Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 168
1.
J Pharm Pharmacol ; 74(1): 22-31, 2022 Jan 05.
Article En | MEDLINE | ID: mdl-34586411

OBJECTIVES: 6-Hydroxykynurenic acid (6-HKA) is an organic acid component in extracts of Ginkgo biloba leaves and acts as a major contributor to neurorestorative effects, while its oral bioavailability was low. Therefore, using prodrug method to improve the bioavailability and brain content of 6-HKA is significant. METHODS: Three structural modified compounds of 6-HKA were synthesized, and ultra performance liquid chromatography-tandem mass spectrometry methods for quantification of these structural modified compounds in rat plasma and rat brain homogenate were established and comprehensively validated. The methods were effectively applied to investigate the effects of structural modification on apparent permeability coefficients in cells, the pharmacokinetics and the brain distribution in rats. KEY FINDINGS: The results illustrated that esterification can greatly improve the apparent permeability coefficient and bioavailability of 6-HKA. Comparing with direct oral administration of 6-HKA, the bioavailability of isopropyl ester was greatly improved (from 3.96 ± 1.45% to 41.8 ± 15.3%), and the contents of 6-HKA in rat brains (49.7 ± 9.2 ng/g brain) were significantly higher after oral administration. CONCLUSIONS: The bioavailability and the brain content of 6-HKA can be improved by the prodrug method. Among three structural modified compounds, isopropyl-esterified 6-HKA was the most promising treatment.


Biological Availability , Brain , Ginkgo biloba , Kynurenic Acid/analogs & derivatives , Administration, Oral , Animals , Brain/metabolism , Brain/pathology , Chromatography, Liquid/methods , Kynurenic Acid/administration & dosage , Kynurenic Acid/pharmacokinetics , Plant Extracts/administration & dosage , Plant Extracts/pharmacokinetics , Plant Preparations/administration & dosage , Plant Preparations/pharmacokinetics , Prodrugs/pharmacology , Rats , Structure-Activity Relationship , Tandem Mass Spectrometry/methods , Tissue Distribution
2.
Cereb Cortex ; 31(4): 2251-2267, 2021 03 05.
Article En | MEDLINE | ID: mdl-33270817

The Hamilton Search Task (HST) is a test of nonnavigational spatial memory that is dependent on the hippocampus. The parahippocampal cortex (PHC) is a major route for spatial information to reach the hippocampus, but the extent to which the PHC and hippocampus function independently of one another in the context of nonnavigational spatial memory is unclear. Here, we tested the hypotheses that (1) bilateral pharmacological inactivation of the PHC would impair HST performance, and (2) that functional disconnection of the PHC and hippocampus by contralateral (crossed) inactivation would likewise impair performance. Transient inactivation of the PHC impaired HST performance most robustly with 30 s intertrial delays, but not when color cues were introduced. Functional disconnection of the PHC and hippocampus, but not separate unilateral inactivation of either region, also selectively impaired long-term spatial memory. These findings indicate a critical role for the PHC and its interactions with the hippocampus in nonnavigational spatial memory.


Hippocampus/physiology , Parahippocampal Gyrus/physiology , Spatial Memory/physiology , Spatial Navigation , Animals , Excitatory Amino Acid Antagonists/administration & dosage , Hippocampus/drug effects , Kynurenic Acid/administration & dosage , Macaca mulatta , Male , Microinjections , Neural Pathways/drug effects , Neural Pathways/physiology , Parahippocampal Gyrus/drug effects , Spatial Memory/drug effects
3.
Mol Cell Endocrinol ; 518: 110928, 2020 12 01.
Article En | MEDLINE | ID: mdl-32702471

Kynurenic acid (KA), an endogenous product of L-tryptophan metabolism in the kynurenine pathway, regulates adipose tissue energy homeostasis and inflammation. However, its role in palmitate-induced insulin resistance and detailed underlying mechanisms in skeletal muscles and adipose tissues are unclear. Herein, we report that KA ameliorated palmitate-induced inflammation and insulin resistance in differentiated C2C12 and 3T3-L1 cell lines as well as soleus skeletal muscle and subcutaneous adipose tissues in mice. Palmitate-induced inflammatory markers, such as nuclear factor κB translocation, inhibitory κBα phosphorylation, pro-inflammatory cytokine expression, and impaired insulin signaling, were markedly attenuated by KA both in vitro and in vivo. KA significantly increased AMP-activated protein kinase (AMPK) phosphorylation and sirtuin 6 (SIRT6) expressions in C2C12 myocytes and 3T3-L1 adipocytes and skeletal muscle and adipose tissues of mice. siRNA-mediated AMPK or SIRT6 inhibition significantly mitigated the suppressive effects of KA on palmitate-induced inflammation and insulin resistance. KA significantly stimulated expression of genes involved in fatty acid oxidation in C2C12 myocytes and skeletal muscle of mice. Moreover, KA inhibits lipogenesis in 3T3-L1 adipocytes. AMPK or SIRT6 siRNA markedly reversed these changes. The siRNA targeting Gpr35 abrogated the effects of KA on AMPK phosphorylation in C2C12 myocytes and 3T3-L1 adipocytes, except SIRT6 expression. It has therefore been shown that KA could potentially alleviate inflammation and insulin resistance in skeletal muscle and adipose tissues through Gpr35/AMPK and SIRT6-mediated pathways.


Adipocytes/drug effects , Hyperlipidemias/drug therapy , Inflammation/prevention & control , Insulin Resistance , Kynurenic Acid/pharmacology , Muscle, Skeletal/drug effects , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Animals , Cells, Cultured , Diet, High-Fat , Hyperlipidemias/complications , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Inflammation/etiology , Kynurenic Acid/administration & dosage , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Signal Transduction/drug effects
4.
Neurochem Int ; 138: 104752, 2020 09.
Article En | MEDLINE | ID: mdl-32445659

Kynurenic acid (KYNA) is one of the most significant metabolite of the kynurenine pathway both in terms of functional and potential therapeutic value. It is an N-methyl-D-aspartate (NMDA) receptor antagonist, but it can also activate the G-protein coupled receptor 35 (GPR35), which shares several structural and functional properties with cannabinoid receptors. Previously our group demonstrated that systemic chronic KYNA treatment altered opioid receptor G-protein activity. Opioid receptors also overlap in many features with cannabinoid receptors. Thus, our aim was to examine the direct in vitro and systemic, chronic in vivo effect of KYNA on type 1 cannabinoid receptor (CB1R) binding and G-protein activity. Based on competition and [35S]GTPγS G-protein binding assays in rat brain, KYNA alone did not show significant binding towards the CB1R, nor did it alter CB1R ligand binding and agonist activity in vitro. When rats were chronically treated with KYNA (single daily, i.p., 128 mg/kg for 9 days), the KYNA plasma and cerebrospinal fluid levels significantly increased compared to vehicle treated group. Furthermore, in G-protein binding assays, in the whole brain the amount of G-proteins in basal and in maximum activity coupled to the CB1R also increased due to the treatment. At the same time, the overall stimulatory properties of the receptor remained unaltered in vehicle and KYNA treated samples. Similar observations were made in rat hippocampus, but not in the cortex and brainstem. In saturation binding assays the density of CB1Rs in rat whole brain and hippocampus were also significantly enhanced after the same treatment, without significantly affecting ligand binding affinity. Thus, KYNA indirectly and brain region specifically increases the abundance of functional CB1Rs, without modifying the overall binding and activity of the receptor. Supposedly, this can be a compensatory mechanism on the part of the endocannabinoid system induced by the long-term KYNA exposure.


Brain/drug effects , Brain/metabolism , Kynurenic Acid/administration & dosage , Kynurenic Acid/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Benzoxazines/metabolism , Benzoxazines/pharmacology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Male , Morpholines/metabolism , Morpholines/pharmacology , Naphthalenes/metabolism , Naphthalenes/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
5.
Neurotox Res ; 38(1): 50-58, 2020 Jun.
Article En | MEDLINE | ID: mdl-32219734

Kynurenic acid (KYNA) and quinolinic acid (QUIN) are metabolites formed in the degradation of tryptophan (Trp). QUIN is a selective NMDA receptor antagonist and may exert neurotoxic effects, whereas KYNA is an agonist of glutamatergic and cholinergic receptors and presents antioxidant properties. KYNA/QUIN ratio is decreased in several central nervous system disorders, but the mechanisms involved are not well elucidated. In the present study, we try to determine the neuroprotective capacity of KYNA on the QUIN effects in redox homeostasis changes (H2DCF oxidation, superoxide dismutase/catalase (SOD/CAT) ratio, glutathione peroxidase (GPx) activity, sulfhydryl content, and nitrite levels), as well as on inflammatory parameters (levels of TNF-α, IL-1ß, and IL-6). KYNA and QUIN effects on the activities of Na+,K+-ATPase and acetylcholinesterase (AChE) were also evaluated. Thirty-day-old male Wistar rats underwent stereotactic surgery and received intrastriatal injections as follows: group 1-control (PBS-injected), group 2-KYNA (100 µM), group 3-QUIN (150 nM), and group 4-KYNA + QUIN (KYNA-injected followed QUIN-injected). Results demonstrated that the KYNA administration was able to prevent the increase in reactive oxygen species, SOD/CAT ratio, and pro-inflammatory cytokines (IL-1ß and IL-6) and the decrease in GPx activity, sulfhydryl content, and nitrite levels caused by QUIN. KYNA was also able to partially prevent the decrease in Na+,K+-ATPase activity and the increase in AChE activity caused by QUIN. This study may help in the elucidation of neuroprotective effects of KYNA against oxidative and inflammatory insults caused by QUIN in the striatum of young male Wistar rats.


Corpus Striatum/drug effects , Corpus Striatum/metabolism , Encephalitis/metabolism , Homeostasis/drug effects , Kynurenic Acid/administration & dosage , Neuroprotective Agents/administration & dosage , Oxidation-Reduction/drug effects , Quinolinic Acid/administration & dosage , Animals , Antioxidants/administration & dosage , Encephalitis/chemically induced , Inflammation Mediators/metabolism , Male , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
6.
PLoS One ; 14(12): e0226205, 2019.
Article En | MEDLINE | ID: mdl-31809528

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.


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
7.
Sci Rep ; 9(1): 6108, 2019 04 15.
Article En | MEDLINE | ID: mdl-30988385

The global increase in resorting to artificial nutritional formulas replacing breastfeeding has been identified among the complex causes of the obesity epidemic in infants and children. One of the factors recently recognized to influence metabolism and weight gain is kynurenic acid (KYNA), an agonist of G protein-coupled receptor (GPR35). Therefore the aim of the study was to determine the concentration of KYNA in artificial nutritional formulas in comparison with its level in human breast milk and to evaluate developmental changes in rats exposed to KYNA enriched diet during the time of breastfeeding. KYNA levels were measured in milk samples from 25 heathy breast-feeding women during the first six months after labor and were compared with 21 time-adjusted nutritional formulas. Animal experiments were performed on male Wistar rats. KYNA was administered in drinking water. The content of KYNA in human milk increases more than 13 times during the time of breastfeeding while its level is significantly lower in artificial formulas. KYNA was detected in breast milk of rats and it was found that the supplementation of rat maternal diet with KYNA in drinking water results in its increase in maternal milk. By means of the immunoblotting technique, GPR35 was evidenced in the mucosa of the jejunum of 1-day-old rats and distinct morphological changes in the jejunum of 21-day-old rats fed by mothers exposed to water supplemented with KYNA were found. A significant reduction of body weight gain of rats postnatally exposed to KYNA supplementation without changes in total body surface and bone mineral density was observed. The rat offspring fed with breast milk with artificially enhanced KYNA content demonstrated a lower mass gain during the first 21 days of life, which indicates that KYNA may act as an anti-obesogen. Further studies are, therefore, warranted to investigate the mechanisms regulating KYNA secretion via breast milk, as well as the influence of breast milk KYNA on mass gain. In the context of lifelong obesity observed worldwide in children fed artificially, our results imply that insufficient amount of KYNA in baby formulas could be considered as one of the factors associated with increased mass gain.


Gastrointestinal Tract/drug effects , Infant Formula/chemistry , Kynurenic Acid/administration & dosage , Milk, Human/chemistry , Obesity/prevention & control , Animals , Breast Feeding , Dietary Supplements , Disease Models, Animal , Female , Gastrointestinal Tract/growth & development , Humans , Infant , Infant Nutritional Physiological Phenomena/drug effects , Infant, Newborn , Kynurenic Acid/analysis , Male , Metabolic Networks and Pathways/drug effects , Obesity/epidemiology , Obesity/etiology , Rats , Rats, Wistar , Weight Gain/drug effects
8.
Pharmacol Rep ; 71(1): 139-148, 2019 Feb.
Article En | MEDLINE | ID: mdl-30550994

BACKGROUND: The G protein-coupled receptor 35 (GPR35), is considered important for nociceptive transmission, as suggested by accumulating evidence. This receptor was discovered in 1998; however, a lack of pharmacological tools prevented a complete understanding of its function and how to exploit it therapeutically. We studied the influence of CXCL17, kynurenic acid and zaprinast on nociceptive transmission in naïve and neuropathic mice. Additionally, we investigated the influence of kynurenic acid and zaprinast on morphine effectiveness in neuropathic pain. METHODS: The chronic constriction injury (CCI) of the sciatic nerve in Swiss mice was performed. The CXCL17, kynurenic acid, zaprinast and morphine were injected intrathecally into naive and CCI-exposed mice at day 14. To evaluate tactile and thermal hypersensitivity, the von Frey and cold plate tests were used, respectively. RESULTS: Our results have shown, for the first time, that administration of CXCL17 in naïve mice induced strong pain-related behaviours, as measured by von Frey and cold plate tests. Moreover, we demonstrated that kynurenic acid and zaprinast diminished CXCL17-evoked pain-related behaviours in both tests. Kynurenic acid and zaprinast reduced thermal and tactile hypersensitivity developed by sciatic nerve injury and strongly enhanced the effectiveness of morphine in neuropathy. CONCLUSIONS: Our study highlights the importance of GPR35 as a receptor involved in neuropathic pain development. Therefore, these results suggest that the modulation of GPR35 could become a potential strategy for the treatment of neuropathic pain.


Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Behavior, Animal/drug effects , Chemokines, CXC/toxicity , Kynurenic Acid/pharmacology , Morphine/pharmacology , Pain Perception/drug effects , Pain Threshold/drug effects , Purinones/pharmacology , Sciatica/drug therapy , Spinal Cord/drug effects , Analgesics/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Chemokines, CXC/administration & dosage , Disease Models, Animal , Injections, Spinal , Kynurenic Acid/administration & dosage , Male , Mice , Morphine/administration & dosage , Purinones/administration & dosage , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Sciatica/chemically induced , Sciatica/physiopathology , Sciatica/psychology , Spinal Cord/metabolism , Spinal Cord/physiopathology
9.
Pharmacol Rep ; 70(6): 1089-1096, 2018 Dec.
Article En | MEDLINE | ID: mdl-30308459

BACKGROUND: Kynurenic acid (KYNA) is a biologically active metabolite of tryptophan exerting action on several receptors located in the brain and periphery. KYNA can be synthesized endogenously or supplied in the diet. It was documented that KYNA is present in various types of food. However, its presence in beverages was not yet investigated. Here, we measured content of KYNA in tea and coffee as well as analyzed distribution and fate of intragastrically administered labelled KYNA in mice. METHODS: 16 and 13 studied samples of tea and coffee, respectively were of commercial origin. Tea and coffee infusions were prepared according to the producers' guidelines. KYNA content in beverages was measured by means of HPLC detection. Adult male mice were used for analysis of fate of intragastrically administered labelled KYNA and collected samples were analyzed using liquid scintillation counter. RESULTS: KYNA was identified in all studied beverages. Amounts of KYNA found in various types of beverages differed significantly. The highest content of KYNA in tea and coffee was 8.7 µg/100 ml and 0.63 µg/100 ml, respectively. It was found that KYNA administered intragastrically as a liquid is absorbed from the digestive system and readily excreted in urine. The atypical kinetics of KYNA distribution were found in intestinal content of cecum, where it appeared later and persisted longer than in other tissues. CONCLUSIONS: Our data show that tea and coffee intake may contribute to KYNA content in the human organism. The distribution pattern of KYNA delivered as a liquid suggests that it either directly affects digestive system's functioning and intestinal microbiome composition, or participates in the whole body pool of KYNA.


Coffee/metabolism , Kynurenic Acid/administration & dosage , Kynurenic Acid/metabolism , Liver/metabolism , Spleen/metabolism , Tea/metabolism , Animals , Beverages , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Liver/drug effects , Male , Mice , Spleen/drug effects , Tissue Distribution/drug effects , Tissue Distribution/physiology
10.
J. physiol. biochem ; 74(2): 325-334, mayo 2018. graf, ilus, tab
Article En | IBECS | ID: ibc-178988

To assess the possible function of glutamate in the interaction between the dorsomedial hypothalamic nucleus-perifornical area (DMH-PeF) and the A5 pontine region (A5), cardiovascular and respiratory changes were studied in response to electrical stimulation of the DMH-PeF (1 ms pulses, 30-50 miA given at 100 Hz for 5 s) before and after the microinjection of kynurenic acid (non-specific glutamate receptor antagonist; 50 nl, 5 nmol), MK-801 (NMDA receptor antagonist; 50 nl, 50 nmol), CNQX (non-NMDA receptor antagonist; 50 nl, 50 nmol) or MCPG (metabotropic glutamate receptor antagonist; 50 nl, 5 nmol) within the A5 region. DMH-PeF electrical stimulation elicited a pressor (p < 0.001) and tachycardic response (p < 0.001) which was accompanied by an inspiratory facilitation characterised by an increase in respiratory rate (p < 0.001) due to a decrease in expiratory time (p < 0.01). Kynurenic acid within the A5 region decreased the tachycardia (p < 0.001) and the intensity of the blood pressure response (p < 0.001) to DMH-PeF stimulation. After the microinjection of MK-801 and CNQX into the A5 region, the magnitude of the tachycardia and the pressor response were decreased (p < 0.05 and p < 0.01; p < 0.001 and p < 0.05, respectively). After MCPG microinjection into the A5 region, a decrease in the tachycardia (p < 0.001) with no changes in the pressor response was observed during DMH-PeF stimulation. The respiratory response elicited by DMH-PeF stimulation was not changed after the microinjection of kynurenic acid, MK-801, CNQX or MCPG within the A5 region. These results suggest that A5 region glutamate receptors play a role in the cardiovascular response elicited from the DMH-PeF. The possible mechanisms involved in these interactions are discussed


Animals , Male , Rats , Cardiovascular Physiological Phenomena , Dorsomedial Hypothalamic Nucleus/physiology , Fornix, Brain/physiology , Receptors, Glutamate/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/administration & dosage , Blood Pressure , Dizocilpine Maleate/administration & dosage , Electric Stimulation , Glycine/administration & dosage , Glycine/antagonists & inhibitors , Kynurenic Acid/administration & dosage , Tachycardia/physiopathology , Microinjections
11.
J Physiol Biochem ; 74(2): 325-334, 2018 May.
Article En | MEDLINE | ID: mdl-29577176

To assess the possible function of glutamate in the interaction between the dorsomedial hypothalamic nucleus-perifornical area (DMH-PeF) and the A5 pontine region (A5), cardiovascular and respiratory changes were studied in response to electrical stimulation of the DMH-PeF (1 ms pulses, 30-50 µA given at 100 Hz for 5 s) before and after the microinjection of kynurenic acid (non-specific glutamate receptor antagonist; 50 nl, 5 nmol), MK-801 (NMDA receptor antagonist; 50 nl, 50 nmol), CNQX (non-NMDA receptor antagonist; 50 nl, 50 nmol) or MCPG (metabotropic glutamate receptor antagonist; 50 nl, 5 nmol) within the A5 region. DMH-PeF electrical stimulation elicited a pressor (p < 0.001) and tachycardic response (p < 0.001) which was accompanied by an inspiratory facilitation characterised by an increase in respiratory rate (p < 0.001) due to a decrease in expiratory time (p < 0.01). Kynurenic acid within the A5 region decreased the tachycardia (p < 0.001) and the intensity of the blood pressure response (p < 0.001) to DMH-PeF stimulation. After the microinjection of MK-801 and CNQX into the A5 region, the magnitude of the tachycardia and the pressor response were decreased (p < 0.05 and p < 0.01; p < 0.001 and p < 0.05, respectively). After MCPG microinjection into the A5 region, a decrease in the tachycardia (p < 0.001) with no changes in the pressor response was observed during DMH-PeF stimulation. The respiratory response elicited by DMH-PeF stimulation was not changed after the microinjection of kynurenic acid, MK-801, CNQX or MCPG within the A5 region. These results suggest that A5 region glutamate receptors play a role in the cardiovascular response elicited from the DMH-PeF. The possible mechanisms involved in these interactions are discussed.


Cardiovascular Physiological Phenomena , Dorsomedial Hypothalamic Nucleus/physiology , Fornix, Brain/physiology , Receptors, Glutamate/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/administration & dosage , Animals , Blood Pressure , Dizocilpine Maleate/administration & dosage , Electric Stimulation , Excitatory Amino Acid Antagonists/administration & dosage , Glycine/administration & dosage , Glycine/analogs & derivatives , Heart Rate , Kynurenic Acid/administration & dosage , Male , Microinjections , Rats , Respiratory Rate , Tachycardia/physiopathology
12.
J Pharm Sci ; 107(6): 1572-1576, 2018 06.
Article En | MEDLINE | ID: mdl-29421218

Scarring is a consequence of biological tissue repair following trauma. Currently, there are no generally agreed ways to prevent scarring. Recently, kynurenic acid has shown to be a potent modulator of extracellular matrix deposition and remodeling. Kynurenic acid can reduce matrix deposition and other fundamental characteristics of fibrosis in vitro and in vivo. Specifically, kynurenic acid has shown to increase matrix metalloproteinase-1 activity and subsequently reduce collagen deposition in a rabbit ear scar model. In the present study kynurenic acid cream in different concentrations was topically applied on healthy skin on volunteers to assess skin reactions and skin sensitivity in both acute and chronic application settings. Skin reactions were assessed, and concentrations for kynurenic acid were assessed both form serum and urine. Results showed to acute or delayed skin reactions. Kynurenic acid was not detectable in blood at any time point, and only trace elements of kynurenic acid were found in urine. This study supports safety and tolerability of topically administered FS2 when using a liposomal, compounding base carrier.


Excitatory Amino Acid Antagonists/administration & dosage , Kynurenic Acid/administration & dosage , Skin/drug effects , Administration, Topical , Adolescent , Adult , Aged , Double-Blind Method , Excitatory Amino Acid Antagonists/adverse effects , Excitatory Amino Acid Antagonists/blood , Excitatory Amino Acid Antagonists/urine , Humans , Kynurenic Acid/adverse effects , Kynurenic Acid/blood , Kynurenic Acid/urine , Liposomes/adverse effects , Liposomes/chemistry , Middle Aged , Skin/pathology , Skin Cream/adverse effects , Skin Cream/chemistry , Skin Diseases/chemically induced , Skin Diseases/diagnosis , Skin Tests , Young Adult
13.
Am J Physiol Heart Circ Physiol ; 314(3): H563-H572, 2018 03 01.
Article En | MEDLINE | ID: mdl-29212793

Intermittent hypoxia causes a persistent increase in sympathetic activity that progresses to hypertension in chronic conditions such as obstructive sleep apnea. Pituitary adenylate cyclase-activating polypeptide (PACAP) is an excitatory neurotransmitter that causes long-lasting sympathetic excitation. We aimed to determine if intermittent activation of the rostral ventrolateral medulla (RVLM) causes PACAP-mediated elevation of sympathetic nerve activity, termed sympathetic long-term facilitation (sLTF). The role of PACAP in mediating sLTF in response to intermittent activation of the RVLM was investigated in urethane-anaesthetized and artificially ventilated rats ( n = 65, Sprague-Dawley). Bilateral RVLM microinjections of the PACAP type 1 receptor/vasoactive intestinal polypeptide receptor type 2 receptor antagonist PACAP-(6-38) [ n = 6, change (Δ): -16.4 ± 6.5%) or an ionotropic glutamate antagonist, kynurenate ( n = 6, Δ:-7.2 ± 2.3%), blocked the development of acute intermittent hypoxia-induced sLTF ( n = 6, Δ: 49.2 ± 14.2%). Intermittent RVLM microinjections of glutamate caused sLTF ( n = 5, Δ: 56.9 ± 14.7%) that was abolished by PACAP-(6-38) pretreatment ( n = 5, Δ:-1.2 ± 4.7%). Conversely, intermittent microinjections of PACAP in the RVLM did not elicit sLTF. Intermittent bilateral disinhibition of the RVLM by microinjection of γ-aminobutyric acid in the caudal ventrolateral medulla did not elicit sLTF. Direct activation of RVLM neurons is crucial for the development of sLTF. PACAP and glutamate act synergistically in the RVLM, with both being necessary for the sLTF response. We found that activation of glutamate but not PACAP receptors is necessary and sufficient to generate sLTF, even in the absence of intermittent hypoxia. Our results demonstrate that PACAP within the RVLM may contribute to the development of obstructive sleep apnea -induced hypertension. NEW & NOTEWORTHY Pharmacological blockade of either pituitary adenylate cyclase-activating polypeptide (PACAP) or ionotropic glutamate receptors in the rostral ventrolateral medulla prevents development of sympathetic long-term facilitation. PACAP receptor inhibition prevents the occurrence of hypoxia-induced peripheral chemoreflex sensitization. Thus, PACAP receptors may be a potential therapeutic target serving to reduce heightened sympathetic tone and hypersensitized cardiovascular reflexes.


Hypoxia/physiopathology , Kynurenic Acid/administration & dosage , Long-Term Potentiation/drug effects , Medulla Oblongata/drug effects , Peptide Fragments/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Sleep Apnea, Obstructive/physiopathology , Sympathetic Nervous System/drug effects , Acute Disease , Animals , Disease Models, Animal , Glutamic Acid/administration & dosage , Hypertension/etiology , Hypertension/physiopathology , Hypoxia/complications , Hypoxia/metabolism , Male , Medulla Oblongata/metabolism , Medulla Oblongata/physiopathology , Microinjections , Rats, Sprague-Dawley , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/metabolism , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Synaptic Transmission/drug effects , Time Factors , gamma-Aminobutyric Acid/administration & dosage
14.
Scand J Pain ; 17: 243-251, 2017 10.
Article En | MEDLINE | ID: mdl-29229209

BACKGROUND AND AIMS: Neuropathic pain is a significant medical problem needing more effective treatments with fewer side effects. Overactive glutamatergic transmission via N-methyl-d-aspartate receptors (NMDARs) are known to play a role in central sensitization and neuropathic pain. Although ketamine, a NMDAR channel-blocking antagonist, is often used for neuropathic pain, its side-effect profile and abusive potential has prompted the search for a safer effective oral analgesic. A novel oral prodrug, AV-101 (l-4 chlorokynurenine), which, in the brain, is converted into one of the most potent and selective GlyB site antagonists of the NMDAR, has been demonstrated to be active in animal models of neuropathic pain. The two Phase 1 studies reported herein were designed to assess the safety and pharmacokinetics of AV-101, over a wide dose range, after daily dosing for 14-days. As secondary endpoints, AV-101 was evaluated in the capsaicin-induced pain model. METHODS: The Phase 1A study was a single-site, randomized, double-blind, placebo-controlled, single oral ascending dose (30-1800mg) study involving 36 normal healthy volunteers. The Phase 1B study was a single-site randomized, double-blind, placebo-controlled, study of multiple ascending doses (360, 1080, and 1440mg/day) of AV-101 involving 50 normal healthy volunteers, to whom AV-101 or placebo were administered orally daily for 14 consecutive days. Subjects underwent PK blood analyses, laboratory assessments, physical examination, 12-lead ECG, ophthalmological examination, and various neurocognitive assessments. The effect of AV-101 was evaluated in the intradermally capsaicin-induced pain model (ClinicalTrials.gov Identifier: NCT01483846). RESULTS: Two Phase 1, with an aggregate of 86 subjects, demonstrated that up to 14 days of oral AV-101, up to 1440mg per day, was safe and very well tolerated with AEs quantitively and qualitatively like those observed with placebo. Mean half-life values of AV-101 were consistent across doses, ranging with an average of 1.73h, with the highest Cmax (64.4µg/mL) and AUC0-t (196µgh/mL) values for AV-101 occurring in the 1440-mg dose group. In the capsaicin induce-pain model, there was no significant change in the area under the pain time curve (AUPC) for the spontaneous pain assessment between the treatment and the placebo groups on Day 1 or 14 (the primary endpoint). In contrast, there were consistent reductions at 60-180min on Day 1 after dosing for allodynia, mechanical hyperalgesia, heat hyperalgesia, and spontaneous pain, and on Day 14 after dosing for heat hyperalgesia. CONCLUSIONS: Although, AV-101 did not reach statistical significance in reducing pain, there were consistent reductions, for allodynia pain and mechanical and heat hyperalgesia. Given the excellent safety profile and PK characteristics demonstrated by this study, future clinical trials of AV-101 in neuropathic pain are justified. IMPLICATIONS: This article presents the safety and PK of AV-101, a novel oral prodrug producing a potent and selective GlyB site antagonist of the NMDA receptor. These data indicate that AV-101 has excellent safety and PK characteristics providing support for advancing AV-101 into Phase 2 studies in neuropathic pain, and even provides data suggesting that AV-101 may have a role in treating depression.


Healthy Volunteers , Hyperalgesia , Kynurenic Acid/analogs & derivatives , Kynurenine/analogs & derivatives , Neuralgia/drug therapy , Neuroprotective Agents/pharmacokinetics , Adult , Cross-Over Studies , Female , Humans , Hyperalgesia/drug therapy , Kynurenic Acid/administration & dosage , Kynurenic Acid/blood , Kynurenic Acid/pharmacokinetics , Kynurenine/administration & dosage , Kynurenine/blood , Kynurenine/pharmacokinetics , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Pain Measurement
15.
J Neurosci ; 37(33): 7921-7929, 2017 08 16.
Article En | MEDLINE | ID: mdl-28729445

Increased concentrations of kynurenic acid (KYNA) in the prefrontal cortex (PFC) are thought to contribute to the development of cognitive deficits observed in schizophrenia. Although this view is consistent with preclinical studies showing a negative impact of prefrontal KYNA elevation on executive function, the mechanism underlying such a disruption remains unclear. Here, we measured changes in local field potential (LFP) responses to ventral hippocampal stimulation in vivo and conducted whole-cell patch-clamp recordings in brain slices to reveal how nanomolar concentrations of KYNA alter synaptic transmission in the PFC of male adult rats. Our data show that prefrontal infusions of KYNA attenuated the inhibitory component of PFC LFP responses, a disruption that resulted from local blockade of α7-nicotinic acetylcholine receptors (α7nAChR). At the cellular level, we found that the inhibitory action exerted by KYNA in the PFC occurred primarily at local GABAergic synapses through an α7nAChR-dependent presynaptic mechanism. As a result, the excitatory-inhibitory ratio of synaptic transmission becomes imbalanced in a manner that correlates highly with the level of GABAergic suppression by KYNA. Finally, prefrontal infusion of a GABAAR positive allosteric modulator was sufficient to overcome the disrupting effect of KYNA and normalized the pattern of LFP inhibition in the PFC. Thus, the preferential inhibitory effect of KYNA on prefrontal GABAergic transmission could contribute to the onset of cognitive deficits observed in schizophrenia because proper GABAergic control of PFC output is one key mechanism for supporting such cortical functions.SIGNIFICANCE STATEMENT Brain kynurenic acid (KYNA) is an astrocyte-derived metabolite and its abnormal elevation in the prefrontal cortex (PFC) is thought to impair cognitive functions in individuals with schizophrenia. However, the mechanism underlying the disrupting effect of KYNA remains unclear. Here we found that KYNA biases the excitatory-inhibitory balance of prefrontal synaptic activity toward a state of disinhibition. Such disruption emerges as a result of a preferential suppression of local GABAergic transmission by KYNA via presynaptic inhibition of α7-nicotinic acetylcholine receptor signaling. Therefore, the degree of GABAergic dysregulation in the PFC could be a clinically relevant contributing factor for the onset of cognitive deficits resulting from abnormal increases of cortical KYNA.


GABAergic Neurons/physiology , Kynurenic Acid/toxicity , Prefrontal Cortex/physiology , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/physiology , Animals , Dose-Response Relationship, Drug , GABAergic Neurons/drug effects , Infusions, Intraventricular , Kynurenic Acid/administration & dosage , Male , Organ Culture Techniques , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley
16.
J Headache Pain ; 18(1): 39, 2017 Dec.
Article En | MEDLINE | ID: mdl-28337634

BACKGROUND: Migraine is a debilitating neurological disorder where trigeminovascular activation plays a key role. We have previously reported that local application of Complete Freund's Adjuvant (CFA) onto the dura mater caused activation in rat trigeminal ganglion (TG) which was abolished by a systemic administration of kynurenic acid (KYNA) derivate (SZR72). Here, we hypothesize that this activation may extend to the trigeminal complex in the brainstem and is attenuated by treatment with SZR72. METHODS: Activation in the trigeminal nucleus caudalis (TNC) and the trigeminal tract (Sp5) was achieved by application of CFA onto the dural parietal surface. SZR72 was given intraperitoneally (i.p.), one dose prior CFA deposition and repeatedly daily for 7 days. Immunohistochemical studies were performed for mapping glutamate, c-fos, PACAP, substance P, IL-6, IL-1ß and TNFα in the TNC/Sp5 and other regions of the brainstem and at the C1-C2 regions of the spinal cord. RESULTS: We found that CFA increased c-fos and glutamate immunoreactivity in TNC and C1-C2 neurons. This effect was mitigated by SZR72. PACAP positive fibers were detected in the fasciculus cuneatus and gracilis. Substance P, TNFα, IL-6 and IL-1ß immunopositivity were detected in fibers of Sp5 and neither of these molecules showed any change in immunoreactivity following CFA administration. CONCLUSION: This is the first study demonstrating that dural application of CFA increases the expression of c-fos and glutamate in TNC neurons. Treatment with the KYNA analogue prevented this expression.


Dura Mater/drug effects , Dura Mater/metabolism , Freund's Adjuvant/administration & dosage , Glutamic Acid/biosynthesis , Kynurenic Acid/analogs & derivatives , Proto-Oncogene Proteins c-fos/biosynthesis , Administration, Topical , Animals , Freund's Adjuvant/toxicity , Gene Expression Regulation , Kynurenic Acid/administration & dosage , Male , Migraine Disorders/chemically induced , Migraine Disorders/metabolism , Migraine Disorders/prevention & control , Rats , Rats, Sprague-Dawley
17.
Auton Neurosci ; 203: 67-73, 2017 Mar.
Article En | MEDLINE | ID: mdl-28173996

Adrenaline is an important counter-regulatory hormone that helps restore glucose homeostasis during hypoglycaemia. However, the neurocircuitry that connects the brain glucose sensors and the adrenal sympathetic outflow to the chromaffin cells is poorly understood. We used electrical microstimulation of the perifornical hypothalamus (PeH) and the rostral ventrolateral medulla (RVLM) combined with adrenal sympathetic nerve activity (ASNA) recording to examine the relationship between the RVLM, the PeH and ASNA. In urethane-anaesthetised male Sprague-Dawley rats, intermittent single pulse electrical stimulation of the rostroventrolateral medulla (RVLM) elicited an evoked ASNA response that consisted of early (60±3ms) and late peaks (135±4ms) of preganglionic and postganglionic activity. In contrast, RVLM stimulation evoked responses in lumbar sympathetic nerve activity that were almost entirely postganglionic. PeH stimulation also produced an evoked excitatory response consisting of both preganglionic and postganglionic excitatory peaks in ASNA. Both peaks in ASNA following RVLM stimulation were reduced by intrathecal kynurenic acid (KYN) injection. In addition, the ASNA response to systemic neuroglucoprivation induced by 2-deoxy-d-glucose was abolished by bilateral microinjection of KYN into the RVLM. This suggests that a glutamatergic pathway from the perifornical hypothalamus (PeH) relays in the RVLM to activate the adrenal SPN and so modulate ASNA. The main findings of this study are that (i) adrenal premotor neurons in the RVLM may be, at least in part, glutamatergic and (ii) that the input to these neurons that is activated during neuroglucoprivation is also glutamatergic.


Adrenal Glands/metabolism , Autonomic Pathways/metabolism , Glucose/metabolism , Glutamic Acid/metabolism , Hypothalamus/metabolism , Sympathetic Nervous System/metabolism , Adrenal Glands/drug effects , Adrenal Glands/innervation , Anesthetics, Intravenous/pharmacology , Animals , Autonomic Pathways/drug effects , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Hypothalamus/drug effects , Kynurenic Acid/administration & dosage , Kynurenic Acid/metabolism , Lumbar Vertebrae , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism , Sympathetic Nervous System/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Urethane/pharmacology
18.
Drug Des Devel Ther ; 11: 27-34, 2017.
Article En | MEDLINE | ID: mdl-28053504

BACKGROUND: The complex pathophysiology of migraine is not yet clearly understood; therefore, experimental models are essential for the investigation of the processes related to migraine headache, which include cortical spreading depression (CSD) and NO donor-induced neurovascular changes. Data on the assessment of drug efficacy in these models are often limited, which prompted us to investigate a novel combined migraine model in which an effective pharmacon could be more easily identified. MATERIALS AND METHODS: In vivo electrophysiological experiments were performed to investigate the effect of nitroglycerin (NTG) on CSD induced by KCl application. In addition, sumatriptan and newly synthesized neuroactive substances (analogues of the neuromodulator kynurenic acid [KYNA]) were also tested. RESULTS: The basic parameters of CSDs were unchanged following NTG administration; however, propagation failure was decreased compared to the controls. Sumatriptan decreased the number of CSDs, whereas propagation failure was as minimal as in the NTG group. On the other hand, both of the KYNA analogues restored the ratio of propagation to the control level. DISCUSSION: The ratio of propagation appeared to be the indicator of the effect of NTG. This is the first study providing direct evidence that NTG influences CSD; furthermore, we observed different effects of sumatriptan and KYNA analogues. Sumatriptan changed the generation of CSDs, whereas the analogues acted on the propagation of the waves. Our experimental design overlaps with a large spectrum of processes present in migraine pathophysiology, and it can be a useful experimental model for drug screening.


Cortical Spreading Depression/drug effects , Kynurenic Acid/pharmacology , Migraine Disorders/drug therapy , Nitroglycerin/pharmacology , Sumatriptan/pharmacology , Vasodilator Agents/pharmacology , Dose-Response Relationship, Drug , Humans , Injections, Intraperitoneal , Kynurenic Acid/administration & dosage , Nitroglycerin/administration & dosage , Sumatriptan/administration & dosage , Vasodilator Agents/administration & dosage
19.
Nat Commun ; 7: 13501, 2016 11 22.
Article En | MEDLINE | ID: mdl-27874011

Impaired glucose homeostasis and energy balance are integral to the pathophysiology of diabetes and obesity. Here we show that administration of a glycine transporter 1 (GlyT1) inhibitor, or molecular GlyT1 knockdown, in the dorsal vagal complex (DVC) suppresses glucose production, increases glucose tolerance and reduces food intake and body weight gain in healthy, obese and diabetic rats. These findings provide proof of concept that GlyT1 inhibition in the brain improves glucose and energy homeostasis. Considering the clinical safety and efficacy of GlyT1 inhibitors in raising glycine levels in clinical trials for schizophrenia, we propose that GlyT1 inhibitors have the potential to be repurposed as a treatment of both obesity and diabetes.


Diabetes Mellitus, Experimental/chemically induced , Glycine Plasma Membrane Transport Proteins/metabolism , Obesity/metabolism , Receptors, Lipoxin/administration & dosage , Animals , Brain/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism , Gene Expression Regulation/drug effects , Glycemic Index , Glycine Plasma Membrane Transport Proteins/genetics , Homeostasis , Kynurenic Acid/administration & dosage , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
20.
Eur J Pharm Sci ; 86: 67-74, 2016 Apr 30.
Article En | MEDLINE | ID: mdl-26924227

Core-shell nanoparticles (CSNPs) were developed to get over therapeutic amount of kynurenic acid (KYNA) across the blood-brain barrier (BBB). Bovine serum albumin (BSA) was used as core for encapsulation of KYNA and the BSA/KYNA composite was finally encapsulated by poly(allylamine) hydrochloride (PAH) polymer as shell. In the interest of the optimization of the synthesis the BSA and KYNA interaction was studied by two-dimensional surface plasmon resonance (SPR) technique as well. The average size of d~100 nm was proven by dynamic light scattering (DLS) and transmission electron microscopy (TEM), while the structure of the composites was characterized by fluorescence (FL) and circular dichroism (CD) spectroscopy. The in vitro release properties of KYNA were investigated by a vertical diffusion cell at 25.0 °C and 37.5 °C and the kinetic of the release were discussed. The penetration capacity of the NPs into the central nervous system (CNS) was tested by an in vitro BBB model. The results demonstrated that the encapsulated KYNA had significantly higher permeability compared to free KYNA molecules. In the neurobiological serial of in vivo experiments the effects of peripherally administered KYNA with CSNPs were studied in comparison with untreated KYNA. These results clearly proved that KYNA in the CSNPs, administrated peripherally is suitable to cross the BBB and to induce electrophysiological effects within the CNS. As the neuroprotective properties of KYNA nowadays are proven, the importance of the results is obvious.


Blood-Brain Barrier/metabolism , Drug Carriers/administration & dosage , Kynurenic Acid/administration & dosage , Nanoparticles/administration & dosage , Polyamines/administration & dosage , Serum Albumin, Bovine/administration & dosage , Animals , Circular Dichroism , Coculture Techniques , Drug Carriers/chemistry , Drug Liberation , Endothelial Cells/metabolism , Kynurenic Acid/chemistry , Kynurenic Acid/pharmacokinetics , Nanoparticles/chemistry , Neuroglia/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Pericytes/metabolism , Polyamines/chemistry , Polyamines/pharmacokinetics , Rats, Wistar , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/pharmacokinetics , Spectrometry, Fluorescence , Surface Plasmon Resonance
...