Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Eur Rev Med Pharmacol Sci ; 28(8): 3085-3098, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38708467

ABSTRACT

OBJECTIVE: Dysgeusia is characterized by a loss of taste perception, leading to malnutrition. This situation affects inflammatory conditions such as respiratory and neurological conditions, obesity, cancer, chemotherapy, aging, and many others. To date, there is not much information on the prevalence and risk of dysgeusia in an inflammatory condition; also, it is unclear which flavor is altered. MATERIALS AND METHODS: We systematically searched three databases from January 2018 to January 2023. Participants were children, adults, or elderly persons with an inflammatory condition and evaluated taste loss. A random effects model was used for statistical analysis to calculate the pooled odds ratio with its corresponding 95.0% confidence interval to estimate the probability of taste alteration (dysgeusia) in an inflammatory condition. RESULTS: The data allowed us to conduct a systematic review, including 63 original articles and 15 studies to perform the meta-analysis. The meta-analysis indicated a heterogenicity of 84.7% with an odds ratio of 3.25 (2.66-3.96), indicating a significant risk of Alzheimer's disease, SARS-CoV-2, chemotherapy, and rhinosinusitis. CONCLUSIONS: Inflammatory conditions and taste alterations are linked. Dysgeusia is associated with a higher risk of malnutrition and poorer general health status, especially in vulnerable populations.


Subject(s)
Dysgeusia , Inflammation , Taste Perception , Humans , Dysgeusia/epidemiology , COVID-19/epidemiology , Alzheimer Disease/epidemiology , Taste/physiology , Malnutrition/epidemiology , SARS-CoV-2
2.
Behav Neurol ; 2021: 6651492, 2021.
Article in English | MEDLINE | ID: mdl-33833828

ABSTRACT

Symptoms of depressive disorders such as anhedonia and despair can be a product of an aberrant adaptation to stress conditions. Chronic unpredictable stress model (CUS) can generate an increase in the activity of the hypothalamic-pituitary-adrenal axis (HPA) and induce a reduction of neurotrophin signaling and the proliferation of neural progenitors in the adult dentate gyrus, together with increased oxidative stress. Levels of the endocannabinoid anandamide (AEA) seem to affect these depression-by-stress-related features and could be modulated by fatty acid amide hydrolase (FAAH). We aimed to evaluate the effects of FAAH inhibitor, URB597, on depressive-like behavior and neural proliferation of mice subjected to a model of CUS. URB597 was administered intraperitoneally at a dose of 0.2 mg/kg for 14 days after CUS. Depressive-like behaviors, anhedonia, and despair were evaluated in the splash and forced swimming tests, respectively. Alterations at the HPA axis level were analyzed using the relative weight of adrenal glands and serum corticosterone levels. Oxidative stress and brain-derived neurotrophic factor (BDNF) were also evaluated. Fluorescence immunohistochemistry tests were performed for the immunoreactivity of BrdU and Sox2 colabeling for comparison of neural precursors. The administration of URB597 was able to reverse the depressive-like behavior generated in mice after the model. Likewise, other physiological responses associated with CUS were reduced in the treated group, among them, increase in the relative weight of the adrenal glands, increased oxidative stress, and decreased BDNF and number of neural precursors. Most of these auspicious responses to enzyme inhibitor administration were blocked by employing a cannabinoid receptor antagonist. In conclusion, the chronic inhibition of FAAH generated an antidepressant effect, promoting neural progenitor proliferation and BDNF expression, while reducing adrenal gland weight and oxidative stress in mice under the CUS model.


Subject(s)
Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Amidohydrolases , Animals , Cell Proliferation , Corticosterone , Dentate Gyrus , Disease Models, Animal , Mice , Stress, Psychological/drug therapy
3.
Neurosci Lett ; 742: 135534, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33271195

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder, caused by the selective death of dopaminergic neurons in the substantia nigra pars compacta. ß-caryophyllene (BCP) is a phytocannabinoid with several pharmacological properties, producing anti-inflammatory and antihypertensive effects. In addition, BCP protects dopaminergic neurons from neuronal death induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), yet it remains unclear if this effect is due to its antioxidant activity. To assess whether this is the case, the effect of BCP on the expression and activity of NAD(P)H quinone oxidoreductase (NQO1) was evaluated in mice after the administration of MPTP. Male C57BL/6 J mice were divided into four groups, the first of which received saline solution i.p. in equivalent volume and served as a control group. The second group received MPTP. The second group received MPTP hydrochloride (5 mg/kg, i.p.) daily for seven consecutive days. The third group received BCP (10 mg/kg) for seven days, administered orally and finally, the fourth group received MPTP as described above and BCP for 7 days from the fourth day of MPTP administration. The results showed that BCP inhibits oxidative stress-induced cell death of dopaminergic neurons exposed to MPTP at the same time as it enhances the expression and enzymatic activity of NQO1. Also, the BCP treatment ameliorated motor dysfunction and protected the dopaminergic cells of the SNpc from damage induced by MPTP. Hence, BCP appears to achieve at least some of its antioxidant effects by augmenting NQO1 activity, which protects cells from MPTP toxicity. Accordingly, this phytocannabinoid may represent a promising pharmacological option to safeguard dopaminergic neurons and prevent the progression of PD.


Subject(s)
Antioxidants/therapeutic use , MPTP Poisoning/metabolism , MPTP Poisoning/prevention & control , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Polycyclic Sesquiterpenes/therapeutic use , Animals , Antioxidants/pharmacology , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred C57BL , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , Polycyclic Sesquiterpenes/pharmacology , Random Allocation
5.
Neurologia (Engl Ed) ; 34(3): 143-152, 2019 Apr.
Article in English, Spanish | MEDLINE | ID: mdl-28104279

ABSTRACT

INTRODUCTION: Parkinson's disease (PD) is a neurodegenerative disorder characterised by balance problems, muscle rigidity, and slow movement due to low dopamine levels and loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The endocannabinoid system is known to modulate the nigrostriatal pathway through endogenous ligands such as anandamide (AEA), which is hydrolysed by fatty acid amide hydrolase (FAAH). The purpose of this study was to increase AEA levels using FAAH inhibitor URB597 to evaluate the modulatory effect of AEA on dopaminergic neuronal death induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). METHODS: Our study included 4 experimental groups (n = 6 mice per group): a control group receiving no treatment, a group receiving URB597 (0.2mg/kg) every 3 days for 30 days, a group treated with MPTP (30mg/kg) for 5 days, and a group receiving URB597 and subsequently MPTP injections. Three days after the last dose, we conducted a series of behavioural tests (beam test, pole test, and stride length test) to compare motor coordination between groups. We subsequently analysed immunoreactivity of dopaminergic cells and microglia in the SNpc and striatum. RESULTS: Mice treated with URB597 plus MPTP were found to perform better on behavioural tests than mice receiving MPTP only. According to the immunohistochemistry study, mice receiving MPTP showed fewer dopaminergic cells and fibres in the SNpc and striatum. Animals treated with URB597 plus MPTP displayed increased tyrosine hydroxylase immunoreactivity compared to those treated with MPTP only. Regarding microglial immunoreactivity, the group receiving MPTP showed higher Iba1 immunoreactivity in the striatum and SNpc than did the group treated with URB597 plus MPTP. CONCLUSION: Our results show that URB597 exerts a protective effect since it inhibits dopaminergic neuronal death, decreases microglial immunoreactivity, and improves MPTP-induced motor alterations.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Amidohydrolases/metabolism , Dopaminergic Neurons/drug effects , Substantia Nigra/drug effects , Animals , Benzamides , Carbamates , Disease Models, Animal , Dopaminergic Neurons/pathology , Male , Mice , Mice, Inbred C57BL , Motor Skills/drug effects , Neuroprotective Agents/therapeutic use , Parkinson Disease , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase
6.
Neurologia ; 28(4): 212-8, 2013 May.
Article in English, Spanish | MEDLINE | ID: mdl-22703630

ABSTRACT

INTRODUCTION: Nicotinic acetylcholine receptors (nAChRs) are widely expressed throughout several brain regions. Formation of the α4ß2 and α7 subtypes in particular is involved in the organisation of different types of memory. Furthermore, due to their location, these receptors can control the release of various types of neurotransmitters and contribute to synaptic plasticity. METHODS: Rats were divided into three groups, an experimental group (E), a sham-operated group, (S) and an intact group (T). In group E, stereotactic guidance was used to induce a chemical lesion with 1 µ/µL of 5,7-dihydroxytryptamine (5,7-DHT) in the anteroventral part of the dorsal raphe nucleus (DRN). In the sham-operated group (S), animals underwent surgery including delivery of the same excipient solution to the same site. The intact group (T) received no treatment whatsoever. Twenty days after surgery, animals in all groups were euthanised by decapitation to evaluate the expression of α4 and α7 nAChRs by means of molecular biology techniques. RESULTS: 5-HT denervation of the rat PFC differentially modified the expression of α4 and α7 receptors: while α4 receptor expression increased, α7 expression decreased. CONCLUSION: Expression differences observed between the two subtypes may be due to their separate locations. The α4 subtype is found in postsynaptic locations and may be related to adaptive changes in postsynaptic cells, while the location of α7 is presynaptic. This explains why the lesion and the elimination of 5-HT fibres in the CPF would cause a decrease in α7 expression.


Subject(s)
Prefrontal Cortex/physiology , Receptors, Nicotinic/biosynthesis , Serotonergic Neurons/physiology , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , 5,7-Dihydroxytryptamine/toxicity , Animals , Denervation , Female , Memory/physiology , Neuronal Plasticity/drug effects , Polymerase Chain Reaction , Prefrontal Cortex/drug effects , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Serotonergic Neurons/drug effects , Serotonin Agents/toxicity , alpha7 Nicotinic Acetylcholine Receptor/drug effects
7.
Neurología (Barc., Ed. impr.) ; 27(5): 301-310, jun. 2012. ilus, tab
Article in Spanish | IBECS | ID: ibc-102008

ABSTRACT

Introducción: Realizar una revisión de la fisiología de las subunidades del receptor a glutamato tipo N-metil-D-aspartato (NMDA). Desarrollo: El acido glutámico (Glu) es el principal neurotransmisor excitador del sistema nervioso central la cual interactúa con dos tipos de receptores clasificados como: metabotrópicos y ionotrópicos. Los receptores ionotrópicos se dividen de acuerdo a la afinidad de sus agonistas específicos en: N-metil-D-aspartato (NMDA), ácido α-amino-3-hidroxi-5-metil-4-isoxazol (AMPA) y acido kaínico (KA). Los receptores NMDA son estructuras macromoleculares que se forman por combinaciones de diferentes subunidades: NMDAR1 (NR1), NMDAR2 (NR2) y (NR3). Conclusiones:El estudio de este receptor ha sido de gran interés por la función que desempeña en la plasticidad sináptica, pero sobre todo por la permeabilidad que tiene para el ion Ca++. En esta revisión se analiza la composición molecular del receptor NMDA, así como las distintas variantes de edición de la subunidad NR1 que en asociación con la subunidad NR2 forman el principal dímero de este receptor. La composición, estructura y funcionalidad y sus distintos patrones de expresión tanto temporal y espacial, ha permitido conocer la versatilidad y la diversidad funcional tanto de las diferentes isoformas de la subunidad NR1, así como las distintas propiedades farmacológicas de la subunidad NR2 (AU)


Introducion: To review the physiology of the glutamate receptor subunits such as N-methyl-D-aspartate (NMDA). Development:Glutamic acid (Glu) is the major excitatory neurotransmitter in the central nervous system which interacts with two types classified into two types: metabotropic and ionotropic. Ionotropic receptors are classified according to the affinity of their specific agonists: N-methyl-D-aspartate (NMDA), α-amino acid-3-hydroxy-5-methyl-4-isoxazole (AMPA) and kainic acid (KA). NMDA receptors are macromolecular structures that are formed by different combinations of subunits, NMDAR1 (NR1), NMDAR2 (NR2) and NMDAR3 (NR3). Conclusions: The study of this receptor has been of great interest due to its role in synaptic plasticity, but mainly due to the permeability it has to Ca++ ion. This review examines the molecular composition of NMDA receptor and the variants of NR1 subunit edition in association with NR2 subunit dimer, the main form of this receptor. The composition, structure and function and their distinct expression patterns in both time and space, has shown the versatility and diversity of functionally different isoforms of the NR1 subunit and various pharmacological properties of the NR2 subunit (AU)


Subject(s)
Humans , Receptors, Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Neuronal Plasticity/physiology , Neurotoxins/analysis , Glutamic Acid/pharmacokinetics , Electrophysiology/methods , Alzheimer Disease/physiopathology , Huntington Disease/physiopathology
8.
Neurologia ; 27(5): 261-7, 2012 Jun.
Article in Spanish | MEDLINE | ID: mdl-22217522

ABSTRACT

INTRODUCTION: In cirrhosis some toxic substances accumulate in brain and modify the expression of several neuronal receptors. Thus, the use of medicinal plants such as Rosmarinus officinalis L. has been proposed in several pathologies due to its hepatoprotective, antioxidant and neuroprotective activity. In this study we evaluated the expression of the subunits NR1, NR2A and NR2B of the glutamate receptor in rat prefrontal cortex in a model of hepatic damage induced with carbon tetrachloride after a treatment with Rosmarinus officinalis L. METHODS: We used a total of 24 male Wistar rats weighing 80-90 g. body weight. We formed three study groups: control group (C) without a treatment, carbon tetrachloride group (CC14), and CC14 group plus Rosmarinus officinalis L (CCl4+ROM; 1.5 g/kg of extract orally). RESULTS: The expression of the NR1, NR2A and NR2B subunits in cirrhotic animals increased compared to the control group, however treatment with Rosmarinus officinalis L. was able to reduce this expression to normal levels compared with CC14 and CCl4+ROM groups. These results could be due to an improvement in hepatic function. CONCLUSION: Treatment with extract of Rosmarinus officinalis L. in cirrhotic animals modifies the expression of subunits of the NMDA receptor due to an improvement in hepatocellular function in the presence of antioxidant compounds and flavonoids.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Liver Diseases/metabolism , Plant Extracts/administration & dosage , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Rosmarinus , Animals , Carbon Tetrachloride/administration & dosage , Male , Rats , Rats, Wistar
9.
Neurologia ; 27(5): 301-10, 2012 Jun.
Article in Spanish | MEDLINE | ID: mdl-22217527

ABSTRACT

INTRODUCTION: To review the physiology of the glutamate receptor subunits such as N-methyl-D-aspartate (NMDA). DEVELOPMENT: Glutamic acid (Glu) is the major excitatory neurotransmitter in the central nervous system which interacts with two types classified into two types: metabotropic and ionotropic. Ionotropic receptors are classified according to the affinity of their specific agonists: N-methyl-D-aspartate (NMDA), α-amino acid-3-hydroxy-5-methyl-4-isoxazole (AMPA) and kainic acid (KA). NMDA receptors are macromolecular structures that are formed by different combinations of subunits, NMDAR1 (NR1), NMDAR2 (NR2) and NMDAR3 (NR3) CONCLUSIONS: The study of this receptor has been of great interest due to its role in synaptic plasticity, but mainly due to the permeability it has to Ca(++) ion. This review examines the molecular composition of NMDA receptor and the variants of NR1 subunit edition in association with NR2 subunit dimer, the main form of this receptor. The composition, structure and function and their distinct expression patterns in both time and space, has shown the versatility and diversity of functionally different isoforms of the NR1 subunit and various pharmacological properties of the NR2 subunit.


Subject(s)
Receptors, N-Methyl-D-Aspartate/physiology , Structure-Activity Relationship
10.
J Neuroimmunol ; 238(1-2): 12-8, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21807419

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF-1α) is a master transcription factor that regulates the response to hypoxia and ischemia and induces the expression of various genes, including vascular endothelial growth factor (VEGF) and erythropoietin (EPO). This study shows the systemic response of increased HIF-1α, EPO, and VEGF mRNA and protein. In addition, VEGF expression was increased in neurons and over-expressed in glial cells in a model of neuroexcitotoxicity in the hippocampus, in which rats were neonatally exposed to high glutamate concentrations. Simultaneous increases in HIF-1α, EPO and VEGF mRNA in peritoneal macrophages were also observed. Our study is consistent with the hypothesis that these genes exert a protective effect in response to neurotoxicity.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/metabolism , Neurotoxicity Syndromes/pathology , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/toxicity , Hippocampus/drug effects , Hippocampus/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Macrophages/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxins/toxicity , Pregnancy , RNA, Messenger , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Clin Biochem ; 43(3): 234-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19804771

ABSTRACT

OBJECTIVES: HIF-1 alpha (hypoxia-inducible factor-1 alpha) mediates the responses of mammalian cells to hypoxia/ischemia by inducing the expression of adaptive gene products (e.g., vascular endothelial growth factor (VEGF) and erythropoietin (EPO)). Persistent pulmonary hypertension of the newborn (PPHN) and cyanotic congenital heart disease (CCHD) are common neonatal diseases considered as paradigms of hypoxemia. Since the expression HIF-1 alpha, VEGF and EPO in newborns diagnosed with these diseases has yet to be studied, we set out to define the expression of these genes in peripheral blood from newborn infants diagnosed with PPHN and CCHD. DESIGN AND METHODS: The mRNA transcripts encoding HIF-1 alpha, VEGF and EPO were measured by RT-PCR in healthy newborn infants and infants diagnosed with PPHN and CCHD. RESULTS: An important increase in HIF-1 alpha expression was observed in both pathological conditions, accompanied by significant increases in VEGF and EPO expression when compared to healthy infants. CONCLUSIONS: HIF-1 alpha mRNA expression increases in newborn infants with PPHN or CCHD, as does the expression of its target genes VEGF and EPO.


Subject(s)
Erythropoietin , Heart Diseases , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Persistent Fetal Circulation Syndrome , Vascular Endothelial Growth Factor A , Erythropoietin/blood , Erythropoietin/genetics , Heart Diseases/blood , Heart Diseases/congenital , Heart Diseases/physiopathology , Humans , Hypoxia/blood , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/blood , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Infant , Infant, Newborn , Persistent Fetal Circulation Syndrome/blood , Persistent Fetal Circulation Syndrome/genetics , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics
12.
Int J Dev Neurosci ; 27(2): 197-204, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18930801

ABSTRACT

Overactivation of NMDA-Rs may mediate excitotoxic cell death associated with epileptic seizures, and hypoxic-ischemic conditions. We assessed whether repeated subcutaneous administration of l-glutamate to neonatal rats affects the subunit composition of NMDA-Rs. Accordingly, cortical and hippocampal tissue from 14-day-old rats was analyzed by Western blotting and RT-PCR to quantify the protein and mRNA expression of different NMDA-R subunits. In addition, tissue sections were Nissl stained to assess the cell damage in this tissue. Early exposure of neonatal rats to L-glutamate differentially affects the expression of mRNA transcripts for NMDA-R subunits in the cerebral cortex and hippocampus. In the cerebral cortex, a decrease in NR2B subunit mRNA expression was observed, as well as a loss of NR1 and NR2A protein. By contrast, neonatal L-glutamate administration augmented the transcripts encoding the NR1, NR2B, and NR2C subunits in the hippocampal formation. The expression of mRNA encoding the NR2A subunit was not affected by neonatal L-glutamate administration in either of the brain regions examined. This differential expression of NMDA-R subunits following neonatal exposure to L-glutamate may represent an adaptive response of the glutamate receptors to overactivation in order to reduce the effect of high L-glutamate during the early period of life when the animal is more vulnerable to excitotoxicity.


Subject(s)
Glutamic Acid/toxicity , Hippocampus/drug effects , Neurotoxins/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Analysis of Variance , Animals , Blotting, Western , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Gene Expression/drug effects , Hippocampus/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
Int J Dev Neurosci ; 26(5): 487-95, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18396383

ABSTRACT

Pro-inflammatory cytokines TNF-alpha, IL-1beta and IL-6 rises significantly during neuronal damage and activate the signaling p38 MAPK pathway, which is involved in the apoptotic (AP) neuronal death. Systemic administration of glutamate as monosodium salt (MSG) to newborn animals induces neuronal death, however whether neurons die by AP or necrosis through MAPK p38 pathway activation it is unknown. In this study, TNF-alpha, IL-1beta and IL-6 expression levels, AP neuronal death and cellular type that produces TNF-alpha was also identified in the cerebral cortex (CC) and striatum (St) of rats at 8, 10, and 14 days of age after neonatal exposure to MSG. TNF-alpha production and AP neuronal death was significantly increased in the CC at PD8-10, and in the St in all ages studied by excitotoxicity effect induced with MSG. This effect was completely inhibited by SB203580 (p38 inhibitor) in both regions studied. TNF-alpha, IL-1beta and IL-6 RNAm increased after MSG administration, whereas SB203580 did not modify their expression. These data indicates that neuronal death induced by excitotoxicity appears to be mediated through p38 signaling pathway activated by TNF-alpha and their inhibition may have an important neuroprotective role as part of anti-inflammatory therapeutic strategy.


Subject(s)
Cytokines/genetics , Sodium Glutamate/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Expression/drug effects , Imidazoles/pharmacology , Immunohistochemistry , Injections, Subcutaneous , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pregnancy , Pyridines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sodium Glutamate/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
Int J Dev Neurosci ; 24(6): 401-10, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16843632

ABSTRACT

Sparteine is a quinolizidine alkaloid (QA) produced by Lupine species that has generated much interest due to its anti-hypertensive, anti-pyretic, and anti-inflammatory properties. In the nervous system, sparteine has been shown to display anti-cholinergic and depressive activity, although how sparteine exerts its toxic effects in the brain remains unclear. We have addressed this issue by administering subcutaneous injections of sparteine (25 mg/kg of body weight) to rats on postnatal days 1 and 3, and then examining the expression of the muscarinic acetylcholine receptor (mAChR) subunits m1-m4 in the brains of the neonatal rats 14-60 days later. Administration of sparteine to neonatal rats caused neuronal damage in the cerebral motor cortex accompanied by transient changes in the expression of m1-m4 mAChR subunits as revealed by both RT-PCR and Western blotting. This effect could be prevented by pre-treatment with atropine (10 mg/kg) 1 h prior to the injection of sparteine, suggesting that the cytotoxic activity of sparteine is mediated through mAChRs.


Subject(s)
Cerebral Cortex , Neurons , Protein Subunits/metabolism , Receptors, Muscarinic/metabolism , Sparteine/toxicity , Animals , Animals, Newborn , Cell Shape , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Female , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pregnancy , Protein Subunits/genetics , Rats , Rats, Wistar , Receptors, Muscarinic/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sparteine/administration & dosage , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Neurosci Lett ; 403(3): 233-8, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16782274

ABSTRACT

Excitotoxic neuronal death occurs through the activation of NMDA and non-NMDA glutamatergic receptors in the CNS. Glutamate also induces strong activation of p38 and indeed, cell death can be prevented by inhibitors of the p38 pathway. Furthermore, intracellular signals generated by AMPA receptors activate the stress sensitive MAP kinases implicated in apoptotic neuronal death, such as JNK and p38. To investigate the relationship between these elements, we have used immunohistochemistry to analyze the expression of GluR2 in the cerebral cortex of postnatal rats (postnatal Day [PD] 8 and 14) after administering them with monosodium glutamate (MSG; 4 mg/g body weight on PD1, 3, 5, and 7). Similarly, the expression of REST, Fas-L and Bcl-2 mRNA transcripts in animals exposed to a p38 inhibitor, SB203580 (0.42 microg/g body weight, administered subcutaneously) was determined by reverse transcriptase-PCR. The enhanced GluR2-expression in the cerebral cortex at PD8 and the down regulation of this receptor at PD14 was correlated with neuronal damage induced by excitotoxicity. In addition, the enhanced expression of REST at PD8 and PD14 suggests that the induction of REST transcription contributes to glutamate-induced excitotoxic neurodegeneration, possibly by modulating GluR2 expression. Fas-L and Bcl-2 over expression at PD8 and their subsequent down regulation at PD14 also suggests that Fas-L could be the direct effector of apoptosis in the cerebral cortex. On the other hand, the presence of Bcl-2 at PD8 could attenuate certain survival signals in neurons under these neurotoxic conditions. Thus, a change in glutamate receptor composition, and enhanced Fas-L and Bcl-2 expression, coupled with activation of the p38/SAPK pathway appear to be events involved in the neuronal apoptosis induced under neurotoxic conditions.


Subject(s)
Cerebral Cortex/metabolism , Glutamic Acid/physiology , Neurons/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Cell Death , Enzyme Activation , Fas Ligand Protein , Female , Glutamic Acid/toxicity , Immunohistochemistry , Membrane Glycoproteins/biosynthesis , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Receptors, AMPA/biosynthesis , Sodium Glutamate/toxicity , Tumor Necrosis Factors/biosynthesis , fas Receptor/biosynthesis
16.
Neurochem Int ; 47(7): 491-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16099549

ABSTRACT

This paper served to evaluate the expression levels of subunits NR1, NR2A and NR2C which are implicated in neuronal plasticity events. A 50% (right half) 4 mm longitudinal resection of the spinal cord was done at the C5-C6 level with preservation of the anterior spinal artery. This was effected in a dog model after either a homologous transplant or a pure spinal cord section. In this study we used two groups of dogs with four individuals each, as well as a control group. The transplant group (n=4) was analyzed at days 3 and 28 post surgery. The section group (n=4) was also analyzed at days 3 and 28 post op. All three groups (transplant, section and control) were evaluated as to the subunit expression in each of the segments corresponding to the transplanted or sectioned sites, the site contralateral to the transplanted or sectioned sites at levels half a centimeter both proximal and distal to the site of transplant and section. The results showed a variety of changes in each of the subunits depending on the group, the segment and the time of evaluation (acute versus chronic). This could be closely related to mechanisms which participate in regeneration and functional recuperation.


Subject(s)
Receptors, N-Methyl-D-Aspartate/biosynthesis , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Spinal Cord/transplantation , Animals , Dogs , Male , Neuronal Plasticity/physiology , RNA/analysis , RNA/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
J Neuroimmunol ; 165(1-2): 53-62, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15972237

ABSTRACT

The proinflammatory cytokines TNF-alpha, IL-1beta, and IL-6 rise during neuronal damage and activate the apoptotic mitogen-activated protein kinase p38. We studied apoptosis, the levels of TNF-alpha, IL-1beta, and IL-6, and the cell type producing TNF-alpha in rats at 8, 10, and 14 days of age after neonatal exposure to glutamate, which induces neuronal damage. TNF-alpha production was significantly increased by glutamate, but inhibited by SB203580 (a p38 inhibitor). TNF-alpha, IL-1beta, and IL-6 mRNA levels increased, but SB203580 did not modify their expression. Thus, the p38 signaling pathway influences the expression of inflammatory genes and its inhibition may offer anti-inflammatory therapy.


Subject(s)
Apoptosis/immunology , Cytokines/metabolism , Hippocampus/enzymology , Hippocampus/pathology , Inflammation Mediators/metabolism , Sodium Glutamate/toxicity , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Cytokines/biosynthesis , Cytokines/physiology , Hippocampus/immunology , Hippocampus/metabolism , Imidazoles/administration & dosage , Inflammation Mediators/physiology , Injections, Subcutaneous , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Neuroglia/immunology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Pyridines/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/immunology , Sodium Glutamate/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Neurosci Lett ; 321(1-2): 9-12, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11872244

ABSTRACT

Monosodium glutamate (MSG) was administered subcutaneously to male neonate rats, and the effects on N-methyl-D-asparatate (NMDA) subunit receptor types NR2C and NR2D from different brain regions were studied. A semi-quantitative reverse transcription-polymerase chain reaction was used to measure NR2C and NR2D expression levels in the cerebral cortex, hippocampus and striatum. MSG treatment (4 mg/g body weight, on postnatal days 1, 3, 5, and 7) produced an important increase of NR2C and NR2D subunit gene expression levels in the hippocampus and striatum of adults rats. No change was observed in the cerebral cortex. We propose that an early excessive activation of glutamate receptors could modify NMDA subunit expression and its structural composition on postnatal development. This, as part of a compensatory response by an altered neuronal circuitry, mainly in the hippocampus and striatum, suggests that the NMDA receptor could be a determinant factor to modulate the dendritic arrangement and the synaptogenesis.


Subject(s)
Brain/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Sodium Glutamate/pharmacology , Aging/drug effects , Aging/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Brain/growth & development , Brain/metabolism , Cell Differentiation/physiology , Dendrites/drug effects , Dendrites/metabolism , Gene Expression Regulation/physiology , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Male , Neostriatum/drug effects , Neostriatum/growth & development , Neostriatum/metabolism , Neurons/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Synapses/drug effects , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...