Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros













Intervalo de año de publicación
1.
Biol Aujourdhui ; 217(3-4): 157-160, 2023.
Artículo en Francés | MEDLINE | ID: mdl-38018943

RESUMEN

In France, suicidal behaviors remain a major public health issue. Depressed patients with suicidal ideation have more severe depressive symptoms, a more unfavorable disease course, and a greater number of suicide attempts than patients without suicidal ideation. Unfortunately, conventional antidepressants tend to be less effective in patients with suicidal tendencies than in those without. Nevertheless, promising advancements have emerged with the use of ketamine, which has shown significant and rapid efficacy in reducing the intensity of suicidal ideation in depressed patients within the first 72 h after its administration. Several mechanisms are potentially involved: (1) reduction of anhedonia. It has been demonstrated that ketamine reduces both anhedonia and suicidal ideation. In depressed patients, the reduction of anhedonia observed 2 h after ketamine administration is associated with metabolic changes in the anterior cingulate cortex involved in suicidal ideation; (2) activation of neuroplasticity cascades. The reduction in suicidal ideation within 24 h following ketamine administration is correlated with changes in plasma BDNF levels and is modulated by the Val66Met functional polymorphism of the BDNF gene. Moreover, preclinical and clinical studies have shown that ketamine induces functional and connectivity changes in the prefrontal and anterior cingulate regions, which are strongly implicated in suicidal behaviors; (3) reduction of inflammation. It is now widely accepted that suicidal behaviors are associated with low-grade inflammation, and with elevated quinolinic acid and reduced kynurenic acid levels. Interestingly, predictors of a reduction in suicidal ideation after ketamine infusion include initial severity of suicidal thoughts and depression, as well as baseline blood levels of kynurenic acid; (4) involvement of the opioidergic system. Post-mortem studies have indicated alterations in the opioidergic system related to suicidal behaviors. A recent study suggested that the antisuicidal effect of ketamine may depend on this system because naltrexone, an antagonist of mu opioid receptors, abolished the typical antidepressant effect and reduction in suicidal ideation observed following ketamine administration. In conclusion, ketamine exhibits promising potential in mitigating suicidal ideation - its effects are specific, rapid, albeit temporary. The suggested mechanisms driving its efficacy are multifaceted. Nevertheless, it is yet to be determined whether ketamine administration can effectively prevent suicidal behaviors.


Title: Comment la kétamine peut-elle aider à mieux prendre en charge le risque suicidaire ? Abstract: Les patients souffrant de dépression suicidaire répondent moins bien aux traitements antidépresseurs conventionnels que ceux qui n'ont pas d'idées suicidaires. Une avancée prometteuse dans ce domaine est l'utilisation de la kétamine, qui a montré une efficacité significative et rapide dans la réduction de l'intensité des idées suicidaires chez les patients déprimés. Des études ont montré qu'une seule perfusion intraveineuse de kétamine à faible dose pouva3it réduire de manière significative et durable les idées de suicide. De plus, la kétamine semble également réduire l'anhédonie, un symptôme associé aux idées suicidaires. Les mécanismes d'action de la kétamine sont multiples. Elle stimule la neuroplasticité via l'activation de la voie du BDNF (facteur neurotrophique du cerveau) et réduit l'inflammation. De plus, la kétamine semble agir sur le système opioïdergique, qui est impliqué dans les conduites suicidaires. En conclusion, la kétamine présente un intérêt dans la réduction des idées suicidaires chez les patients déprimés. Cependant, il est nécessaire de déterminer si son administration permet de prévenir les actes suicidaires. De plus amples recherches sont nécessaires pour mieux comprendre les mécanismes d'action de la kétamine et développer des stratégies thérapeutiques ciblées pour prévenir les conduites suicidaires.


Asunto(s)
Trastorno Depresivo Mayor , Ketamina , Humanos , Ketamina/efectos adversos , Anhedonia , Ideación Suicida , Ácido Quinurénico/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Antidepresivos/efectos adversos , Inflamación
2.
Int J Mol Sci ; 23(16)2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-36012164

RESUMEN

Kynurenic acid was included in the three compounds (caffeic acid, chlorogenic acid, and kynurenic acid) that showed high antioxidant and anti-inflammatory potential among the phenolic compounds contained in Gynura procumbens. In this study, the mechanism of cancer cell death induced by kynurenic acid (KYNA), which has the highest molecular binding affinity, in the gastric cancer cell line AGS was confirmed in molecular docking analysis. KYNA showed the most cancer cell death effect on AGS cells among several gastric cancer cell lines (MKN, AGS, and SNU). AGS cells were used for later experiments, and KYNA concentrations of 0, 150, 200, and 250 µM were used. KYNA inhibited cell migration and proliferation in AGS cells in a concentration-dependent manner. G2/M phase cell cycle arrest and reduction of related proteins (Cdc25C, CDK1 and CyclinB1) were confirmed in KYNA-treated AGS cells. Apoptosis of KYNA-treated AGS cells was confirmed by Annexin V/propidium iodide (PI) staining flow cytometry analysis. As a result of morphological chromatin condensation through DAPI (4',6-diamidino-2-phenylindole), intense blue fluorescence was confirmed. The mechanism of apoptosis induction of KYNA-treated AGS cells was confirmed by western blotting. In the extrinsic pathway, apoptosis induction markers FasL, Fas, and Caspase-3 and -8 were increased in a concentration-dependent manner upon KYNA treatment. In the intrinsic pathway, the expression of anti-apoptotic factors PI3K, AKT, and Bcl-xL was down-regulated, and the expression of apoptosis-inducing factors BAD, Bak, Bax, Cytochrom C, and Caspase-9 was up-regulated. Therefore, in the present study, we strongly imply that KYNA induces apoptosis in AGS gastric cancer cells. This suggests that KYNA, a natural compound, could be the basis for drug for the treatment of gastric cancer.


Asunto(s)
Neoplasias Gástricas , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Ácido Quinurénico/farmacología , Ácido Quinurénico/uso terapéutico , Simulación del Acoplamiento Molecular , Neoplasias Gástricas/metabolismo
3.
Science ; 377(6606): 621-629, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35926043

RESUMEN

Kynurenic acid (KynA) is tissue protective in cardiac, cerebral, renal, and retinal ischemia models, but the mechanism is unknown. KynA can bind to multiple receptors, including the aryl hydrocarbon receptor, the a7 nicotinic acetylcholine receptor (a7nAChR), multiple ionotropic glutamate receptors, and the orphan G protein-coupled receptor GPR35. Here, we show that GPR35 activation was necessary and sufficient for ischemic protection by KynA. When bound by KynA, GPR35 activated Gi- and G12/13-coupled signaling and trafficked to the outer mitochondria membrane, where it bound, apparantly indirectly, to ATP synthase inhibitory factor subunit 1 (ATPIF1). Activated GPR35, in an ATPIF1-dependent and pertussis toxin-sensitive manner, induced ATP synthase dimerization, which prevented ATP loss upon ischemia. These findings provide a rationale for the development of specific GPR35 agonists for the treatment of ischemic diseases.


Asunto(s)
Ácido Quinurénico , Mitocondrias Cardíacas , Isquemia Miocárdica , Receptores Acoplados a Proteínas G , Adenosina Trifosfato/metabolismo , Animales , Humanos , Ácido Quinurénico/metabolismo , Ácido Quinurénico/farmacología , Ácido Quinurénico/uso terapéutico , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevención & control , Proteínas/metabolismo , Conejos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Proteína Inhibidora ATPasa
4.
Nord J Psychiatry ; 76(6): 451-456, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34928781

RESUMEN

BACKGROUND: Kynurenic acid (KYNA) is a metabolite of tryptophan (TRP). KYNA levels have been reported with controversial findings in patients with schizophrenia. AIM: This study aimed to investigate the probable effects of medication and illness chronicity on peripheral KYNA levels in schizophrenia. METHODS: We assessed peripheral (plasma) TRP metabolite levels in 38 drug-free patients with first-episode schizophrenia (FES), 65 patients with chronic schizophrenia (CHS), and 70 healthy controls by using high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS). The symptom severity of patients was evaluated by using the Positive and Negative Syndrome Scale (PANSS). Finally, we analyzed the association of TRP metabolites with symptom severity. RESULTS: We found significantly higher KYNA levels in FES patients than in both healthy controls (p < 0.01) and CHS patients (p < 0.05). No significant association was observed between plasma TRP metabolite levels and PANSS scores in either FES or CHS patients. CONCLUSIONS: In conclusion, elevated plasma KYNA levels may be a promising biomarker in FES patients. Medication and illness chronicity may affect peripheral KYNA levels with a currently unknown mechanism.


Asunto(s)
Ácido Quinurénico , Esquizofrenia , Cromatografía Liquida , Humanos , Ácido Quinurénico/uso terapéutico , Datos Preliminares , Esquizofrenia/diagnóstico , Esquizofrenia/tratamiento farmacológico , Espectrometría de Masas en Tándem
5.
CNS Neurosci Ther ; 28(1): 19-35, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34862742

RESUMEN

AIMS: The family of kynurenine pathway (KP) metabolites includes compounds produced along two arms of the path and acting in clearly opposite ways. The equilibrium between neurotoxic kynurenines, such as 3-hydroxykynurenine (3-HK) or quinolinic acid (QUIN), and neuroprotective kynurenic acid (KYNA) profoundly impacts the function and survival of neurons. This comprehensive review summarizes accumulated evidence on the role of KYNA in Alzheimer's, Parkinson's and Huntington's diseases, and discusses future directions of potential pharmacological manipulations aimed to modulate brain KYNA. DISCUSSION: The synthesis of specific KP metabolites is tightly regulated and may considerably vary under physiological and pathological conditions. Experimental data consistently imply that shift of the KP to neurotoxic branch producing 3-HK and QUIN formation, with a relative or absolute deficiency of KYNA, is an important factor contributing to neurodegeneration. Targeting specific brain regions to maintain adequate KYNA levels seems vital; however, it requires the development of precise pharmacological tools, allowing to avoid the potential cognitive adverse effects. CONCLUSIONS: Boosting KYNA levels, through interference with the KP enzymes or through application of prodrugs/analogs with high bioavailability and potency, is a promising clinical approach. The use of KYNA, alone or in combination with other compounds precisely influencing specific populations of neurons, is awaiting to become a significant therapy for neurodegenerative disorders.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/uso terapéutico , Ácido Quinurénico/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Quinurenina/análogos & derivados , Quinurenina/toxicidad , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Ácido Quinolínico/toxicidad , Transducción de Señal/efectos de los fármacos
6.
Front Immunol ; 12: 702764, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745090

RESUMEN

The pathophysiology of acute pancreatitis (AP) is not well understood, and the disease does not have specific therapy. Tryptophan metabolite L-kynurenic acid (KYNA) and its synthetic analogue SZR-72 are antagonists of the N-methyl-D-aspartate receptor (NMDAR) and have immune modulatory roles in several inflammatory diseases. Our aims were to investigate the effects of KYNA and SZR-72 on experimental AP and to reveal their possible mode of action. AP was induced by intraperitoneal (i.p.) injection of L-ornithine-HCl (LO) in SPRD rats. Animals were pretreated with 75-300 mg/kg KYNA or SZR-72. Control animals were injected with physiological saline instead of LO, KYNA and/or SZR-72. Laboratory and histological parameters, as well as pancreatic and systemic circulation were measured to evaluate AP severity. Pancreatic heat shock protein-72 and IL-1ß were measured by western blot and ELISA, respectively. Pancreatic expression of NMDAR1 was investigated by RT-PCR and immunohistochemistry. Viability of isolated pancreatic acinar cells in response to LO, KYNA, SZR-72 and/or NMDA administration was assessed by propidium-iodide assay. The effects of LO and/or SZR-72 on neutrophil granulocyte function was also studied. Almost all investigated laboratory and histological parameters of AP were significantly reduced by administration of 300 mg/kg KYNA or SZR-72, whereas the 150 mg/kg or 75 mg/kg doses were less or not effective, respectively. The decreased pancreatic microcirculation was also improved in the AP groups treated with 300 mg/kg KYNA or SZR-72. Interestingly, pancreatic heat shock protein-72 expression was significantly increased by administration of SZR-72, KYNA and/or LO. mRNA and protein expression of NMDAR1 was detected in pancreatic tissue. LO treatment caused acinar cell toxicity which was reversed by 250 µM KYNA or SZR-72. Treatment of acini with NMDA (25, 250, 2000 µM) did not influence the effects of KYNA or SZR-72. Moreover, SZR-72 reduced LO-induced H2O2 production of neutrophil granulocytes. KYNA and SZR-72 have dose-dependent protective effects on LO-induced AP or acinar toxicity which seem to be independent of pancreatic NMDA receptors. Furthermore, SZR-72 treatment suppressed AP-induced activation of neutrophil granulocytes. This study suggests that administration of KYNA and its derivative could be beneficial in AP.


Asunto(s)
Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/uso terapéutico , Pancreatitis Aguda Necrotizante/tratamiento farmacológico , Animales , Interleucina-1beta/análisis , Ácido Quinurénico/farmacología , Masculino , Microcirculación/efectos de los fármacos , N-Metilaspartato/farmacología , Pancreatitis Aguda Necrotizante/fisiopatología , Gravedad del Paciente , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/análisis
7.
Molecules ; 26(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064778

RESUMEN

Kynurenic acid (KYNA) is derived from tryptophan, formed by the kynurenic pathway. KYNA is being widely studied as a biomarker for neurological and cardiovascular diseases, as it is found in ischemic conditions as a protective agent; however, little is known about its effect after ischemia-reperfusion in the vascular system. We induced ischemia for 30 min followed by 5 min reperfusion (I/R) in the rat aorta for KYNA evaluation using functional assays combined with proteomics. KYNA recovered the exacerbated contraction induced by phenylephrine and relaxation induced by acetylcholine or sodium nitroprussiate in the I/R aorta, with vessel responses returning to values observed without I/R. The functional recovery can be related to the antioxidant activity of KYNA, which may be acting on the endothelium-injury prevention, especially during reperfusion, and to proteins that regulate neurotransmission and cell repair/growth, expressed after the KYNA treatment. These proteins interacted in a network, confirming a protein profile expression for endothelium and neuron repair after I/R. Thus, the KYNA treatment had the ability to recover the functionality of injured ischemic-reperfusion aorta, by tissue repairing and control of neurotransmitter release, which reinforces its role in the post-ischemic condition, and can be useful in the treatment of such disease.


Asunto(s)
Aorta/patología , Ácido Quinurénico/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Proteómica , Acetilcolina/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/fisiopatología , Modelos Animales de Enfermedad , Ácido Quinurénico/farmacología , Contracción Miocárdica/efectos de los fármacos , Daño por Reperfusión Miocárdica/fisiopatología , Nitroprusiato/farmacología , Fenilefrina/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
8.
Oxid Med Cell Longev ; 2021: 8874503, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34055199

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Hiperlipidemias/tratamiento farmacológico , Ácido Quinurénico/uso terapéutico , Obesidad/tratamiento farmacológico , Animales , Cangrejos Herradura , Ácido Quinurénico/farmacología , Masculino , Ratones
9.
Int J Mol Sci ; 22(1)2021 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-33401674

RESUMEN

The incidence of neurodegenerative diseases has increased greatly worldwide due to the rise in life expectancy. In spite of notable development in the understanding of these disorders, there has been limited success in the development of neuroprotective agents that can slow the progression of the disease and prevent neuronal death. Some natural products and molecules are very promising neuroprotective agents because of their structural diversity and wide variety of biological activities. In addition to their neuroprotective effect, they are known for their antioxidant, anti-inflammatory and antiapoptotic effects and often serve as a starting point for drug discovery. In this review, the following natural molecules are discussed: firstly, kynurenic acid, the main neuroprotective agent formed via the kynurenine pathway of tryptophan metabolism, as it is known mainly for its role in glutamate excitotoxicity, secondly, the dietary supplement pantethine, that is many sided, well tolerated and safe, and the third molecule, α-lipoic acid is a universal antioxidant. As a conclusion, because of their beneficial properties, these molecules are potential candidates for neuroprotective therapies suitable in managing neurodegenerative diseases.


Asunto(s)
Ácido Quinurénico/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Panteteína/análogos & derivados , Ácido Tióctico/metabolismo , Animales , Antioxidantes/uso terapéutico , Humanos , Ácido Quinurénico/uso terapéutico , Redes y Vías Metabólicas/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Panteteína/metabolismo , Panteteína/uso terapéutico , Ácido Tióctico/uso terapéutico
10.
Pharmacol Rep ; 72(2): 449-455, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32162182

RESUMEN

BACKGROUND: Kynurenic acid (KYNA) is an L-tryptophan metabolite with neuromodulatory activities, regulating the release of neurotransmitters such as glutamate, dopamine (DA), and acetylcholine (Ach). Dysregulation of the kynurenine pathway has been associated with neurodegenerative, neurological, and psychological disorders such as Alzheimer's disease, Parkinson's disease, Huntington's disease, major depressive disorder, and schizophrenia. METHODS: The antidepressant-like effects of KYNA were studied with a modified mouse forced swimming test (FST), and the potential involvement of the serotonin (SER), norepinephrine, DA, Ach, N-methyl-D-aspartate, or gamma-aminobutyric acid subunit A (GABAA) receptors in its antidepressant-like effect was assayed by modified combination mouse FST. In combination studies, the mice were pretreated with the respective receptor antagonist, cyproheptadine (CPH), phenoxybenzamine, yohimbine, propranolol, haloperidol (HPD), atropine, MK-801, or bicuculline (BCL). RESULTS: The FST revealed that KYNA reversed immobility, climbing, and swimming times, suggesting the antidepressant-like effects of KYNA. Furthermore, the combination studies showed that CPH prevented the antidepressant-like effects of KYNA on immobility, climbing, and swimming times, whereas HPD reduced climbing time and BCL influenced immobility and climbing times and prevented the effects of KYNA on swimming time. CONCLUSIONS: The results demonstrated, for the first time, the presence of antidepressant-like effects of KYNA in a modified mouse FST. Furthermore, modified combination FST showed that the antidepressant-like actions of KYNA strongly interacted with 5-hydroxytryptamine type 2 SER-ergic receptors, weakly interacted with D2, D3, D4 DA-ergic receptors, and interacted moderately with GABAA receptors.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Depresión/tratamiento farmacológico , Ácido Quinurénico/farmacología , Actividad Motora/efectos de los fármacos , Natación , Animales , Antidepresivos/uso terapéutico , Depresión/metabolismo , Modelos Animales de Enfermedad , Ácido Quinurénico/uso terapéutico , Masculino , Ratones Endogámicos , Receptores Dopaminérgicos/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Serotonina 5-HT2/metabolismo
11.
Curr Med Chem ; 26(34): 6261-6281, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29848264

RESUMEN

BACKGROUND: Migraine is one of the most disabling neurological conditions and associated with high socio-economic costs. Though certain aspects of the pathomechanism of migraine are still incompletely understood, the leading hypothesis implicates the role of the activation of the trigeminovascular system. Triptans are considered to be the current gold standard therapy for migraine attacks; however, their use in clinical practice is limited. Prophylactic treatment includes non-specific approaches for migraine prevention. All these support the need for future studies in order to develop innovative anti-migraine drugs. OBJECTIVE: The present study is a review of the current literature regarding new therapeutic lines in migraine research. METHODS: A systematic literature search in the database of PUBMED was conducted concerning therapeutic strategies in a migraine published until July 2017. RESULTS: Ongoing clinical trials with 5-HT1F receptor agonists and glutamate receptor antagonists offer promising new aspects for acute migraine treatment. Monoclonal antibodies against CGRP and the CGRP receptor are revolutionary in preventive treatment; however, further long-term studies are needed to test their tolerability. Preclinical studies show positive results with PACAP- and kynurenic acid-related treatments. Other promising therapeutic strategies (such as those targeting TRPV1, substance P, NOS, or orexin) have failed to show efficacy in clinical trials. CONCLUSION: Due to their side-effects, current therapeutic approaches are not suitable for all migraine patients. Especially frequent episodic and chronic migraine represents a therapeutic challenge for researchers. Clinical and preclinical studies are needed to untangle the pathophysiology of migraine in order to develop new and migraine-specific therapies.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/uso terapéutico , Trastornos Migrañosos/tratamiento farmacológico , Agonistas de Receptores de Serotonina/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Péptido Relacionado con Gen de Calcitonina/inmunología , Ensayos Clínicos como Asunto , Humanos , Ácido Quinurénico/uso terapéutico , Trastornos Migrañosos/diagnóstico , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/antagonistas & inhibidores
12.
Anal Cell Pathol (Amst) ; 2018: 1270483, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30327755

RESUMEN

Acute liver failure (ALF) is a life-threatening disorder of liver function. Kynurenic acid (KYNA), a tryptophan metabolite formed along the kynurenine metabolic pathway, possesses anti-inflammatory and antioxidant properties. Its presence in food and its potential role in the digestive system was recently reported. The aim of this study was to define the effect of KYNA on liver failure. The Wistar rat model of thioacetamide-induced liver injury was used. Morphological and biochemical analyses as well as the measurement of KYNA content in liver and hepatoprotective herbal remedies were conducted. The significant attenuation of morphological disturbances and aspartate and alanine transaminase activities, decrease of myeloperoxidase and tumor necrosis factor-α, and elevation of interleukin-10 levels indicating the protective effect of KYNA in thioacetamide (TAA) - induced liver injury were discovered. In conclusion, the hepatoprotective role of KYNA in an animal model of liver failure was documented and the use of KYNA in the treatment of ALF was suggested.


Asunto(s)
Ácido Quinurénico/uso terapéutico , Hígado/efectos de los fármacos , Hígado/lesiones , Tioacetamida/toxicidad , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Biología Computacional/métodos , Modelos Animales de Enfermedad , Humanos , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar
13.
Curr Med Chem ; 25(42): 5945-5957, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29532751

RESUMEN

Ischemic stroke is one of the leading causes of mortality and permanent disability in developed countries. Stroke induces massive glutamate release, which in turn causes N-Methyl-D-aspartate (NMDA) receptor over-excitation and thus, calcium overload in neurons leading to cell death via apoptotic cascades. The kynurenine pathway is a complex enzymatic cascade of tryptophan catabolism, generating various neuroactive metabolites. One metabolite, kynurenic acid (KYNA), is a potent endogenous NMDA glutamate receptor antagonist, making it a possible therapeutic tool to decrease excitotoxicity and neuroinflammation. Recently, clinical investigations have shown that during the acute phase of ischemic stroke, kynurenine pathway is activated and peripheral levels of metabolites correlated with worse outcome. In this review, we set out to summarize the current literature on the connection of the kynurenine pathway and ischemic stroke and set a course for future investigations and potential drug development.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/uso terapéutico , Quinurenina/análogos & derivados , Accidente Cerebrovascular/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/química , Humanos , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/metabolismo , Ácido Quinurénico/uso terapéutico , Quinurenina/química , Quinurenina/metabolismo , Quinurenina/uso terapéutico , Ácidos Quinolínicos/química , Ácidos Quinolínicos/metabolismo , Ácidos Quinolínicos/uso terapéutico , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Accidente Cerebrovascular/patología , Triptófano/metabolismo
14.
J Headache Pain ; 17(1): 64, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27377707

RESUMEN

BACKGROUND: Neurogenic inflammation has for decades been considered an important part of migraine pathophysiology. In the present study, we asked the question if administration of a novel kynurenic acid analogue (SZR72), precursor of an excitotoxin antagonist and anti-inflammatory substance, can modify the neurogenic inflammatory response in the trigeminal ganglion. METHODS: Inflammation in the trigeminal ganglion was induced by local dural application of Complete Freunds Adjuvant (CFA). Levels of phosphorylated MAP kinase pERK1/2 and IL-1ß expression in V1 region of the trigeminal ganglion were investigated using immunohistochemistry and Western blot. FINDINGS: Pretreatment with one dose of SZR72 abolished the CFA-induced pERK1/2 and IL-1ß activation in the trigeminal ganglion. No significant change was noted in case of repeated treatment with SZR72 as compared to a single dose. CONCLUSIONS: This is the first study that demonstrates that one dose of KYNA analog before application of CFA can give anti-inflammatory response in a model of trigeminal activation, opening a new line for further investigations regarding possible effects of KYNA derivates.


Asunto(s)
Antiinflamatorios/uso terapéutico , Adyuvante de Freund/toxicidad , Interleucina-1beta/biosíntesis , Ácido Quinurénico/análogos & derivados , Sistema de Señalización de MAP Quinasas/fisiología , Ganglio del Trigémino/metabolismo , Animales , Antiinflamatorios/farmacología , Regulación de la Expresión Génica , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Ácido Quinurénico/farmacología , Ácido Quinurénico/uso terapéutico , Masculino , Ratas , Ratas Sprague-Dawley , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/patología
15.
Neuropharmacology ; 108: 136-43, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27131920

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have a key role in the control of cellular excitability. HCN2, a subgroup of the HCN family channels, are heavily expressed in small dorsal root ganglia (DRG) neurons and their activation seems to be important in the determination of pain intensity. Intracellular elevation of cAMP levels activates HCN-mediated current (Ih) and small DRG neurons excitability. GPR35, a Gi/o coupled receptor, is highly expressed in small DRG neurons, and we hypothesized that its activation, mediated by endogenous or exogenous ligands, could lead to pain control trough a reduction of Ih current. Patch clamp recordings were carried out in primary cultures of rat DRG neurons and the effects of GPR35 activation on Ih current and neuronal excitability were studied in control conditions and after adenylate cyclase activation with either forskolin or prostaglandin E2 (PGE2). We found that both kynurenic acid (KYNA) and zaprinast, the endogenous and synthetic GPR35 agonist respectively, were able to antagonize the forskolin-induced depolarization of resting membrane potential by reducing Ih-mediated depolarization. Similar results were obtained when PGE2 was used to activate adenylate cyclase and to increase Ih current and the overall neuronal excitability. Finally, we tested the analgesic effect of both GPR35 agonists in an in vivo model of PGE2-induced thermal hyperalgesia. In accord with the hypothesis, both KYNA and zaprinast showed a dose dependent analgesic effect. In conclusion, GPR35 activation leads to a reduced excitability of small DRG neurons in vitro and causes a dose-dependent analgesia in vivo. GPR35 agonists, by reducing adenylate cyclase activity and inhibiting Ih in DRG neurons may represent a promising new group of analgesic drugs.


Asunto(s)
Analgesia/métodos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Ácido Quinurénico/uso terapéutico , Purinonas/uso terapéutico , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/agonistas , Ácido Quinurénico/farmacología , Purinonas/farmacología , Ratas , Ratas Wistar
16.
Curr Pharm Des ; 21(17): 2250-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25557633

RESUMEN

Though migraine and neurodegenerative disorders have a high socioeconomic impact, their therapeutic management has not been fully addressed. Their pathomechanisms are not completely understood, but glutamateinduced excitotoxicity, mitochondrial disturbances and oxidative stress all seem to play crucial roles. The overactivation of glutamate receptors contributes to the hyperexcitability observed in migraine and also to the neurodegenerative process. The kynurenine pathway of the tryptophan metabolism produces the only known endogenous Nmethyl- D-aspartate receptor antagonist, kynurenic acid, which has been proven in different preclinical studies to exert a neuroprotective effect. Influencing the kynurenine pathway might be beneficial in migraine and neurodegenerative diseases, and in the normalization of glutamatergic neurotransmission and the prevention of excitotoxic neuronal damage. The synthesis of kynurenic acid analogues may offer a valuable tool for drug development.


Asunto(s)
Diseño de Fármacos , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/uso terapéutico , Trastornos Migrañosos/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Humanos , Ácido Quinurénico/síntesis química , Ácido Quinurénico/química , Estructura Molecular
17.
Ann Allergy Asthma Immunol ; 113(2): 209-216.e7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24893766

RESUMEN

BACKGROUND: Evidence suggests that intestinal microbiota play an important role in the pathogenesis of atopic dermatitis (AD) through induction of immunosuppression and immune tolerance; however, the exact underlying mechanism is unclear. Few studies to date have examined the effects of probiotics on adult-type AD. OBJECTIVE: To examine the effects of the probiotic Bifidobacterium animalis subsp lactis LKM512 on adult-type AD and the expression of metabolites that are known to be influenced by gut microbiota in fecal samples. METHODS: Forty-four patients were randomly assigned to receive LKM512 or a placebo and underwent medical examinations. Fecal microbiota were analyzed with real-time polymerase chain reaction. Metabolomic analysis was conducted to search for antipruritic metabolites produced by intestinal bacteria using feces derived from 3 patients whose itch scores had improved using capillary electrophoresis with time-of-flight mass spectrometry. Antipruritic effects of kynurenic acid were observed using AD-induced NC/Nga mice. RESULTS: LKM512 administration alleviated itch in AD patients compared with controls and improved the dermatology-specific quality-of-life scores when compared with the controls. Administration of LKM512 also increased the expression of the antipruritic and antinociceptive metabolite kynurenic acid (KYNA) in patients whose itch score had improved after LKM512 treatment. In mouse experiments, scratching behavior counts tended to be decreased by KYNA injection when compared with no treatment. CONCLUSION: LKM512 administration may exert antipruritic effects by increasing KYNA production. LKM512 could therefore be a potentially effective therapeutic candidate for the reduction of pruritus. TRIAL REGISTRATION: umin.ac.jp/ctr Identifier: UMIN000005695.


Asunto(s)
Antipruriginosos/uso terapéutico , Bifidobacterium/inmunología , Dermatitis Atópica/tratamiento farmacológico , Ácido Quinurénico/uso terapéutico , Probióticos/uso terapéutico , Adulto , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Antipruriginosos/farmacología , Heces/microbiología , Femenino , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Intestinos/microbiología , Ácido Quinurénico/farmacología , Ácido Láctico/análogos & derivados , Ácido Láctico/metabolismo , Masculino , Metabolómica , Ratones , Microbiota , Poliaminas/metabolismo , Probióticos/farmacología , Prurito/tratamiento farmacológico , Prurito/microbiología , Calidad de Vida
18.
Ann Neurol ; 75(4): 492-507, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24242287

RESUMEN

OBJECTIVE: Hypoglycemia is a common adverse event and can injure central nervous system (CNS) white matter (WM). We determined whether glutamate receptors were involved in hypoglycemic WM injury. METHODS: Mouse optic nerves (MON), CNS WM tracts, were maintained at 37°C with oxygenated artificial cerebrospinal fluid (ACSF) containing 10mM glucose. Aglycemia was produced by switching to 0 glucose ACSF. Supramaximal compound action potentials (CAPs) were elicited using suction electrodes, and axon function was quantified as the area under the CAP. Amino acid release was measured using high-performance liquid chromatography. Extracellular lactate concentration ([lactate(-)]o) was measured using an enzyme electrode. RESULTS: About 50% of MON axons were injured after 60 minutes of aglycemia (90% after 90 minutes); injury extent was not affected by animal age. Blockade of N-methyl-D-aspartate (NMDA)-type glutamate receptors improved recovery after 90 minutes of aglycemia by 250%. Aglycemic injury was increased by reducing [Mg(2+)]o or increasing [glycine]o , and decreased by lowering pHo , expected results for NMDA receptor-mediated injury. pHo increased during aglycemia due to a drop in [lactate(-)]o. Aglycemic injury was dramatically reduced in the absence of [Ca(2+)]o. Extracellular aspartate, a selective NMDA receptor agonist, increased during aglycemia ([glutamate]o fell). INTERPRETATION: Aglycemia injured WM by a unique excitotoxic mechanism involving NMDA receptors (located primarily on oligodendrocytes). During WM aglycemia, the selective NMDA agonist aspartate is released, probably from astrocytes. Injury is mediated by Ca(2+) influx through aspartate-activated NMDA receptors made permeable by an accompanying alkaline shift in pHo caused by a fall in [lactate(-)]o. These insights have important clinical implications.


Asunto(s)
Leucoencefalopatías/etiología , Traumatismos del Nervio Óptico/etiología , Traumatismos del Nervio Óptico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Ácido Aspártico/metabolismo , Encéfalo/efectos de los fármacos , Calcio/metabolismo , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Ácido Glutámico/metabolismo , Glicina/metabolismo , Glucógeno/metabolismo , Concentración de Iones de Hidrógeno , Hipoglucemia/complicaciones , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/uso terapéutico , Ácido Láctico/metabolismo , Leucoencefalopatías/tratamiento farmacológico , Leucoencefalopatías/metabolismo , Ratones , Ratones Endogámicos C57BL , Traumatismos del Nervio Óptico/tratamiento farmacológico , Quinoxalinas/uso terapéutico , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
19.
Postepy Hig Med Dosw (Online) ; 66: 431-6, 2012 Jun 22.
Artículo en Polaco | MEDLINE | ID: mdl-22922142

RESUMEN

Atherosclerosis together with its cardiovascular consequences is the most common and significant cause of death, particularly in highly developed countries. The process of atherogenesis begins as soon as in childhood and depends on classical risk factors. Atherosclerosis also results from a chronic inflammatory-immune process which takes place in the vascular walls. Furthermore, it has been known for a number of years that the development of atherosclerotic lesions is closely connected with the concentration of homocysteine in serum. Homocysteine is a sulfur amino acid originating from methionine. An increased concentration of homocysteine in blood harmfully influences blood vessels, leading to a higher risk of ischemic heart disease and stroke. Since tackling classical atherosclerosis risk factors is not efficient enough when it comes to protecting the cardiovascular system from diseases, new substances possessing anti-atherogenic properties, especially endogenous ones, are sought. Recently, researchers have paid attention to a connection between homocysteine and an endogenous tryptophan derivative, kynurenic acid. Recently, it was revealed that kynurenic acid counteracts the harmful effects of homocysteine on endothelium cells in vitro. The hypothesis assuming homocysteine-kynurenate interplay suggests the existence of a new mechanism of atherogenesis and gives us an opportunity to use this knowledge in both prevention and treatment of cardiovascular diseases.


Asunto(s)
Hiperhomocisteinemia/tratamiento farmacológico , Ácido Quinurénico/uso terapéutico , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Homocisteína/sangre , Humanos , Hiperhomocisteinemia/complicaciones , Inflamación/complicaciones , Ácido Quinurénico/farmacocinética , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control , Distribución Tisular
20.
J Neural Transm (Vienna) ; 119(2): 151-4, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22231843

RESUMEN

The concentration of kynurenic acid (KYNA) in the cerebrospinal fluid, which is in the nanomolar range, is known to decrease in epilepsy. The experimental data suggest that treatment with L: -KYN dose dependently increases the concentration of the neuroprotective KYNA in the brain, which itself hardly crosses the blood-brain barrier. However, it is suggested that new synthetic KYNA analogs may readily cross the blood-brain barrier. In this study, we tested the hypothesis that a new KYNA analog administered systemically in a sufficient dose results in a decreased population spike activity recorded from the pyramidal layer of area CA1 of the hippocampus, and also provides protection against pentylenetetrazole-induced epileptiform seizures.


Asunto(s)
Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/uso terapéutico , Quinurenina , Pentilenotetrazol/toxicidad , Convulsiones/prevención & control , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Ácido Quinurénico/farmacología , Masculino , Pentilenotetrazol/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA