Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Oxid Med Cell Longev ; 2021: 8874503, 2021.
Article in English | MEDLINE | ID: mdl-34055199

ABSTRACT

The marine horseshoe crab (Tachypleus tridentatus) has been considered as food and traditional medicine for many years. Kynurenic acid (KA) was isolated from horseshoe crab in this study for the first time in the world. A previous study in 2018 reported that intraperitoneal administration of KA prevented high-fat diet- (HFD-) induced body weight gain. Now, we investigated the effects of intragastric gavage of KA on HFD mice and found that KA (5 mg/kg/day) inhibited both the body weight gain and the increase of average daily energy intake. KA reduced serum triglyceride and increased serum high-density lipoprotein cholesterol. KA inhibited HFD-induced the increases of serum low-density lipoprotein cholesterol, coronary artery risk index, and atherosclerosis index. KA also suppressed HFD-induced the increase of the ratio of Firmicutes to Bacteroidetes (two dominant gut microbial phyla). KA partially reversed HFD-induced the changes in the composition of gut microbial genera. These overall effects of KA on HFD mice were similar to that of simvastatin (positive control). But the effects of 1.25 mg/kg/day KA on HFD-caused hyperlipidemia were similar to the effects of 5 mg/kg/day simvastatin. The pattern of relative abundance in 40 key genera of gut microbiota from KA group was closer to that from the normal group than that from the simvastatin group. In addition, our in vitro results showed the potential antioxidant activity of KA, which suggests that the improvement effects of KA on HFD mice may be partially associated with antioxidant activity of KA. Our findings demonstrate the potential role of KA as a functional food ingredient for the treatment of obesity and hyperlipidemia as well as the modulation of gut microbiota.


Subject(s)
Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/drug therapy , Kynurenic Acid/therapeutic use , Obesity/drug therapy , Animals , Horseshoe Crabs , Kynurenic Acid/pharmacology , Male , Mice
2.
PLoS One ; 14(12): e0226205, 2019.
Article in English | MEDLINE | ID: mdl-31809528

ABSTRACT

Kynurenic acid (KYNA) is a neuroactive metabolite of tryptophan. KYNA naturally occurs in breast milk and its content increases with lactation, indicating the role of neonatal nutrition in general growth with long-term health effects. KYNA is also an antagonist of ionotropic glutamate receptors expressed in bone cells. The aim of this study was to establish the effects of chronic KYNA supplementation on bone homeostasis in young rats, using mandible as a model bone. Female and male newborn Wistar rats were divided into control and KYNA-administered groups until 60 days of age (25x101 mg/L or 25x102 mg/L in drinking water). Hemimandibles were subjected to densitometry, computed tomography analysis and mechanical testing. Rats supplemented with KYNA at both doses showed a decrease in body weight. There were no effects of KYNA administration and mandible histomorphometry. In males, a significant quadratic effect (P < 0.001) was observed in the densitometry of the hemimandible, where BMD increased in the group supplemented with 2.5x101 mg/L of KYNA. Analysis of mechanical tests data showed that when fracture forces were corrected for bone geometry and rats body weight the improvement of bone material properties was observed in male and female rats supplemented with lower dose of KYNA. This study showed that chronic supplementation with KYNA may limit weight gain in the young, without adversely affecting the development of the skeleton.


Subject(s)
Kynurenic Acid/administration & dosage , Mandible/physiology , Weight Loss/drug effects , Animals , Animals, Newborn , Biomechanical Phenomena/drug effects , Bone Density/drug effects , Case-Control Studies , Dietary Supplements , Female , Kynurenic Acid/pharmacology , Male , Mandible/drug effects , Rats , Rats, Wistar , Tomography, X-Ray Computed
3.
Elife ; 72018 05 31.
Article in English | MEDLINE | ID: mdl-29848447

ABSTRACT

Reward contingencies are dynamic: outcomes that were valued at one point may subsequently lose value. Action selection in the face of dynamic reward associations requires several cognitive processes: registering a change in value of the primary reinforcer, adjusting the value of secondary reinforcers to reflect the new value of the primary reinforcer, and guiding action selection to optimal choices. Flexible responding has been evaluated extensively using reinforcer devaluation tasks. Performance on this task relies upon amygdala, Areas 11 and 13 of orbitofrontal cortex (OFC), and mediodorsal thalamus (MD). Differential contributions of amygdala and Areas 11 and 13 of OFC to specific sub-processes have been established, but the role of MD in these sub-processes is unknown. Pharmacological inactivation of the macaque MD during specific phases of this task revealed that MD is required for reward valuation and action selection. This profile is unique, differing from both amygdala and subregions of the OFC.


Subject(s)
Behavior, Animal , Goals , Thalamus/physiology , Animals , Behavior, Animal/drug effects , Kynurenic Acid/pharmacology , Macaca , Male , Thalamus/drug effects , Time Factors
4.
Brain Res ; 1695: 53-64, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29800553

ABSTRACT

BACKGROUND: Pneumococcal meningitis is one of the most common infectious diseases with a high-mortality rate and long-term neurological sequelae, affecting up to 50% of survivors. Pneumococcal compounds are pro-inflammatory mediators that induce an innate immune response and tryptophan degradation through the kynurenine pathway. Vitamin B6 (vitB6) is an important vitamin which acts as a cofactor at the active sites of enzymes that catalyze a great number of reactions involved in the metabolism of tryptophan through the kynurenine pathway and may thus limit the accumulation of neurotoxic metabolites and preserve the cellular energy status. The aim of this study was to investigate the neuroprotective effect of adjuvant treatment with vitB6 in pneumococcal meningitis. METHODS: The effects of vitB6 on the clinical symptoms, the expression of kynureninase (KYN), Kynurenic acid (KYNA), nicotinamide adenine dinucleotide (NAD) and cytokines in brain tissue and memory of infant Wistar rats subjected to pneumococcal meningitis were researched. At the same time, Kynurenine 3-monooxygenase (KMO) inhibitor Ro 61-8048 was applied in order to further investigate the brain protective effect of vitB6 in bacterial meningitis. RESULTS: Adjuvant therapy of bacterial meningitis with vitB6 could improve the clinical symptoms, learning performance, lead to the maintenance of cellular NAD+ and ATP homeostasis and significantly down-regulate the levels of cytokines in the brain tissue by affecting the KYN pathway. CONCLUSIONS: Adjuvant treatment with vitB6 in pneumococcal meningitis could exert neuroprotective effect via increasing the preservation of cellular energy through affecting the KYN pathway and reducing of the inflammatory response.


Subject(s)
Brain/drug effects , Ceftriaxone/pharmacology , Memory/drug effects , Meningitis, Pneumococcal/drug therapy , Vitamin B 6/pharmacology , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Animals , Cytokines/metabolism , Kynurenic Acid/pharmacology , Meningitis, Pneumococcal/chemically induced , Rats, Wistar
5.
Food Chem Toxicol ; 109(Pt 1): 465-471, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28951307

ABSTRACT

High glucose and insulin lead to neuronal insulin resistance. Glucose transport into the neurons is achieved by regulatory induction of surface glucose transporter-3 (GLUT3) instead of the insulin. N-methyl-D aspartate (NMDA) receptor activity increases GLUT3 expression. This study explored whether an endogenous NMDA receptor antagonist, kynurenic acid (KynA) affects the neuronal cell viability at high glucose concentrations. SH-SY5Y neuroblastoma cells were exposed to 150-250 mg/dL glucose and 40 µU/mL insulin. In KynA and N-nitro-l-arginine methyl ester (L-NAME) supplemented cultures, oxidative stress, mitochondrial metabolic activity (MTT), nitric oxide as nitrite+nitrate (NOx) and GLUT3 were determined at the end of 24 and 48-h incubation periods. Viable cells were counted by trypan blue dye. High glucose-exposed SH-SY5Y cells showed two-times more GLUT3 expression at second 24-h period. While GLUT3-stimulated glucose transport and oxidative stress was increased, total mitochondrial metabolic activity was significantly reduced. Insulin supplementation to high glucose decreased NOx synthesis and GLUT3 levels, in contrast oxidative stress increased three-fold. KynA significantly reduced oxidative stress, and increased MTT by regulating NOx production and GLUT3 expression. KynA is a noteworthy compound, as an endogenous, specific NMDA receptor antagonist; it significantly reduces oxidative stress, while increasing cell viability at high glucose and insulin concentrations.


Subject(s)
Dopaminergic Neurons/metabolism , Glucose Transporter Type 3/metabolism , Glucose/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Biological Transport/drug effects , Cell Line , Cell Survival/drug effects , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Glucose Transporter Type 3/genetics , Humans , Insulin/metabolism , Kynurenic Acid/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics
6.
J Nat Med ; 70(1): 8-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26232896

ABSTRACT

The present study was designed to explore the possible effects of Pteris vittata on visual sensitivity, ERG waves, and other components of the visual system. Electrophysiological techniques including electroretinography (ERG) were used in the present study. The phytochemical composition of the extract was investigated using liquid chromatography-mass spectrometry (LC-MS) techniques. The results indicated that the extract significantly augmented dark- and light-adapted ERG b-wave amplitude. Furthermore, these findings showed that P. vittata extract does not have Gamma-aminobutyric acid receptor antagonistic activity but may function as a retinal neural antagonist in bullfrog retina. P. vittata extract improved the visual sensitivity by 0.8 log unit of light intensity, and reduced the regeneration time for rhodopsin. The six main peaks obtained through LC-MS were identified as flavonoids. Based on these results, it was concluded that P. vittata extract or its constituents may be used to treat eye diseases.


Subject(s)
Electroretinography/drug effects , Plant Extracts/pharmacology , Pteris/metabolism , Retina/metabolism , Rhodopsin/metabolism , Vision, Ocular/drug effects , Animals , Chick Embryo , Chromatography, Liquid , GABA Antagonists/pharmacology , Kynurenic Acid/pharmacology , Light , Mass Spectrometry , Phytochemicals , Picrotoxin/pharmacology , Rana catesbeiana , gamma-Aminobutyric Acid/metabolism
7.
Brain Res ; 1630: 73-82, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26562663

ABSTRACT

Previous studies showed that acute 7-chlorokynurenic acid treatment produced a rapid antidepressant-like action in depression-like animal models. However, the underlying mechanism involved in neurotrophin system about 7-chlorokynurenic acid is unclear. Our present study aimed to verify whether chronic 7-chlorokynurenic acid treatment produced an antidepressant-like effect through the activation of brain-derived neurotrophic factor (BDNF) signaling in mice exposed to chronic unpredictable mild stress (CUMS). In addition, we performed an oral toxicological evaluation of chronic 7-chlorokynurenic acid administration in mice. The results showed that a two-week administration with 7-chlorokynurenic acid reversed the decreased sucrose preference and prolonged first feeding latency. In addition, 7-chlorokynurenic acid significantly reversed the CUMS-induced down-regulation of BDNF, p-ERK, p-Akt, PSD-95, synapsin I and cell proliferation in the hippocampus. In contrast, K252a, an inhibitor of BDNF receptor tropomyosin-related kinase receptor B (TrkB), blocked the antidepressant-like effect and the improvement of 7-chlorokynurenic acid. Furthermore, we found that 7-chlorokynurenic acid did not produce any toxicological effect in mice. In conclusion, our findings suggest that the antidepressant-like effect of 7-chlorokynurenic acid may be mediated, at least in part, by activating BDNF signaling in the hippocampus.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/drug therapy , Hippocampus/drug effects , Kynurenic Acid/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Antidepressive Agents/toxicity , Depressive Disorder/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Hippocampus/metabolism , Hippocampus/pathology , Kidney/drug effects , Kidney/pathology , Kynurenic Acid/pharmacology , Kynurenic Acid/toxicity , Liver/drug effects , Liver/pathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred ICR , Random Allocation , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Uncertainty
8.
Epilepsia ; 54(7): 1251-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23663038

ABSTRACT

PURPOSE: Dravet syndrome (DS) is caused by dominant mutations of the SCN1A gene, encoding the NaV 1.1 sodium channel α subunit. Gene targeted mouse models of DS mutations replicate patients' phenotype and show reduced γ-aminobutyric acid (GABA)ergic inhibition. However, little is known on the properties of network hyperexcitability and on properties of seizure generation in these models. In fact, seizures have been studied thus far with surface electroencephalography (EEG), which did not show if specific brain regions are particularly involved. We have investigated hyperexcitability and epileptiform activities generated in neuronal networks of a mouse model of DS. METHODS: We have studied heterozygous NaV 1.1 knock-out mice performing field potential recordings in combined hippocampal/cortical slices in vitro and video/depth electrode intracerebral recordings in vivo during hyperthermia-induced seizures. KEY FINDINGS: In slices, we have disclosed specific signs of hyperexcitability of hippocampal circuits in both the pre-epileptic and epileptic periods, and a specific epileptiform activity was generated in the hippocampus upon application of the convulsant 4-aminopyridine in the epileptic period. During in vivo hyperthermia-induced seizures, we have observed selective hippocampal activity in early preictal phases and pronounced hippocampal activity in the ictal phase. SIGNIFICANCE: We have identified specific epileptiform activities and signs of network hyperexcitability, and disclosed the important role of the hippocampus in seizure generation in this model. These activities may be potentially used as targets for screenings of antiepileptic approaches.


Subject(s)
Epilepsies, Myoclonic/pathology , Epilepsies, Myoclonic/physiopathology , Hippocampus/physiopathology , 4-Aminopyridine/adverse effects , Age Factors , Animals , Animals, Newborn , Bicuculline/toxicity , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Electric Stimulation/adverse effects , Electroencephalography , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/genetics , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Antagonists/toxicity , Hippocampus/drug effects , Hyperthermia, Induced/adverse effects , In Vitro Techniques , Kynurenic Acid/pharmacology , Mice , Mice, Knockout , NAV1.1 Voltage-Gated Sodium Channel/deficiency , NAV1.1 Voltage-Gated Sodium Channel/genetics , Potassium Channel Blockers/adverse effects , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Pyramidal Cells/physiology
9.
J Neurosci ; 33(11): 4935-46, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486964

ABSTRACT

Associative stimulation causes enduring changes in the nervous system based on the Hebbian concept of spike-timing-dependent plasticity. The present study aimed to characterize the immediate and long-term electrophysiological effects of associative stimulation at the level of spinal cord and to test how trans-spinal direct current stimulation (tsDC) modulates associative plasticity. The effect of combined associative stimulation and tsDC on locomotor recovery was tested in a unilateral model of spinal cord injury (SCI). Two associative protocols were tested: (1) spino-sciatic associative (SSA) protocol, in which the first stimulus originated from the sciatic nerve and the second from the spinal cord; and (2) cortico-sciatic associative (CSA) protocol, in which the first stimulus originated from the sciatic nerve and the second from the motor cortex. In addition, those two protocols were repeated in combination with cathodal tsDC application. SSA and CSA stimulation produced immediate enhancement of spinal and cortical outputs, respectively, depending on the duration of the interstimulus interval. Repetitive SSA or CSA stimulation produced long-term potentiation of spinal and cortical outputs, respectively. Applying tsDC during SSA or CSA stimulation markedly enhanced their immediate and long-term effects. In behaving mice with unilateral SCI, four consecutive 20 min sessions of CSA + tsDC markedly reduced error rate in a horizontal ladder-walking test. Thus, this form of artificially enhanced associative connection can be translated into a form of motor relearning that does not depend on practice or experience.


Subject(s)
Cerebral Cortex/physiology , Electric Stimulation Therapy/methods , Recovery of Function/physiology , Sciatic Nerve/physiology , Spinal Cord Injuries/therapy , Spinal Cord/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Biophysics , Disease Models, Animal , Electrodes , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality/drug effects , Kynurenic Acid/pharmacology , Male , Mice , Neural Pathways/physiology , Neuronal Plasticity/physiology , Psychomotor Performance/drug effects , Reaction Time/drug effects , Spinal Cord/drug effects , Time Factors , Walking/physiology
10.
J Neurosci ; 32(31): 10699-712, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22855818

ABSTRACT

In the developing nervous system, spontaneous neuronal activity arises independently of experience or any environmental input. This activity may play a major role in axonal pathfinding, refinement of topographic maps, dendritic morphogenesis, and the segregation of axonal terminal arbors. In the auditory system, endogenously released ATP in the cochlea activates inner hair cells to trigger bursts of action potentials (APs), which are transferred to the central auditory system. Here we show the modulatory role of purinergic signaling beyond the cochlea, i.e., the developmentally regulated and cell-type-specific depolarizing effects on auditory brainstem neurons of Mongolian gerbil. We assessed the effects of P2X receptors (P2XRs) on neuronal excitability from prehearing to early stages of auditory signal processing. Our results demonstrate that in neurons expressing P2XRs, extracellular ATP can evoke APs in sync with Ca(2+) signals. In cochlear nucleus (CN) bushy cells, ATP increases spontaneous and also acoustically evoked activity in vivo, but these effects diminish with maturity. Moreover, ATP not only augmented glutamate-driven firing, but it also evoked APs in the absence of glutamatergic transmission. In vivo recordings also revealed that endogenously released ATP in the CN contributes to neuronal firing activity by facilitating AP generation and prolonging AP duration. Given the enhancing effect of ATP on AP firing and confinement of P2XRs to certain auditory brainstem nuclei, and to distinct neurons within these nuclei, it is conceivable that purinergic signaling plays a specific role in the development of neuronal brainstem circuits.


Subject(s)
Action Potentials/physiology , Brain Stem/cytology , Brain Stem/growth & development , Receptors, Purinergic P2X/metabolism , Sensory Receptor Cells/physiology , Acoustic Stimulation , Action Potentials/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Female , Gerbillinae , Glycine/pharmacology , In Vitro Techniques , Kynurenic Acid/pharmacology , Male , Patch-Clamp Techniques , Psychoacoustics , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Stilbamidines/metabolism , Synaptic Potentials/drug effects
11.
Auton Neurosci ; 169(2): 124-34, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22748567

ABSTRACT

To characterize the possible role of glutamate in the interaction between Hypothalamic Defense Area (HDA) and Parabrachial complex (PBc) nuclei, cardiorespiratory changes were analyzed in response to electrical stimulation of the HDA (1 ms pulses, 30-50 µA given at 100 Hz for 5s) before and after the microinjection of the nonspecific glutamate receptor antagonist kynurenic acid (50 nl, 5 nmol), NMDA receptor antagonist MK-801 (50 nl, 50 nmol), non-NMDA receptor antagonist CNQX (50 nl, 50 nmol) or metabotropic glutamate receptor antagonist MCPG (50 nl, 5 nmol) within the PBc. HDA stimulation evoked an inspiratory facilitatory response, consisting of an increase in respiratory rate (p<0.001) due to a decrease in expiratory time (p<0.01). The respiratory response was accompanied by a pressor (p<0.001) and a tachycardic response (p<0.001). Kynurenic acid within the lateral parabrachial region (lPB) abolished the tachycardia (p<0.001) and decreased the magnitude of blood pressure response (p<0.001) to HDA stimulation. Similarly, the magnitude of the tachycardia and the pressor response was decreased after the microinjection of MK-801 (p<0.01 and p<0.001, respectively) and CNQX (p<0.05 in both cases) into the lPB. Kynurenic acid microinjection in this region produced an inhibition of the tachypnea (p<0.001) to HDA stimulation but the respiratory response persisted unchanged after MK-801 or CNQX microinjection into the lPB. Kynurenic acid within the medial parabrachial region (mPB) abolished the tachycardia (p<0.01) and decreased the magnitude of the pressor response (p<0.001) to HDA stimulation. MK-801 and CNQX microinjection in this region decreased the magnitude of the tachycardia (p<0.05, in both cases) and pressor response (p<0.05, in both cases). The respiratory response evoked by HDA stimulation was not changed after the microinjection of kynurenic acid, MK-801 or CNQX within the mPB. No changes were observed in the cardiorespiratory response evoked to HDA stimulation after MCPG microinjection within lPB and mPB. These results indicate that glutamate PBc receptors are involved in the cardiorespiratory response evoked from the HDA. The possible mechanisms involved in these interactions are discussed.


Subject(s)
6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Dizocilpine Maleate/pharmacology , Hypothalamus/metabolism , Receptors, Glutamate/metabolism , Animals , Blood Pressure/physiology , Cardiovascular Physiological Phenomena , Electric Stimulation , Heart Rate/drug effects , Heart Rate/physiology , Hypothalamus/drug effects , Hypothalamus/physiology , Kynurenic Acid/pharmacology , Male , Microinjections , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Respiration/drug effects , Tachycardia/physiopathology
12.
J Pharmacol Sci ; 118(3): 382-90, 2012.
Article in English | MEDLINE | ID: mdl-22450196

ABSTRACT

Microinjection of the α(2)-adrenoceptor agonist clonidine into the hypothalamic periventricular nuclei (PVN) induces the pressor response associated with bradycardia in freely-moving conscious rats. This study investigated the involvement of γ-aminobutyric acid nerves (GABAergic nerves) and glutamatergic nerves in the cardiovascular response to microinjection of clonidine in the PVN. Male Wistar rats were chronically implanted with a microinjection cannula into the PVN and an arterial catheter into the abdominal aorta through the femoral artery. Blood pressure and heart rate were measured under a conscious unrestrained state. PVN injection of clonidine induced a dose-dependent pressor response concomitant with bradycardia. PVN pretreatment with GABA, muscimol (GABA(A)-receptor agonist), or bicuculline (GABA(A)-receptor antagonist) significantly inhibited the pressor response to PVN-injected clonidine without affecting bradycardia. PVN pretreatment with baclofen (GABA(B)-receptor agonist), 2-hydroxysaclofen (GABA(B)-receptor antagonist), or kynurenic acid (non-selective NMDA-type glutamate-receptor and ionotropic glutamate-receptor antagonist) did not affect the pressor response to PVN-injected clonidine. These results suggest that clonidine induces a pressor response by stimulating the presynaptic α(2)-adrenoceptor of GABAergic nerves in the PVN, thereby inhibiting GABAergic nerve activity.


Subject(s)
Antihypertensive Agents/pharmacology , Clonidine/pharmacology , GABAergic Neurons/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Baclofen/analogs & derivatives , Baclofen/pharmacology , Bicuculline/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Excitatory Amino Acid Antagonists/pharmacology , GABA Agents/pharmacology , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , GABA-B Receptor Agonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , GABAergic Neurons/physiology , Heart Rate/drug effects , Hypothalamus , Kynurenic Acid/pharmacology , Male , Muscimol/pharmacology , Paraventricular Hypothalamic Nucleus/physiology , Rats , Rats, Wistar , Receptors, Glutamate/physiology , gamma-Aminobutyric Acid/pharmacology
13.
J Neurosci ; 31(38): 13644-53, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21940455

ABSTRACT

Prepulse inhibition (PPI) of startle is the suppression of the startle reflex when a weaker sensory stimulus (the prepulse) shortly precedes the startling stimulus. PPI can be attentionally enhanced in both humans and laboratory animals. This study investigated whether the following three forebrain structures, which are critical for initial cortical processing of auditory signals, auditory fear conditioning/memories, and spatial attention, respectively, play a role in the top-down modulation of PPI in rats: the primary auditory cortex (A1), lateral nucleus of the amygdala (LA), and posterior parietal cortex (PPC). The results show that, under the noise-masking condition, PPI was enhanced by fear conditioning of the prepulse in a prepulse-specific manner, and the conditioning-induced PPI enhancement was further increased by perceptual separation between the conditioned prepulse and the noise masker. Reversibly blocking glutamate receptors in the A1 with 2 mm kynurenic acid eliminated both the conditioning-induced and perceptual separation-induced PPI enhancements. Blocking the LA eliminated the conditioning-induced but not the perceptual separation-induced PPI enhancement, and blocking the PPC specifically eliminated the perceptual separation-induced PPI enhancement. The two types of PPI enhancements were also eliminated by the extinction manipulation. Thus, the top-down modulation of PPI is differentially organized and depends on operations of various forebrain structures. Due to the fine-tuned modulation by higher-order cognitive processes, functions of PPI can be more flexible to complex environments. The top-down enhancements of PPI in rats are also useful for modeling some mental disorders, such as schizophrenia, attention deficit/hyperactivity disorder, and posttraumatic stress disorder.


Subject(s)
Amygdala/physiology , Auditory Cortex/physiology , Neural Inhibition/physiology , Parietal Lobe/physiology , Reflex, Startle/physiology , Acoustic Stimulation/methods , Amygdala/drug effects , Animals , Attention/physiology , Auditory Cortex/drug effects , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/physiology , Fear/physiology , Kynurenic Acid/pharmacology , Male , Neural Inhibition/drug effects , Parietal Lobe/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects
14.
Arch Pharm (Weinheim) ; 343(10): 561-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20938950

ABSTRACT

The synthesis and the pharmacological activity of a series of 1-aroyl derivatives of kynurenic acid methyl ester (4-oxo-quinolin-2-carboxy methyl (KYNA) esters), structurally related to NSAID indomethacin are described. The derivatives were screened in vivo for anti-inflammatory and analgesic activities. Most of the compounds exhibited good anti-inflammatory and analgesic activities. An automatic docking of the synthesized compounds was performed using X-ray structures of COX-1 and COX-2. Docking results are in good accordance with the experimental biological data.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/chemical synthesis , Kynurenic Acid/pharmacology , Analgesics/chemistry , Analgesics/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Carrageenan/chemistry , Carrageenan/metabolism , Cyclooxygenase 1/pharmacology , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/pharmacology , Drug Evaluation, Preclinical , Indomethacin/pharmacology , Kynurenic Acid/chemistry , Models, Molecular , Rats , Structure-Activity Relationship
15.
Neurogastroenterol Motil ; 22(2): 217-25, e68, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19735360

ABSTRACT

BACKGROUND: Inflammatory bowel diseases are accompanied by severe motility disorders. The aim of our study was to investigate whether the blockade of peripheral N-methyl-D-aspartate (NMDA)-sensitive glutamate receptors (NMDA-Rs) alters motility changes in chemically induced acute colitis and how this modulation is accomplished. METHODS: The inflammatory and motility changes in 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis were studied in anaesthetized Wistar rats following treatment with the natural NMDA-R antagonist kynurenic acid (KynA) or SZR-72, a blood-brain barrier-permeable synthetic KynA analogue. The macrohaemodynamics, serosal microcirculation (visualized by intravital videomicroscopy), plasma levels of tumour necrosis factor alpha (TNF-alpha), inflammatory enzyme activities (xanthine oxidoreductase (XOR), myeloperoxidase (MPO) and nitric oxide synthase (NOS)), and colonic motility (with a strain-gauge technique) were evaluated 17 h after colitis induction and compared with the control conditions. KEY RESULTS: The TNBS enema induced a systemic hyperdynamic circulatory reaction, increased the serosal capillary blood flow, significantly elevated the mucosal XOR, MPO and NOS activities and augmented the colonic motility relative to the controls. The NMDA-R antagonist treatment with KynA or SZR-72 significantly reduced the XOR, NOS and MPO activities, decreased the motility and increased the tone of the colon. CONCLUSIONS & INFERENCES: These data demonstrate a potential modulatory mechanism of NMDA-R in altered colonic motility in TNBS colitis. Inhibition of the enteric NMDA-Rs may provide a therapeutic option via which to influence intestinal hypermotility, microcirculatory changes and inflammatory activation simultaneously.


Subject(s)
Colitis/physiopathology , Colon/physiopathology , Gastrointestinal Motility/drug effects , Inflammation/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Analysis of Variance , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Colitis/chemically induced , Colitis/metabolism , Colon/drug effects , Colon/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Gastrointestinal Motility/physiology , Inflammation/physiopathology , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/pharmacology , Male , Nitric Oxide Synthase/metabolism , Peroxidase/metabolism , Random Allocation , Rats , Rats, Wistar , Time Factors , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/blood , Xanthine Oxidase/metabolism
16.
J Neurophysiol ; 101(3): 1647-59, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19036862

ABSTRACT

Survival in natural environments for small animals such as rats often depends on precise neural coding of life-threatening acoustic signals, and binaural unmasking of species-specific pain calls is especially critical. This study investigated how species-specific tail-pain chatter is represented in the rat amygdala, which receives afferents from both auditory thalamus and auditory association cortex, and whether the amygdaloid representation of the chatter can be binaurally unmasked. The results show that chatter with a fundamental frequency (F0) of 2.1 kHz was able to elicit salient phase-locked frequency-following responses (FFRs) in the lateral amygdala nucleus in anesthetized rats. FFRs to the F0 of binaurally presented chatter were sensitive to the interaural time difference (ITD), with the preference of ipsilateral-ear leading, as well as showing features of binaural inhibition. When interaurally correlated masking noises were added and ipsilateral chatter led contralateral chatter, introducing an ITD disparity between the chatter and masker significantly enhanced (unmasked) the FFRs. This binaural unmasking was further enhanced by chemically blocking excitatory glutamate receptors in the auditory association cortex. When the chatter was replaced by a harmonic tone complex with an F0 of 0.7 kHz, both the binaural-inhibition feature and the binaural unmasking were preserved only for the harmonic of 2.1 kHz but not the tone F0. These results suggest that both frequency-dependent ascending binaural modulations and cortical descending modulations of the precise auditory coding of the chatter in the amygdala are critical for processing life-threatening acoustic signals in noisy and even reverberant environments.


Subject(s)
Amygdala/physiology , Auditory Pathways/physiology , Auditory Perception/physiology , Functional Laterality/physiology , Perceptual Masking/physiology , Psychoacoustics , Acoustic Stimulation/methods , Amygdala/drug effects , Analysis of Variance , Animals , Evoked Potentials, Auditory/physiology , Excitatory Amino Acid Antagonists/pharmacology , Kynurenic Acid/pharmacology , Male , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Spectrum Analysis
17.
J Neurosci ; 28(22): 5691-5, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18509030

ABSTRACT

Retinal horizontal cells (HCs) provide negative feedback to cones, but, largely because annular illumination fails to evoke a depolarizing response in rods, it is widely believed that there is no feedback from HCs to rods. However, feedback from HCs to cones involves small changes in the calcium current (I(Ca)) that do not always generate detectable depolarizing responses. We therefore recorded I(Ca) directly from rods to test whether they were modulated by feedback from HCs. To circumvent problems presented by overlapping receptive fields of HCs and rods, we manipulated the membrane potential of voltage-clamped HCs while simultaneously recording from rods in a salamander retinal slice preparation. Like HC feedback in cones, hyperpolarizing HCs from -14 to -54, -84, and -104 mV increased the amplitude of I(Ca) recorded from synaptically connected rods and caused hyperpolarizing shifts in I(Ca) voltage dependence. These effects were blocked by supplementing the bicarbonate-buffered saline solution with HEPES. In rods lacking light-responsive outer segments, hyperpolarizing neighboring HCs with light caused a negative activation shift and increased the amplitude of I(Ca). These changes in I(Ca) were blocked by HEPES and by inhibiting HC light responses with a glutamate antagonist, indicating that they were caused by HC feedback. These results show that rods, like cones, receive negative feedback from HCs that regulates the amplitude and voltage dependence of I(Ca). HC-to-rod feedback counters light-evoked decreases in synaptic output and thus shapes the transmission of rod responses to downstream visual neurons.


Subject(s)
Feedback/physiology , Retina/cytology , Retinal Horizontal Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Animals , Computer Simulation , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , HEPES/pharmacology , In Vitro Techniques , Kynurenic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Models, Neurological , Patch-Clamp Techniques , Photic Stimulation/methods , Urodela
18.
J Pharmacol Exp Ther ; 324(1): 342-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17940199

ABSTRACT

GPR35 is a G protein-coupled receptor recently "de-orphanized" using high-throughput intracellular calcium measurements in clonal cell lines expressing a chimeric G-protein alpha-subunit. From these screens, kynurenic acid, an endogenous metabolite of tryptophan, and zaprinast, a synthetic inhibitor of cyclic guanosine monophosphate-specific phosphodiesterase, emerged as potential agonists for GPR35. To investigate the coupling of GPR35 to natively expressed neuronal signaling pathways and effectors, we heterologously expressed GPR35 in rat sympathetic neurons and examined the modulation of N-type (Ca(V)2.2) calcium channels. In neurons expressing GPR35, calcium channels were inhibited in the absence of overt agonists, indicating a tonic receptor activity. Application of kynurenic acid or zaprinast resulted in robust voltage-dependent calcium current inhibition characteristic of Gbetagamma-mediated modulation. Both agonist-independent and -dependent effects of GPR35 were blocked by Bordetella pertussis toxin pretreatment indicating the involvement of G(i/o) proteins. In neurons expressing GPR35a, a short splice variant of GPR35, zaprinast was more potent (EC(50) = 1 microM) than kynurenic acid (58 microM) but had a similar efficacy (approximately 60% maximal calcium current inhibition). Expression of GPR35b, which has an additional 31 residues at the N terminus, produced similar results but with much greater variability. Both GPR35a and GPR35b appeared to have similar expression patterns when fused to fluorescent proteins. These results suggest a potential role for GPR35 in regulating neuronal excitability and synaptic release.


Subject(s)
Calcium Channels, N-Type/physiology , Neurons/physiology , Receptors, G-Protein-Coupled/physiology , Superior Cervical Ganglion/physiology , Animals , Calcium Channel Blockers/pharmacology , DNA, Complementary/genetics , HeLa Cells , Humans , Kynurenic Acid/pharmacology , Male , Pertussis Toxin , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/physiology , Purinones/pharmacology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics
19.
J Physiol ; 586(1): 227-45, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17962328

ABSTRACT

Heteromeric NMDARs are composed of coagonist glycine-binding NR1 subunits and glutamate-binding NR2 subunits. The majority of functional NMDARs in the mammalian central nervous system (CNS) contain two NR1 subunits and two NR2 subunits of which there are four types (A-D). We show that the potency of a variety of endogenous and synthetic glycine-site coagonists varies between recombinant NMDARs such that the highest potency is seen at NR2D-containing and the lowest at NR2A-containing NMDARs. This heterogeneity is specified by the particular NR2 subunit within the NMDAR complex since the glycine-binding NR1 subunit is common to all NMDARs investigated. To identify the molecular determinants responsible for this heterogeneity, we generated chimeric NR2A/2D subunits where we exchanged the S1 and S2 regions that form the ligand-binding domains and coexpressed these with NR1 subunits in Xenopus laevis oocytes. Glycine concentration-response curves for NMDARs containing NR2A subunits including the NR2D S1 region gave mean glycine EC(50) values similar to NR2A(WT)-containing NMDARs. However, receptors containing NR2A subunits including the NR2D S2 region or both NR2D S1 and S2 regions gave glycine potencies similar to those seen in NR2D(WT)-containing NMDARs. In particular, two residues in the S2 region of the NR2A subunit (Lys719 and Tyr735) when mutated to the corresponding residues found in the NR2D subunit influence glycine potency. We conclude that the variation in glycine potency is caused by interactions between the NR1 and NR2 ligand-binding domains that occur following agonist binding and which may be involved in the initial conformation changes that determine channel gating.


Subject(s)
Glycine/pharmacology , Oocytes/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Amino Acid Sequence , Animals , Chimera , Dose-Response Relationship, Drug , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Female , Glycine/antagonists & inhibitors , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/pharmacology , Molecular Sequence Data , Oocytes/drug effects , Patch-Clamp Techniques , RNA, Complementary/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/drug effects , Xenopus laevis
20.
Am J Physiol Heart Circ Physiol ; 293(6): H3627-35, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17890425

ABSTRACT

Electroacupuncture (EA) causes prolonged suppression of reflex elevations in blood pressure for 1-2 h in anesthetized preparations. A long-loop pathway involving the arcuate nucleus (ARC), ventrolateral periaqueductal gray, and rostral ventrolateral medulla (rVLM) is involved in sympathoinhibitory cardiovascular EA effects. However, the mechanisms and locations of the prolonged EA inhibition are unknown. We hypothesized that this effect is mediated through a long-loop pathway involving opioid, nociceptin, and gamma-aminobutyric acid (GABA) receptor activation in the rVLM. In anesthetized, ventilated cats application of bradykinin to the gallbladder every 10 min induced consistent reflex increases in blood pressure. Bilateral EA stimulation at the cardiovascular acupoints P5-6 overlying the median nerves reduced the reflex responses for at least 80 min. Bilateral blockade with kynurenic acid in the ARC 60 min after onset of EA inhibition reversed the cardiovascular response, suggesting a role for the ARC in the long-loop pathway during the prolonged inhibitory response. Unilateral microinjection with either an opioid or a GABA(A) antagonist in rVLM 50-60 min after the beginning of the EA response reversed EA inhibition of the cardiovascular excitatory reflex. Gabazine also reversed EA inhibition of cardiovascular premotor sympathetic rVLM neurons. Conversely, microinjection of a nociceptin/orphanin FQ peptide antagonist did not affect the prolonged inhibitory effect. Thus the ARC, an important component in the long-loop pathway in the EA cardiovascular response, is required for prolonged suppression of reflex cardiovascular excitatory responses by EA. Furthermore, in the rVLM, opioids and GABA, but not nociceptin, participate in the long-term EA-related inhibition of sympathoexcitatory cardiovascular responses.


Subject(s)
Baroreflex , Cardiovascular System/innervation , Electroacupuncture , Medulla Oblongata/metabolism , Neural Inhibition , Opioid Peptides/metabolism , Sympathetic Nervous System/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Baroreflex/drug effects , Blood Pressure , Bradykinin/metabolism , Cats , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Gallbladder/innervation , Kynurenic Acid/pharmacology , Median Nerve/physiology , Medulla Oblongata/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neural Inhibition/drug effects , Neural Pathways/metabolism , Peptides/pharmacology , Pyridazines/pharmacology , Receptors, GABA-A/metabolism , Receptors, Opioid/metabolism , Sympathetic Nervous System/drug effects , Time Factors , Nociceptin Receptor , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL