) modulates Gonadotropin Releasing Hormone (GnRH) system in vitro and down-regulates steroidogenic enzymes in vivo. Thus, male frogs were injected with AEA ± SR141716A, a cannabinoid receptor 1 (CB1) antagonist, to evaluate possible effects on GnRH and Kiss1/Gpr54 systems, gonadotropin receptors and steroid levels. In frog diencephalons, AEA negatively affected both GnRH and Kiss1/Gpr54 systems. In testis, AEA induced the expression of gonadotropin receptors, cb1, gnrh2 and gnrhr3 meanwhile reducing gnrhr2 mRNA and Kiss1/Gpr54 proteins. Furthermore, aromatase (Cyp19) expression increased in parallel to testosterone decrease and estradiol increase. In vitro treatment of testis with AEA revealed direct effects on Cyp19 and induced the expression of the AEA-degrading enzyme Faah. Lastly, AEA effects on Faah were counteracted by the antiestrogen ICI182780, indicating estradiol mediated effect. In conclusion, for the first time we show in a vertebrate that AEA regulates testicular activity through kisspeptin system.


Subject(s)
Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , Kisspeptins/metabolism , Polyunsaturated Alkamides/pharmacology , Rana esculenta/metabolism , Testis/metabolism , Amidohydrolases/metabolism , Animals , Aromatase/metabolism , Diencephalon/drug effects , Diencephalon/metabolism , Estradiol/metabolism , Male , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Gonadotropin/metabolism , Rimonabant , Testosterone/metabolism
11.
Pharmacol Res ; 103: 328-39, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26657418

ABSTRACT

Histone deacetylases (HDACs) are key epigenetic enzymes and emerging drug targets in cancer and neurodegeneration. Pan-HDAC inhibitors provided neuroprotection in Parkinson's Disease (PD) models, however, the HDAC isoforms with highest neuroprotective potential remain unknown. Zebrafish larvae (powerful pharmacological testing tools bridging cellular and in vivo studies) have thus far been used in PD modelling with limited phenotypic characterization. Here we characterize the behavioural and metabolic phenotypes of a zebrafish PD model induced with MPP(+), assess the feasibility of targeting zebrafish HDAC1 and HDAC6 isoforms, and test the in vivo effects of their selective inhibitors MS-275 and tubastatin A, respectively. MPP(+) induced a concentration-dependent decrease in metabolic activity and sensorimotor reflexes, and induced locomotor impairments rescuable by the dopaminergic agonist apomorphine. Zebrafish HDAC1 and HDAC6 isoforms show high sequence identity with mammalian homologues at the deacetylase active sites, and pharmacological inhibition increased acetylation of their respective histone and tubulin targets. MS-275 and tubastatin rescued the MPP(+)-induced decrease in diencephalic tyrosine hydroxylase immunofluorescence and in whole-larvae metabolic activity, without modifying mitochondrial complex activity or biogenesis. MS-275 or tubastatin alone modulated spontaneous locomotion. When combined with MPP(+), however, neither MS-275 nor tubastatin rescued locomotor impairments, although tubastatin did ameliorate the head-reflex impairment. This study demonstrates the feasibility of pharmacologically targeting the zebrafish HDAC1 and HDAC6 isoforms, and indicates that their inhibition can rescue cellular metabolism in a PD model. Absence of improvement in locomotion, however, suggests that monotherapy with either HDAC1 or HDAC6 inhibitors is unlikely to provide strong benefits in PD. This study highlights parameters dependent on the integrity of zebrafish neuronal circuits as a valuable complement to cell-based studies. Also, the demonstrated feasibility of pharmacologically targeting HDAC1 and HDAC6 in this organism paves the way for future studies investigating HDAC inhibitors in other diseases modelled in zebrafish.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Parkinson Disease, Secondary , Pyridines/pharmacology , Zebrafish Proteins/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium , Animals , Behavior, Animal/drug effects , Diencephalon/drug effects , Diencephalon/metabolism , Disease Models, Animal , Histone Deacetylase 1/genetics , Histone Deacetylase 6 , Histone Deacetylases/genetics , Larva , Locomotion/drug effects , Neurotoxins , Oxazines/metabolism , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/physiopathology , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/metabolism , Xanthenes/metabolism , Zebrafish , Zebrafish Proteins/genetics
12.
Neurotoxicol Teratol ; 52(Pt A): 51-6, 2015.
Article in English | MEDLINE | ID: mdl-26546233

ABSTRACT

Dopamine signaling is conserved across all animal species and has been implicated in the disease process of many neurological disorders, including Parkinson's disease (PD). The primary neuropathology in PD involves the death of dopaminergic cells in the substantia nigra (SN), an anatomical region of the brain implicated in dopamine production and voluntary motor control. Increasing evidence suggests that the neurotransmitter dopamine may have a neurotoxic metabolic product (DOPAL) that selectively damages dopaminergic cells. This study was designed to test this theory of oxidative damage in an animal model of Parkinson's disease, using a transgenic strain of zebrafish with fluorescent labeling of cells that express the dopamine transporter. The pretectum and ventral diencephalon exhibited reductions in cell numbers due to L-DOPA treatment while reticulospinal neurons that do not express the DAT were unaffected, and this was partially rescued by monoamine oxidase inhibition. Consistent with the MPTP model of PD in zebrafish larvae, spontaneous locomotor behavior in L-DOPA treated animals was depressed following a 24-h recovery period, while visually-evoked startle response rates and latencies were unaffected.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Levodopa/toxicity , Motor Activity/drug effects , Animals , Diencephalon/drug effects , Diencephalon/metabolism , Disease Models, Animal , Oxidative Stress/drug effects , Pretectal Region/drug effects , Pretectal Region/metabolism , Zebrafish
13.
Neuropeptides ; 48(5): 305-11, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25005752

ABSTRACT

Exogenous administration of substance P (SP) exerts anorexigenic effects in both chicks and rats, but the central mechanism mediating this response is poorly understood. Therefore, this study was designed to elucidate mechanisms of SP-induced anorexia using chicks as models. Chicks that received intracerebroventricular (ICV) injections of SP dose-dependably reduced their food intake with no effect on water intake. Next, the diencephalon was isolated from SP-injected chicks and mRNA expression of neuropeptide Y (NPY), corticotropin releasing factor (CRF), urocortin 3 (UCN 3) and CRF receptors were measured but were not affected. When measured in the hypothalamus, mRNA abundance of these and NPY receptors, urotensin 2 (UTS2) and melanocortin receptor 4 (MCR4) were not affected by SP-injection. Quantification of c-Fos immunoreactivity in appetite-associated hypothalamic nuclei demonstrated that the paraventricular nucleus (PVN) was activated in SP-injected chicks. Finally, in the PVN isolated from SP-injected chicks, there was increased expression of UTS2 mRNA while CRF and UCN3 were not affected. Thus, the anorexigenic effects of SP appear to be mediated by PVN activation and may involve UTS2.


Subject(s)
Appetite Depressants/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Substance P/pharmacology , Urotensins/metabolism , Animals , Anorexia/chemically induced , Anorexia/metabolism , Chickens , Diencephalon/drug effects , Diencephalon/metabolism , Disease Models, Animal , Drinking/drug effects , Eating/drug effects , Female , Infusions, Intraventricular , Male , RNA, Messenger/metabolism , Substance P/administration & dosage
14.
Neurosci Lett ; 567: 63-7, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24686181

ABSTRACT

Central congenital hypoventilation syndrome is a neuro-respiratory disease characterized by the dysfunction of the CO2/H(+) chemosensitive neurons of the retrotrapezoid nucleus/parafacial respiratory group. A recovery of CO2/H(+) chemosensitivity has been observed in some central congenital hypoventilation syndrome patients coincidental with contraceptive treatment by a potent progestin, desogestrel (Straus et al., 2010). The mechanisms of this progestin effect remain unknown, although structures of medulla oblongata, midbrain or diencephalon are known to be targets for progesterone. In the present study, on ex vivo preparations of central nervous system of newborn rats, we show that acute exposure to etonogestrel (active metabolite of desogestrel) enhanced the increased respiratory frequency induced by metabolic acidosis via a mechanism involving supramedullary structures located in pontine, mesencephalic or diencephalic regions.


Subject(s)
Acidosis/physiopathology , Desogestrel/pharmacology , Progestins/pharmacology , Respiratory Center/drug effects , Animals , Animals, Newborn , Diencephalon/drug effects , Diencephalon/physiopathology , Hydrogen-Ion Concentration , Medulla Oblongata/drug effects , Medulla Oblongata/physiopathology , Mesencephalon/drug effects , Mesencephalon/physiopathology , Rats, Sprague-Dawley , Respiratory Center/physiopathology
15.
Gen Comp Endocrinol ; 195: 9-20, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24505600

ABSTRACT

The first studies that identified leptin and its receptor (LepR) in mammals were based on mutant animals that displayed dramatic changes in body-weight and regulation of energy homeostasis. Subsequent studies have shown that a deficiency of leptin or LepR in homoeothermic mammals results in hyperphagia, obesity, infertility and a number of other abnormalities. The physiological roles of leptin-mediated signaling in ectothermic teleosts are still being explored. Here, we produced medaka with homozygous LepR gene mutation using the targeting induced local lesions in a genome method. This knockout mutant had a point mutation of cysteine for stop codon at the 357th amino acid just before the leptin-binding domain. The evidence for loss of function of leptin-mediated signaling in the mutant is based on a lack of response to feeding in the expression of key appetite-related neuropeptides in the diencephalon. The mutant lepr−/− medaka expressed constant up-regulated levels of mRNA for the orexigenic neuropeptide Ya and agouti-related protein and a suppressed level of anorexigenic proopiomelanocortin 1 in the diencephalon independent of feeding, which suggests that the mutant did not possess functional LepR. Phenotypes of the LepR-mutant medaka were analyzed in order to understand the effects on food intake, growth, and fat accumulation in the tissues. The food intake of the mutant medaka was higher in post-juveniles and adult stages than that of wild-type (WT) fish. The hyperphagia led to a high growth rate at the post-juvenile stage, but did not to significant alterations in final adult body size. There was no additional deposition of fat in the liver and muscle in the post-juvenile and adult mutants, or in the blood plasma in the adult mutant. However, adult LepR mutants possessed large deposits of visceral fat, unlike in the WT fish, in which there were none. Our analysis confirms that LepR in medaka exert a powerful influence on the control on food intake. Further analyses using the mutant will contribute to a better understanding of the role of leptin in fish. This is the first study to produce fish with leptin receptor deficiency.


Subject(s)
Animals, Genetically Modified/growth & development , Body Weight/drug effects , Eating/physiology , Gene Knockout Techniques , Intra-Abdominal Fat/drug effects , Neuropeptides/pharmacology , Receptors, Leptin/physiology , Agouti-Related Protein/metabolism , Animals , Animals, Genetically Modified/metabolism , Appetite/drug effects , Appetite/physiology , Diencephalon/drug effects , Diencephalon/metabolism , Eating/drug effects , Hyperphagia/genetics , Hyperphagia/pathology , Leptin/metabolism , Mutation/genetics , Obesity/metabolism , Oryzias/genetics , Oryzias/growth & development , Oryzias/metabolism , Up-Regulation
16.
Gen Comp Endocrinol ; 199: 86-93, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24472706

ABSTRACT

In sheep as in rat, it has been highly suggested that neuronal histamine (HA) participates to the estradiol (E2)-induced GnRH and LH surges, through H1 receptor. With the aim of determining if E2 could act directly on HA neurons, we examined here whether HA neurons express estrogen receptor alpha (ERα) in the ewe diencephalon during the breeding season. We first produced a specific polyclonal antibody directed against recombinant ovine histidine decarboxylase (oHDC), the HA synthesizing enzyme. Using both this anti-oHDC antibody and an anti-ERα monoclonal antibody in double label immunohistochemistry, we showed that HA neurons do not express ERα in diencephalon of ewes with different hormonal status. This result diverges from those obtained in rat, in which around three quarters of HA neurons express ERα in their nucleus. This discrepancy between these two mammal species may reflect difference in their neuronal network.


Subject(s)
Breeding , Diencephalon/metabolism , Estrogen Receptor alpha/metabolism , Histamine/metabolism , Neurons/metabolism , Seasons , Sheep/metabolism , Animals , Antibodies/metabolism , Antibody Specificity/immunology , Diencephalon/cytology , Diencephalon/drug effects , Diencephalon/enzymology , Electrophoresis, Polyacrylamide Gel , Estradiol/pharmacology , Female , Histidine Decarboxylase/immunology , Immunohistochemistry , Male , Neurons/drug effects , Rats , Recombinant Proteins/isolation & purification
17.
J Steroid Biochem Mol Biol ; 141: 37-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24434281

ABSTRACT

As one of the third generation of aromatase inhibitors, letrozole is a favored drug for the treatment of hormone receptor-positive breast cancer with some adverse effects on the nervous system, but the knowledge is limited and the results are controversial, the mechanism underlying its central action is also unclear. Accumulated evidences have demonstrated that estrogens derived from androgens by aromatase play profound roles in the brain through their receptors, which needs coactivator for the transcription regulation, among which steroid receptor coactivator-1 (SRC-1) has been shown to be multifunctional potentials in the brain, but whether it is regulated by letrozole is currently unknown. In this study, we examined letrozole regulation on SRC-1 expression in adult mice brain using immunohistochemistry. The results showed that letrozole induced dramatic decrease of SRC-1 in the medial septal, hippocampus, medial habenular nucleus, arcuate hypothalamic nucleus and superior colliculus (p<0.01). Significant decrease was detected in the dorsal lateral septal nucleus, bed nucleus of stria terminalis, ventral taenia tecta, dorsomedial and ventromedial hypothalamic nuclei, dorsomedial periaqueductal gray, superior paraolivary nucleus and pontine nucleus (p<0.05). In the hippocampus, levels of estradiol content, androgen receptor, estrogen receptor α and ß also decreased significantly after letrozole injection. The above results demonstrated letrozole downregulation of SRC-1 in specific regions that are primarily related to learning and memory, cognition and mood, neuroendocrine as well as information integration, indicating that SRC-1 may be one important downstream central target of letrozole. Furthermore, these potential central adverse effects of letrozole should be taken into serious considerations.


Subject(s)
Aromatase Inhibitors/pharmacology , Down-Regulation/drug effects , Hippocampus/metabolism , Nitriles/pharmacology , Nuclear Receptor Coactivator 1/metabolism , Triazoles/pharmacology , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Cerebellum/drug effects , Cerebellum/metabolism , Diencephalon/drug effects , Diencephalon/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Hippocampus/drug effects , Letrozole , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Motor Cortex/drug effects , Motor Cortex/metabolism , Neurosecretory Systems/drug effects , Neurosecretory Systems/metabolism , Nuclear Receptor Coactivator 1/genetics , Organ Specificity , Receptors, Androgen/metabolism , Telencephalon/drug effects , Telencephalon/metabolism
18.
Neuroscience ; 260: 106-19, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24361177

ABSTRACT

Brain injury due to neonatal hypoxia-ischemia (HI) is more homogenously severe in male than in female mice. Because, necrostatin-1 (nec-1) prevents injury progression only in male mice, we hypothesized that changes in brain-derived neurotrophic factor (BDNF) signaling after HI and nec-1 are also sex-specific providing differential conditions to promote recovery of those more severely injured. The increased aromatization of testosterone in male mice during early development and the link between 17-ß-estradiol (E2) levels and BDNF transcription substantiate this hypothesis. Hence, we aimed to investigate if sexual differences in BDNF signaling existed in forebrain and diencephalon after HI and HI/nec-1 and their correlation with estrogen receptors (ER). C57B6 mice (p7) received nec-1 (0.1µl [8µM]) or vehicle (veh) intracerebroventricularly after HI. At 24h after HI, BDNF levels increased in both sexes in forebrain without evidence of tropomyosin-receptor-kinase B (TrkB) activation. At 96h after HI, BDNF levels in forebrain decreased below those seen in control mice of both sexes. Additionally, only in female mice, truncated TrkB (Tc.TrkB) and p75 neurotrophic receptor (p75ntr) levels increased in forebrain and diencephalon. In both, forebrain and diencephalon, nec-1 treatment increased BDNF levels and TrkB activation in male mice while, nec-1 prevented Tc.TrkB and p75ntr increases in female mice. While E2 levels were unchanged by HI or HI/nec-1 in either sex or treatment, ERα:ERß ratios were increased in diencephalon of nec-1-treated male mice and directly correlated with BDNF levels. Neonatal HI produces sex-specific signaling changes in the BDNF system, that are differentially modulated by nec-1. The regional differences in BDNF levels may be a consequence of injury severity after HI, but sexual differences in response to nec-1 after HI may represent a differential thalamo-cortical preservation or alternatively off-target regional effect of nec-1. The biological significance of ERα predominance and its correlation with BDNF levels is still unclear.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Diencephalon/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Imidazoles/therapeutic use , Indoles/therapeutic use , Neuroprotective Agents/therapeutic use , Prosencephalon/metabolism , Animals , Animals, Newborn , Diencephalon/drug effects , Estradiol/analysis , Female , Male , Mice , Mice, Inbred C57BL , Parvalbumins/metabolism , Prosencephalon/drug effects , Receptors, Estrogen/metabolism , Sex Factors
19.
Physiol Behav ; 120: 40-5, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23816984

ABSTRACT

We have recently identified prolactin (PRL)-releasing peptides (PrRPs) and their stimulating effects on feeding behavior in chicks. To investigate further metabolic functions of PrRP, the present study was performed to clarify whether intracerebroventricular (ICV) injection of PrRP31, an active form of PrRP in chicks, affects heat production (HP), respiratory quotient (RQ) and plasma concentrations of metabolic fuels in chicks. The ICV injection of PrRP31 (94 and 375 pmol) did not affect HP but significantly lowered RQ. The change in RQ implies that PrRP31 shifted the utility of metabolic fuels in the body. This idea was confirmed by subsequent results in which ICV injection of PrRP31 significantly reduced glucose but increased non-esterified fatty acid concentrations in plasma. These shifts in blood metabolic fuels would not be through the increased plasma insulin, because the ICV injection of PrRP31 significantly decreased plasma insulin concentration. On the other hand, ICV injection of another orexigenic peptide, neuropeptide Y (NPY) also induced the insulin release and the metabolic effects were similar to those of PrRP31. Because ICV injection of PrRP31 increased NPY mRNA in the diencephalon, the NPY may mediate the metabolic functions of PrRP31. In summary, the present study suggests that central PrRP31 shifts the utilities of peripheral energy sources, which is not via hyperinsulinemia but via the diencephalon.


Subject(s)
Carbohydrate Metabolism/drug effects , Chickens/physiology , Lipid Metabolism/drug effects , Prolactin-Releasing Hormone/pharmacology , Animals , Animals, Newborn , Blood Glucose/metabolism , Carbon Dioxide/metabolism , Diencephalon/drug effects , Diencephalon/metabolism , Fatty Acids, Nonesterified/blood , Injections, Intraventricular , Insulin/blood , Male , Neuropeptide Y/biosynthesis , Neuropeptide Y/pharmacology , Oxygen Consumption/drug effects , Prolactin-Releasing Hormone/administration & dosage , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Thermogenesis/drug effects , Triglycerides/blood
20.
Article in Russian | MEDLINE | ID: mdl-23887452

ABSTRACT

We studied an effect of original domestic derivatives of 3-oxypyridine and succinic acid (emoxipine, reamberin and mexidol) on changes in the cellular composition of cortical and diencephalic structures in the rat brain in relation to the hyperglycemia severity in rats with alloxan diabetes. The effect of 3-oxypyridine and succinic acid derivatives was compared with the results of alpha-lipoic acid treatment. We determined that administration of 14 optimal doses of any medication in this study prevented the decrease in neuronal count in the primary somatosensory cortex (Par1 field). This effect was particularly evident after treatment with 3-oxypyridine derivatives (emoxipine and mexidol). Additionally, a two-week administration of emoxipine and mexidol led to a decreased percentage of lipofuscin-positive neurons in the neocortex and field CA1 of the hippocampus. Concurrently, emoxipine and mexidol increased the number of basket neurons as well as oligodendrocytes and microglia in the studied structure of the Ammon's horn. Moreover, these two substances prevented the decrease in the number of astrocytes in the somatosensory cortex and the paraventricular nucleus of the hypothalamus. The cerebroprotective activity of reamberin and alpha-lipoic acid in alloxan diabetes was less effective than that of 3-oxypyridine derivatives. Also, reamberin and alpha-lipoic acid induced undesirable side-effects manifested in the decreased number of pyramid neurons in field CA1 in the hippocampus and the increased number of lipofuscin-positive neurons in the somatosensory cortex and the paraventricular nucleus of the hypothalamus. Changes in the cellular composition of cortical and diencephalic structures resulting from administration of medications used in this study did not depend on the ability of these substances to restrain the hyperglycemia in alloxan diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diencephalon/metabolism , Neocortex/metabolism , Pyridines/therapeutic use , Succinic Acid/therapeutic use , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diencephalon/drug effects , Dose-Response Relationship, Drug , Female , Lipid Peroxidation/drug effects , Male , Neocortex/drug effects , Neocortex/physiopathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pyridines/administration & dosage , Rats , Succinic Acid/administration & dosage , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...