Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 327
1.
J Clin Invest ; 133(4)2023 02 15.
Article En | MEDLINE | ID: mdl-36787221

Obesity is a risk factor for neurodegenerative disease associated with cognitive dysfunction, including Alzheimer's disease. Low-grade inflammation is common in obesity, but the mechanism between inflammation and cognitive impairment in obesity is unclear. Accumulative evidence shows that quinolinic acid (QA), a neuroinflammatory neurotoxin, is involved in the pathogenesis of neurodegenerative processes. We investigated the role of QA in obesity-induced cognitive impairment and the beneficial effect of butyrate in counteracting impairments of cognition, neural morphology, and signaling. We show that in human obesity, there was a negative relationship between serum QA levels and cognitive function and decreased cortical gray matter. Diet-induced obese mice had increased QA levels in the cortex associated with cognitive impairment. At single-cell resolution, we confirmed that QA impaired neurons, altered the dendritic spine's intracellular signal, and reduced brain-derived neurotrophic factor (BDNF) levels. Using Caenorhabditis elegans models, QA induced dopaminergic and glutamatergic neuron lesions. Importantly, the gut microbiota metabolite butyrate was able to counteract those alterations, including cognitive impairment, neuronal spine loss, and BDNF reduction in both in vivo and in vitro studies. Finally, we show that butyrate prevented QA-induced BDNF reductions by epigenetic enhancement of H3K18ac at BDNF promoters. These findings suggest that increased QA is associated with cognitive decline in obesity and that butyrate alleviates neurodegeneration.


Cognitive Dysfunction , Neurodegenerative Diseases , Mice , Animals , Humans , Quinolinic Acid/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Butyrates , Obesity/drug therapy , Obesity/genetics , Obesity/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Inflammation/complications
2.
Altern Lab Anim ; 51(1): 30-38, 2023 Jan.
Article En | MEDLINE | ID: mdl-36550678

The pathological accumulation of quinolinic acid (QA) is often associated with neuritis and neuronal cell death in several neurodegenerative diseases, through the overproduction of free radicals. Urolithin B and auraptene have been reported to exert potent antioxidant effects - however, little is known about the protective effects of these compounds against QA-induced neurotoxicity. Therefore, this study aimed to explore the in vitro protective effects of urolithin B and auraptene against QA-induced neurotoxicity in the SH-SY5Y neuroblastoma cell line. The MTT assay was used to evaluate cell viability, and flow cytometry was carried out to evaluate effects on the cell cycle and apoptosis. The intracellular levels of reactive oxygen species (ROS) were also determined. Our findings showed that auraptene at non-toxic concentrations had no protective effect on QA-induced toxicity. However, urolithin B at concentrations of 0.6 µM and 2.5 µM enhanced the viability of cells treated with QA. Moreover, while the percentage of apoptotic cells (i.e. in the sub-G1 phase) was shown to significantly increase after QA treatment, pre-treatment with urolithin B reduced the number of these apoptotic cells. Furthermore, urolithin B, as an antioxidant, also significantly reduced QA-induced ROS production. Our findings suggest that urolithin B may possess potent antioxidant and neuroprotective effects against QA-induced neurotoxicity that merit further investigation.


Antioxidants , Neuroblastoma , Humans , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Quinolinic Acid/pharmacology , Cell Line, Tumor , Neuroblastoma/metabolism , Neuroblastoma/pathology , Apoptosis , Cell Survival , Oxidative Stress/physiology
3.
Aging Cell ; 21(10): e13706, 2022 10.
Article En | MEDLINE | ID: mdl-36148631

The effect of a ketogenic diet (KD) on middle aged female mice is poorly understood as most of this work have been conducted in young female mice or diseased models. We have previously shown that an isocaloric KD started at middle age in male mice results in enhanced mitochondrial mass and function after 2 months on diet and improved cognitive behavior after being on diet for 14 months when compared with their control diet (CD) fed counterparts. Here, we aimed to investigate the effect of an isocaloric 2-month KD or CD on healthy 14-month-old female mice. At 16 months of age cognitive behavior tests were performed and then serum, skeletal muscle, cortex, and hippocampal tissues were collected for biochemical analysis. Two months on a KD resulted in enhanced cognitive behavior associated with anxiety, memory, and willingness to explore. The improved neurocognitive function was associated with increased PGC1α protein in the gastrocnemius (GTN) muscle and nuclear fraction. The KD resulted in a tissue specific increase in mitochondrial mass and kynurenine aminotransferase (KAT) levels in the GTN and soleus muscles, with a corresponding decrease in kynurenine and increase in kynurenic acid levels in serum. With KAT proteins being responsible for converting kynurenine into kynurenic acid, which is unable to cross the blood brain barrier and be turned into quinolinic acid-a potent neurotoxin, this study provides a potential mechanism of crosstalk between muscle and brain in mice on a KD that may contribute to improved cognitive function in middle-aged female mice.


Diet, Ketogenic , Animals , Cognition , Female , Kynurenic Acid/metabolism , Kynurenic Acid/pharmacology , Kynurenine/metabolism , Kynurenine/pharmacology , Male , Mice , Muscle, Skeletal/metabolism , Neurotoxins , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Quinolinic Acid/pharmacology
4.
Mol Psychiatry ; 27(9): 3679-3691, 2022 09.
Article En | MEDLINE | ID: mdl-35422466

The tryptophan catabolite (TRYCAT) pathway is implicated in the pathophysiology of schizophrenia (SCZ) since the rate-limiting enzyme indoleamine-dioxygenase (IDO) may be induced by inflammatory and oxidative stress mediators. This systematic review searched PubMed, Web of Science, and Google Scholar for papers published from inception until August 2021 and meta-analyzed the association between SCZ and TRYCATs in the central nervous system (CNS) and peripheral blood. We included 61 studies comprising 2813 patients and 2948 healthy controls. In the CNS we found a significant (p < 0.001) increase in the kynurenine/tryptophan (KYN/TRP) (standardized mean difference, SMD = 0.769, 95% confidence interval, CI: 0.456; 1.082) and kynurenic acid (KA)/KYN + TRP (SMD = 0.697, CI: 0.478-0.917) ratios, KA (SMD = 0.646, CI: 0.422; 0.909) and KYN (SMD = 1.238; CI: 0.590; 1.886), while the 3OH-kynurenine (3HK) + KYN-3-monooxygenase (KMO)/KYN ratio was significantly reduced (SMD = -1.089, CI: -1.682; -0.496). There were significant differences between KYN/TRP, (KYN + KA)/TRP, (3HK + KMO)/KYN, KA, and KYN levels among the CNS and peripheral blood, and among serum and plasma KYN. The only useful peripheral marker of CNS TRYCATs findings was the increased KYN/TRP ratio in serum (SMD = 0.211, CI: 0.056; 0.366, p = 0.007), but not in plasma. There was no significant increase in a neurotoxic composite score based on KYN, 3HK, and picolinic, xanthurenic, and quinolinic acid. SCZ is accompanied by increased IDO activity in the CNS and serum, and reduced KMO activity and a shift towards KA production in the CNS. This CNS TRYCATs profile indicates neuroprotective, negative immunoregulatory and anti-inflammatory effects. Peripheral blood levels of TRYCATs are dissociated from CNS findings except for a modest increase in serum IDO activity.


Kynurenine , Schizophrenia , Humans , Tryptophan/metabolism , Schizophrenia/metabolism , Kynurenic Acid , Quinolinic Acid/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase
5.
PLoS Pathog ; 18(3): e1010366, 2022 03.
Article En | MEDLINE | ID: mdl-35235615

Tryptophan (Trp) metabolism through the kynurenine pathway (KP) is well known to play a critical function in cancer, autoimmune and neurodegenerative diseases. However, its role in host-pathogen interactions has not been characterized yet. Herein, we identified that kynurenine-3-monooxygenase (KMO), a key rate-limiting enzyme in the KP, and quinolinic acid (QUIN), a key enzymatic product of KMO enzyme, exerted a novel antiviral function against a broad range of viruses. Mechanistically, QUIN induced the production of type I interferon (IFN-I) via activating the N-methyl-d-aspartate receptor (NMDAR) and Ca2+ influx to activate Calcium/calmodulin-dependent protein kinase II (CaMKII)/interferon regulatory factor 3 (IRF3). Importantly, QUIN treatment effectively inhibited viral infections and alleviated disease progression in mice. Furthermore, kmo-/- mice were vulnerable to pathogenic viral challenge with severe clinical symptoms. Collectively, our results demonstrated that KMO and its enzymatic product QUIN were potential therapeutics against emerging pathogenic viruses.


Kynurenine 3-Monooxygenase , Virus Diseases , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Interferon Regulatory Factor-3/metabolism , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/metabolism , Mice , Quinolinic Acid/metabolism , Quinolinic Acid/pharmacology , Virus Diseases/drug therapy
6.
Int J Mol Sci ; 22(24)2021 Dec 16.
Article En | MEDLINE | ID: mdl-34948292

A meta-analysis of publicly available transcriptomic datasets was performed to identify metabolic pathways profoundly implicated in the progression and treatment of inflammatory bowel disease (IBD). The analysis revealed that genes involved in tryptophan (Trp) metabolism are upregulated in Crohn's disease (CD) and ulcerative colitis (UC) and return to baseline after successful treatment with infliximab. Microarray and mRNAseq profiles from multiple experiments confirmed that enzymes responsible for Trp degradation via the kynurenine pathway (IDO1, KYNU, IL4I1, KMO, and TDO2), receptor of Trp metabolites (HCAR3), and enzymes catalyzing NAD+ turnover (NAMPT, NNMT, PARP9, CD38) were synchronously coregulated in IBD, but not in intestinal malignancies. The modeling of Trp metabolite fluxes in IBD indicated that changes in gene expression shifted intestinal Trp metabolism from the synthesis of 5-hydroxytryptamine (5HT, serotonin) towards the kynurenine pathway. Based on pathway modeling, this manifested in a decline in mucosal Trp and elevated kynurenine (Kyn) levels, and fueled the production of downstream metabolites, including quinolinate, a substrate for de novo NAD+ synthesis. Interestingly, IBD-dependent alterations in Trp metabolites were normalized in infliximab responders, but not in non-responders. Transcriptomic reconstruction of the NAD+ pathway revealed an increased salvage biosynthesis and utilization of NAD+ in IBD, which normalized in patients successfully treated with infliximab. Treatment-related changes in NAD+ levels correlated with shifts in nicotinamide N-methyltransferase (NNMT) expression. This enzyme helps to maintain a high level of NAD+-dependent proinflammatory signaling by removing excess inhibitory nicotinamide (Nam) from the system. Our analysis highlights the prevalent deregulation of kynurenine and NAD+ biosynthetic pathways in IBD and gives new impetus for conducting an in-depth examination of uncovered phenomena in clinical studies.


Inflammatory Bowel Diseases/metabolism , Kynurenine/metabolism , Nicotinamide N-Methyltransferase/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Colitis/drug therapy , Colitis/metabolism , Humans , Inflammatory Bowel Diseases/drug therapy , Infliximab/pharmacology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Quinolinic Acid/pharmacology , Tryptophan/metabolism
7.
Inorg Chem ; 60(20): 15435-15444, 2021 Oct 18.
Article En | MEDLINE | ID: mdl-34546735

Coordination polymers have emerged as a new class of potent biologically active agents due to a variety of important characteristics such as the presence of bioactive metal centers and linkers, low toxicity, stability, tailorable structures, and bioavailability. The research on intermediate metabolites has also been explored with implications toward the development of selective anticancer, antimicrobial, and antiviral therapeutic strategies. In particular, quinolinic acid (H2quin) is a recognized metabolite in kynurenine pathway and potent neurotoxic molecule, which has been selected in this study as a bioactive building block for assembling a new silver(I) coordination polymer, [Ag(Hquin)(µ-PTA)]n·H2O (1). This product has been prepared from silver oxide, H2quin, and 1,3,5-triaza-7-phosphaadamantane (PTA), and fully characterized by standard methods including single-crystal X-ray diffraction. Compound 1 has revealed distinctive bioactive features, namely (i) a remarkable antiviral activity against herpes simplex virus type 1 (HSV-1) and adenovirus 36 (Ad-36), (ii) a significant antibacterial activity against clinically important bacteria (Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa), and (iii) a selective cytotoxicity against HeLa (human cervix carcinoma) cell line. The present work widens a growing family of bioactive coordination polymers with potent antiviral, antibacterial, and antiproliferative activity.


Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Polymers/pharmacology , Quinolinic Acid/pharmacology , Silver/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , Escherichia coli/drug effects , HeLa Cells , Humans , Microbial Sensitivity Tests , Polymers/chemical synthesis , Polymers/chemistry , Pseudomonas aeruginosa/drug effects , Quinolinic Acid/chemistry , Silver/chemistry , Staphylococcus aureus/drug effects
8.
Int J Mol Sci ; 22(16)2021 Aug 06.
Article En | MEDLINE | ID: mdl-34445176

Due to similar coordination chemistry of palladium and platinum, a large number of palladium compounds as well have been investigated for their anticancer activity. In the present study, we describe synthesis, characterization, and anticancer activity of palladium complex [Bis(1,8-quinolato)palladium (II)], coded as NH3 against seven different cancer cell lines. NH3 is found to have higher antitumor activity than cisplatin against both parent ovarian A2780 cell line and cisplatin-resistant cell lines. Also, NH3 has the lower IC50 value in HT-29 colorectal cancer cell line. The higher antitumor activity of NH3 is due to the presence of bulky 8-Hydroxyquinoline ligand, thus reducing its reactivity. Proteomic study has identified significantly expressed proteins which have been validated through bioinformatics. NH3 has been found to be less toxic than cisplatin at 2.5 mg/kg and 5 mg/kg dosages on mice models. Binary combinations of NH3 with curcumin and epigallocatechin gallate (EGCG) have demonstrated dose and sequence-dependent synergism in ovarian and colorectal cancer models. All of the preclinical studies indicate promising therapeutic potential of NH3 [Bis(1,8-quinolato)palladium (II)] as an anticancer drug.


Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Palladium/chemistry , Palladium/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Humans , Male , Mice , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Interaction Maps/drug effects , Quinolinic Acid/chemical synthesis , Quinolinic Acid/chemistry , Quinolinic Acid/pharmacology
9.
J Cell Mol Med ; 23(9): 6343-6354, 2019 09.
Article En | MEDLINE | ID: mdl-31318159

Protein kinase C (PKC) shows a neuronal protection effect in neurodegenerative diseases. In this study, we test whether berberine has a positive effect on the activity of PKC in quinolinic acid (QA)-induced neuronal cell death. We used intrastriatal injections of QA mice model to test the effect of berberine on motor and cognitive deficits, and the PKC signalling pathway. Treatment with 50 mg/kg b.w of berberine for 2 weeks significantly prevented QA-induced motor and cognitive impairment and related pathologic changes in the brain. QA inhibited the phosphorylation of PKC and its downstream molecules, GSK-3ß, ERK and CREB, enhanced the glutamate level and release of neuroinflammatory cytokines; these effects were attenuated by berberine. We used in vivo infusion of Go6983, a PKC inhibitor to disturb PKC activity in mice brain, and found that the effect of berberine to reverse motor and cognitive deficits was significantly reduced. Moreover, inhibition of PKC also blocked the anti-excitotoxicity effect of berberine, which is induced by glutamate in PC12 cells and BV2 cells, as well as anti-neuroinflammatory effect in LPS-stimulated BV2 cells. Above all, berberine showed neuroprotective effect against QA-induced acute neurotoxicity by activating PKC and its downstream molecules.


Berberine/pharmacology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Protein Kinase C/metabolism , Quinolinic Acid/pharmacology , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Disease Models, Animal , Glutamic Acid/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Neurodegenerative Diseases/chemically induced , Neurons/drug effects , Neurotoxicity Syndromes/metabolism , Signal Transduction/drug effects
10.
Mol Neurobiol ; 56(2): 844-856, 2019 Feb.
Article En | MEDLINE | ID: mdl-29802570

The endocannabinoid system (ECS) actively participates in several physiological processes within the central nervous system. Among such, its involvement in the downregulation of the N-methyl-D-aspartate receptor (NMDAr) through a modulatory input at the cannabinoid receptors (CBr) has been established. After its production via the kynurenine pathway (KP), quinolinic acid (QUIN) can act as an excitotoxin through the selective overactivation of NMDAr, thus participating in the onset and development of neurological disorders. In this work, we evaluated whether the pharmacological inhibition of fatty acid amide hydrolase (FAAH) by URB597, and the consequent increase in the endogenous levels of anandamide, can prevent the excitotoxic damage induced by QUIN. URB597 (0.3 mg/kg/day × 7 days, administered before, during and after the striatal lesion) exerted protective effects on the QUIN-induced motor (asymmetric behavior) and biochemical (lipid peroxidation and protein carbonylation) alterations in rats. URB597 also preserved the structural integrity of the striatum and prevented the neuronal loss (assessed as microtubule-associated protein-2 and glutamate decarboxylase localization) induced by QUIN (1 µL intrastriatal, 240 nmol/µL), while modified the early localization patterns of CBr1 (CB1) and NMDAr subunit 1 (NR1). Altogether, these findings support the concept that the pharmacological manipulation of the endocannabinoid system plays a neuroprotective role against excitotoxic insults in the central nervous system.


Amidohydrolases/drug effects , Corpus Striatum/drug effects , Quinolinic Acid/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Animals , Arachidonic Acids/pharmacology , Corpus Striatum/injuries , Endocannabinoids/pharmacology , Lipid Peroxidation/drug effects , Male , Neostriatum/drug effects , Neostriatum/metabolism , Polyunsaturated Alkamides/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism
11.
Ultrasound Med Biol ; 45(1): 129-136, 2019 01.
Article En | MEDLINE | ID: mdl-30309748

The goal of this study was to test different combinations of acoustic pressure and doses of quinolinic acid (QA) for producing a focal neuronal lesion in the murine hippocampus without causing unwanted damage to adjacent brain structures. Sixty male CD-1 mice were divided into 12 groups that underwent magnetic resonance-guided focused ultrasound at high (0.67 MPa), medium (0.5 MPa) and low (0.33 MPa) acoustic peak negative pressures and received QA at high (0.012 mmol), medium (0.006 mmol) and low (0.003 mmol) dosages. Neuronal loss occurred only when magnetic resonance-guided focused ultrasound with adequate acoustic power (0.67 or 0.5 MPa) was combined with QA. The animals subjected to the highest acoustic power had larger lesions than those treated with medium acoustic power, but two mice had evidence of bleeding. When the intermediate acoustic power was used, medium and high dosages of QA produced lesions larger than those produced by the low dosage.


Brain/pathology , Neurons/pathology , Quinolinic Acid/pharmacology , Ultrasonic Waves , Acoustics , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Pressure
12.
Neurochem Res ; 44(2): 421-427, 2019 Feb.
Article En | MEDLINE | ID: mdl-30523577

Under pathological conditions, nitric oxide can become a mediator of oxidative cellular damage, generating an unbalance between oxidant and antioxidant systems. The participation of neuronal nitric oxide synthase (nNOS) in the neurodegeneration mechanism has been reported; the activation of N-methyl-D-aspartate (NMDA) receptors by agonist quinolinic acid (QUIN) triggers an increase in nNOS function and promotes oxidative stress. The aim of the present work was to elucidate the participation of nNOS in QUIN-induced oxidative stress in knock-out mice (nNOS-/-). To do so, we microinjected saline solution or QUIN in the striatum of wild-type (nNOS +/+), heterozygote (nNOS+/-), and knock-out (nNOS-/-) mice, and measured circling behavior, GABA content levels, oxidative stress, and NOS expression and activity. We found that the absence of nNOS provides a protection against striatal oxidative damage induced by QUIN, resulting in decreased circling behavior, oxidative stress, and a partial protection reflected in GABA depletion. We have shown that nNOS-derived NO is involved in neurological damage induced by oxidative stress in a QUIN-excitotoxic model.


Corpus Striatum/drug effects , Nitric Oxide Synthase Type I/drug effects , Oxidative Stress/drug effects , Quinolinic Acid/pharmacology , Animals , Antioxidants/pharmacology , Corpus Striatum/metabolism , Lipid Peroxidation/drug effects , Male , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/metabolism , Quinolinic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
13.
Domest Anim Endocrinol ; 64: 59-65, 2018 07.
Article En | MEDLINE | ID: mdl-29753195

l-tryptophan (l-Trp), an essential amino acid, is well known as a precursor of 5-hydroxytryptamine (5-HT) and melatonin. In mammals, l-Trp itself has been reported to suppress gastric emptying in mammals. In addition, 5-HT and melatonin are found in the gastrointestinal tract and affect food passage from the digestive tract in mammals. While the function of these factors in mammals is documented, there is little knowledge on their function in the digestive tract of birds. Therefore, the purpose of the present study was to determine if l-Trp and its metabolites affect the crop emptying rate in chicks (Gallus gallus). We also investigated the effects of kynurenic acid (KYNA) and quinolinic acid (QA), which are metabolites of the kynurenine pathway for l-Trp. Oral administration of l-Trp significantly reduced the crop emptying rate in chicks. Among the metabolites, intraperitoneal injection of 5-HT and melatonin significantly reduced the crop emptying rate, whereas KYNA and QA had no effect. The present study suggests that l-Trp, 5-HT, and melatonin inhibit the movement of food in the digestive tract and thereby affect the utilization of nutrients in the diet of chicks.


Chickens , Crop, Avian/drug effects , Gastrointestinal Motility/drug effects , Tryptophan/pharmacology , Administration, Oral , Alanine , Animals , Asparagine , Glycine , Kynurenic Acid/metabolism , Kynurenic Acid/pharmacology , Male , Quinolinic Acid/metabolism , Quinolinic Acid/pharmacology , Tryptophan/administration & dosage
14.
Mol Neurobiol ; 55(2): 1524-1536, 2018 02.
Article En | MEDLINE | ID: mdl-28181189

Evidence suggests that activation of the tryptophan catabolite (TRYCAT) pathway is involved in the pathophysiology of schizophrenia. However, no previous study examined whether TRYCAT pathway activation is associated with deficit schizophrenia. We measured IgA responses to TRYCATs, namely quinolinic acid, picolinic acid, kynurenic acid, xanthurenic acid, and anthranilic acid and 3-OH-kynurenine, in 40 healthy controls and in schizophrenic patients with (n = 40) and without (n = 40) deficit, defined according to the Schedule for the Deficit Syndrome (SDS). Primary deficit schizophrenia is accompanied by an activated TRYCAT pathway as compared to controls and nondeficit schizophrenia. Participants with deficit schizophrenia show increased IgA responses to xanthurenic acid, picolinic acid, and quinolinic acid and relatively lowered IgA responses to kynurenic and anthranilic acids, as compared to patients with nondeficit schizophrenia. Both schizophrenia subgroups show increased IgA responses to 3-OH-kynurenine as compared to controls. The IgA responses to noxious TRYCATs, namely xanthurenic acid, picolinic acid, quinolinic acid, and 3-OH-kynurenine, but not protective TRYCATS, namely anthranilic acid and kunyrenic acid, are significantly higher in deficit schizophrenia than in controls. The negative symptoms of schizophrenia are significantly and positively associated with increased IgA responses directed against picolinic acid and inversely with anthranilic acid, whereas no significant associations between positive symptoms and IgA responses to TRYCATs were found. In conclusion, primary deficit schizophrenia is characterized by TRYCAT pathway activation and differs from nondeficit schizophrenia by a highly specific TRYCAT pattern suggesting increased excitotoxicity, cytotoxicity, and neurotoxicity, as well as inflammation and oxidative stress. The specific alterations in IgA responses to TRYCATs provide further insight for the biological delineation of deficit versus nondeficit schizophrenia.


Immunoglobulin A/metabolism , Inflammation/metabolism , Picolinic Acids/pharmacology , Quinolinic Acid/pharmacology , Schizophrenia/metabolism , Xanthurenates/pharmacology , Adult , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Young Adult
15.
Neurotox Res ; 34(1): 147-163, 2018 07.
Article En | MEDLINE | ID: mdl-29124681

The brain of patients affected by Alzheimer's disease (AD) develops progressive neurodegeneration linked to the formation of proteins aggregates. However, their single actions cannot explain the extent of brain damage observed in this disorder, and the characterization of co-adjuvant involved in the early toxic processes evoked in AD is essential. In this line, quinolinic acid (QUIN) and homocysteine (Hcy) appear to be involved in the AD neuropathogenesis. Herein, we investigate the effects of QUIN and Hcy on early toxic events in cortical neurons and astrocytes. Exposure of primary cortical cultures to these neurometabolites for 24 h induced concentration-dependent neurotoxicity. In addition, QUIN (25 µM) and Hcy (30 µM) triggered ROS production, lipid peroxidation, diminished of Na+,K+-ATPase activity, and morphologic alterations, culminating in reduced neuronal viability by necrotic cell death. In astrocytes, QUIN (100 µM) and Hcy (30 µM) induced caspase-3-dependent apoptosis and morphologic alterations through oxidative status imbalance. To establish specific mechanisms, we preincubated cell cultures with different protective agents. The combined toxicity of QUIN and Hcy was attenuated by melatonin and Trolox in neurons and by NMDA antagonists and glutathione in astrocytes. Cellular death and morphologic alterations were prevented when co-culture was treated with metabolites, suggesting the activation of protector mechanisms dependent on soluble factors and astrocyte and neuron communication through gap junctions. These findings suggest that early damaging events involved in AD can be magnified by synergistic toxicity of the QUIN and Hcy. Therefore, this study opens new possibilities to elucidate the molecular mechanisms of neuron-astrocyte interactions and their role in neuroprotection against QUIN and Hcy.


Astrocytes/drug effects , Cerebral Cortex/cytology , Homocysteine/pharmacology , Neurons/drug effects , Neurotoxins/pharmacology , Quinolinic Acid/pharmacology , Analysis of Variance , Animals , Annexin A5/metabolism , Astrocytes/ultrastructure , Cells, Cultured , Coculture Techniques , Drug Synergism , Embryo, Mammalian , Female , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/ultrastructure , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Pregnancy , Rats , Sodium-Potassium-Exchanging ATPase/metabolism , Thiobarbituric Acid Reactive Substances
16.
Metab Brain Dis ; 33(1): 151-159, 2018 02.
Article En | MEDLINE | ID: mdl-29086235

Increased accumulation of endogenous neurotoxin quinolinic acid has been found in various neurodegenerative diseases. Oxidative stress caused by quinolinic acid is considered as imperative factor for its toxicity. Asiatic acid, a natural triterpene is widely studied for its various medicinal values. In the present study the effects of asiatic acid in preventing the cognitive impairment and oxidative stress caused by quinolinic acid was investigated. Male Spraque-Dawley rats were orally administered asiatic acid (30 mg/kg/day) for 28 days, while quinolinic acid toxicity-induced animals received quinolinic acid (1.5 mmol/kg/day) from day 15 to day 28 for 14 days. Asiatic acid administration prevented the loss of spatial memory caused due to quinolinic acid-induced toxicity as determined using the novel object location test. In addition, asiatic acid administration alleviated the deleterious effect of quinolinic acid in brain such as increased oxidative stress, decreased antioxidant status and mitochondrial oxidative phosphorylation dysfunction. These data demonstrate that asiatic acid through its potent antioxidant and cognition enhancement property prevented the neuronal impairments caused by quinolinic acid.


Cognition/drug effects , Cognitive Dysfunction/prevention & control , Oxidative Stress/drug effects , Pentacyclic Triterpenes/pharmacology , Spatial Memory/drug effects , Animals , Antioxidants/pharmacology , Cognitive Dysfunction/drug therapy , Lipid Peroxidation/drug effects , Male , Neurodegenerative Diseases/drug therapy , Quinolinic Acid/pharmacology , Rats, Sprague-Dawley
17.
Mol Neurobiol ; 55(3): 2214-2226, 2018 03.
Article En | MEDLINE | ID: mdl-28290151

Deficit schizophrenia is accompanied by mucosa-associated activation of the tryptophan catabolite (TRYCAT) pathway, as indicated by increased IgA responses to noxious (NOX) TRYCATs, but not regulatory or protective (PRO) TRYCATs, suggesting increased neurotoxic, excitotoxic, inflammatory, and oxidative potential. No previous studies examined IgM-mediated autoimmune responses to the TRYCAT pathway in deficit versus nondeficit schizophrenia. We measured IgM responses to NOX TRYCATs, namely, quinolinic acid (QA), 3-OH-kynurenine (3HK), picolinic acid (PA), and xanthurenic (XA) acid, and PRO TRYCATs, including kynurenic acid (KA) and anthranilic acid (AA), in 40 healthy controls and 40 deficit and 40 nondeficit schizophrenic patients. We computed the IgM responses to NOX (QA + PA + 3HK + XA)/PRO (AA + KA) ratio and ∆ differences in IgA - IgM TRYCAT values and NOX/PRO ratio. Deficit schizophrenia is characterized by significantly attenuated IgM responses to all TRYCATs and NOX/PRO ratio and highly increased ∆IgA - IgM NOX/PRO ratio as compared to nondeficit schizophrenia and healthy controls. The negative symptoms of schizophrenia are significantly and positively associated with increased IgM responses directed against the KA/3HK ratio and ∆IgA - IgM NOX/PRO ratio. The findings support the view that deficit schizophrenia is a distinct subtype of schizophrenia that may be significantly discriminated from nondeficit schizophrenia. Deficit schizophrenia is accompanied by a highly specific defect in IgM isotype-mediated regulatory responses directed to the TRYCAT pathway. Lowered IgM regulatory responses together with mucosa-derived activation of the TRYCAT pathway may contribute to neuroprogression, negative symptoms, and deficit schizophrenia. All in all, a highly specific defect in the compensatory (anti-)inflammatory reflex system (CIRS), namely, natural IgM-mediated regulatory responses, may underpin deficit schizophrenia.


Immunity, Cellular/immunology , Immunoglobulin M/immunology , Schizophrenia/immunology , Schizophrenic Psychology , Signal Transduction/physiology , Tryptophan/immunology , Adult , Biomarkers/blood , Female , Humans , Immunity, Cellular/drug effects , Immunoglobulin M/blood , Kynurenic Acid/pharmacology , Male , Metabolism/drug effects , Metabolism/physiology , Middle Aged , Quinolinic Acid/pharmacology , Schizophrenia/blood , Schizophrenia/diagnosis , Signal Transduction/drug effects , Tryptophan/blood , Xanthurenates/pharmacology
18.
Neuroscience ; 367: 85-97, 2017 Dec 26.
Article En | MEDLINE | ID: mdl-29031603

Xanthurenic acid (XA), formed from 3-hydroxykynurenine (3-HK) in the kynurenine pathway of tryptophan degradation, may modulate glutamatergic neurotransmission by inhibiting the vesicular glutamate transporter and/or activating Group II metabotropic glutamate receptors. Here we examined the molecular and cellular mechanisms by which 3-HK controls the neosynthesis of XA in rat, mouse and human brain, and compared the physiological actions of 3-HK and XA in the rat brain. In tissue homogenates, XA formation from 3-HK was observed in all three species and traced to a major role of kynurenine aminotransferase II (KAT II). Transamination of 3-HK to XA was also demonstrated using human recombinant KAT II. Neosynthesis of XA was significantly increased in the quinolinate-lesioned rat striatum, indicating a non-neuronal localization of the process. Studies using rat cortical slices revealed that newly produced XA is rapidly released into the extracellular compartment, and that XA biosynthesis can be manipulated experimentally in the same way as the production of kynurenic acid from kynurenine (omission of Na+ or glucose, depolarizing conditions, or addition of 2-oxoacids). The synthesis of XA from 3-HK was confirmed in vivo by striatal microdialysis. In slices from the rat hippocampus, both 3-HK and XA reduced the slopes of dentate gyrus field EPSPs. The effect of 3-HK was reduced in the presence of the KAT inhibitor aminooxyacetic acid. Finally, both 3-HK and XA reduced the power of gamma-oscillatory activity recorded from the hippocampal CA3 region. Endogenous XA, newly formed from 3-HK, may therefore play a physiological role in attentional and cognitive processes.


Brain/cytology , Brain/metabolism , Kynurenine/analogs & derivatives , Xanthurenates/chemistry , Xanthurenates/metabolism , Aged , Animals , Brain/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Fluoroquinolones/pharmacology , Glucose/metabolism , Glutamine/pharmacology , Humans , In Vitro Techniques , Kynurenine/metabolism , Kynurenine/pharmacology , Male , Mice , Middle Aged , Piperazines/pharmacology , Postmortem Changes , Pyruvic Acid/metabolism , Quinolinic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tacrolimus/analogs & derivatives , Tacrolimus/metabolism , Temperature , Tissue Distribution/drug effects , Tissue Distribution/physiology , Transaminases/metabolism , Veratridine/metabolism
19.
Mult Scler Relat Disord ; 14: 60-67, 2017 May.
Article En | MEDLINE | ID: mdl-28619434

BACKGROUND: Quinolinic acid (QA) is a product of tryptophan degradation and its pathologic accumulation has been found to induce neuroinflammatory and demyelinating diseases such as multiple sclerosis via excessive free radicals generation. Recent studies showed that Terminalia chebula has several pharmacological effects such as antioxidant, anti-inflammatory and neuroprotective properties. The aim of this study was evaluation of the protective effect of T. chebula alcoholic extract (TCAE) on oxidative PC12 and OLN-93 cells death induced by QA. METHODS: The cells were pretreated with TCAE (6.25-50µg/mL) for 2h and then subjected to QA (8mM) for 24h. Cell viability and the parameters of redox status including the levels of intracellular reactive oxygen species (ROS), lipid peroxidation and oxidative DNA damage were measured using 2-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT), 2,7-dicholorofluorecin diacetate (DCF-DA), thiobarbituric acid and comet assays, respectively. RESULTS: Based on Folin-Ciocalteu method, the total phenolic compounds in TCAE were estimated about 1.18%. TCAE at concentration ranges of 6.25-50µg/mL had no toxic effect on cell viability (p>0.05). Treatment with TCAE significantly increased cell viability following QA insult at concentrations above 25µg/mL (p<0.01). Cytoprotective potential of TCAE also ameliorated ROS accumulation, lipid peroxidation and DNA damage induced by QA. CONCLUSION: These data suggest that TCAE exhibits neuroprotection and oligoprotection potential by means of alleviating oxidative stress parameters.


Cell Death/drug effects , DNA Damage/drug effects , Lipid Peroxidation/drug effects , Oligodendroglia/drug effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Quinolinic Acid/pharmacology , Terminalia , Animals , Cell Line , Oligodendroglia/metabolism , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
20.
Dev Neurosci ; 39(5): 399-412, 2017.
Article En | MEDLINE | ID: mdl-28490020

Maternal inflammation has been linked to neurodevelopmental and neuropsychiatric disorders such as cerebral palsy, schizophrenia, and autism. We had previously shown that intrauterine inflammation resulted in a decrease in serotonin, one of the tryptophan metabolites, and a decrease in serotonin fibers in the sensory cortex of newborns in a rabbit model of cerebral palsy. In this study, we hypothesized that maternal inflammation results in alterations in tryptophan pathway enzymes and metabolites in the placenta and fetal brain. We found that intrauterine endotoxin administration at gestational day 28 (G28) resulted in a significant upregulation of indoleamine 2,3-dioxygenase (IDO) in both the placenta and fetal brain at G29 (24 h after treatment). This endotoxin-mediated IDO induction was also associated with intense microglial activation, an increase in interferon gamma expression, and increases in kynurenine and the kynurenine pathway metabolites kynurenine acid and quinolinic acid, as well as a significant decrease in 5-hydroxyindole acetic acid (a precursor of serotonin) levels in the periventricular region of the fetal brain. These results indicate that maternal inflammation shunts tryptophan metabolism away from the serotonin to the kynurenine pathway, which may lead to excitotoxic injury along with impaired development of serotonin-mediated thalamocortical fibers in the newborn brain. These findings provide new targets for prevention and treatment of maternal inflammation-induced fetal and neonatal brain injury leading to neurodevelopmental disorders such as cerebral palsy and autism.


Brain/metabolism , Inflammation/metabolism , Placenta/metabolism , Tryptophan/metabolism , Animals , Brain/embryology , Female , Indoles/pharmacology , Inflammation/chemically induced , Pregnancy , Quinolinic Acid/pharmacology , Rabbits , Serotonin/metabolism
...