Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Respir Physiol Neurobiol ; 294: 103744, 2021 12.
Article in English | MEDLINE | ID: mdl-34302992

ABSTRACT

Central respiratory chemoreceptors play a key role in the respiratory homeostasis by sensing CO2 and H+ in brain and activating the respiratory neural network. This ability of specific brain regions to respond to acidosis and hypercapnia is based on neuronal and glial mechanisms. Several decades ago, glutamatergic transmission was proposed to be involved as a main mechanism in central chemoreception. However, a complete identification of mechanism has been elusive. At the rostral medulla, chemosensitive neurons of the retrotrapezoid nucleus (RTN) are glutamatergic and they are stimulated by ATP released by RTN astrocytes in response to hypercapnia. In addition, recent findings show that caudal medullary astrocytes in brainstem can also contribute as CO2 and H+ sensors that release D-serine and glutamate, both gliotransmitters able to activate the respiratory neural network. In this review, we describe the mammalian astrocytic glutamatergic contribution to the central respiratory chemoreception trying to trace in vertebrates the emergence of several components involved in this process.


Subject(s)
Astrocytes/metabolism , Biological Evolution , Chemoreceptor Cells/metabolism , Glutamic Acid/metabolism , Medulla Oblongata/metabolism , Receptors, Ionotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Respiratory Center/metabolism , Animals
2.
Biochim Biophys Acta Proteins Proteom ; 1868(11): 140484, 2020 11.
Article in English | MEDLINE | ID: mdl-32652125

ABSTRACT

d-serine, released from mouse medullary astrocytes in response to increased CO2 levels, boosts the respiratory frequency to adapt breathing to physiological demands. We analyzed in mouse neonates, the influence of d-serine upon inspiratory/expiratory durations and the architecture of the inspiratory burst, assessed by pwelch's power spectrum density (PSD) and continuous wavelet transform (CWT) analyses. Suction electrode recordings were performed in slices from the ventral respiratory column (VRC), site of generation of the respiratory rhythm, and in brainstem-spinal cord (en bloc) preparations, from the C5 ventral roots, containing phrenic fibers that in vivo innervate and drive the diaphragm, the main inspiratory muscle. In en bloc and slice preparations, d-serine (100 µM) reduced the expiratory, but not the inspiratory duration, and increased the frequency and the regularity of the respiratory rhythm. In en bloc preparations, d-serine (100 µM) also increased slightly the amplitude of the integrated inspiratory burst and the area under the curve of the integrated inspiratory burst, suggesting a change in the recruitment or the firing pattern of neurons within the burst. Time-frequency analyses revealed that d-serine changed the burst architecture of phrenic roots, widening their frequency spectrum and shifting the position of the core of firing frequencies towards the onset of the inspiratory burst. At the VRC, no clear d-serine induced changes in the frequency-time domain could be established. Our results show that d-serine not only regulates the timing of the respiratory cycle, but also the recruitment strategy of phrenic motoneurons within the inspiratory burst.


Subject(s)
Brain Stem/physiology , Respiration , Serine/physiology , Spinal Cord/physiology , Animals , Animals, Newborn , Female , Male , Mice
3.
Nat Commun ; 8(1): 838, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29018191

ABSTRACT

Central chemoreception is essential for adjusting breathing to physiological demands, and for maintaining CO2 and pH homeostasis in the brain. CO2-induced ATP release from brainstem astrocytes stimulates breathing. NMDA receptor (NMDAR) antagonism reduces the CO2-induced hyperventilation by unknown mechanisms. Here we show that astrocytes in the mouse caudal medullary brainstem can synthesize, store, and release D-serine, an agonist for the glycine-binding site of the NMDAR, in response to elevated CO2 levels. We show that systemic and raphe nucleus D-serine administration to awake, unrestrained mice increases the respiratory frequency. Application of D-serine to brainstem slices also increases respiratory frequency, which was prevented by NMDAR blockade. Inhibition of D-serine synthesis, enzymatic degradation of D-serine, or the sodium fluoroacetate-induced impairment of astrocyte functions decrease the basal respiratory frequency and the CO2-induced respiratory response in vivo and in vitro. Our findings suggest that astrocytic release of D-serine may account for the glutamatergic contribution to central chemoreception.Astrocytes are involved in chemoreception in brainstem areas that regulate breathing rhythm, and astrocytes are known to release D-serine. Here the authors show that astrocyte release of D-serine contributes to CO2 sensing and breathing in brainstem slices, and in vivo in awake unrestrained mice.


Subject(s)
Astrocytes/metabolism , Brain Stem/metabolism , Carbon Dioxide/metabolism , Serine/metabolism , Animals , Astrocytes/drug effects , Brain Stem/cytology , Cells, Cultured , Female , Fluoroacetates/pharmacology , Hypercapnia/metabolism , Hypercapnia/physiopathology , Male , Mice, Inbred Strains , Receptors, N-Methyl-D-Aspartate/metabolism , Respiration
4.
Neurotox Res ; 25(1): 68-80, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24114771

ABSTRACT

Scavenger receptor Class A (SR-A) participates in the regulation of inflammatory processes against pathogens and in inflammatory stimulation. We have recently demonstrated the presence of SR-A in astrocytes, but its participation in their inflammatory response is unknown. Astrocytes regulate neuroinflammation through the regulation of microglial cell activation and the production of cytokines, neurotrophic factors, and reactive species. Using astrocytes from SR-A(-/-) mice in culture, we assessed the participation of SR-A in their inflammatory activation, evaluating the activation of IκB/NF-κB and MAPK signaling pathways and the production of nitric oxide (NO) and IL-1ß in response to SR-A ligands. In SR-A(-/-) astrocytes, lipopolysaccharide (LPS) induced higher levels of NO and reduced levels of IL-1ß compared to SR-A(+/+) cells. In addition, SR-A(-/-) astrocytes had a reduced basal and LPS-stimulated JNK phosphorylation, and a delayed activation on IκB/NF-κB signaling pathway in response to LPS. Moreover, inhibition of the ERK pathway reduced NO production by SR-A(-/-) cells, suggesting that this signaling pathway modulated LPS-induced NO production, an effect that depended on the presence of SR-A. Our results suggest that SR-A participates in the modulation of signaling pathways involved in the production of soluble molecules implicated in the neuroinflammatory response.


Subject(s)
Astrocytes/metabolism , MAP Kinase Signaling System , Scavenger Receptors, Class A/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Inflammation/metabolism , Mice , Mice, Inbred ICR , Mice, Knockout , Nitric Oxide/metabolism , Scavenger Receptors, Class A/genetics
5.
J Neuroimmunol ; 251(1-2): 6-13, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22743055

ABSTRACT

Class-A scavenger receptor (SR-A) is expressed by microglia, and we show here that it is also expressed by astrocytes, where it participates on their inflammatory activation. Astrocytes play a key role on the inflammatory response of the central nervous system, secreting several soluble mediators like cytokines and radical species. Exposure to SR ligands activated MAPKs and NF-κB signaling and increased production of IL1ß and nitric oxide (NO). IL1ß classically an inflammatory cytokine surprisingly did not increase but inhibited LPS+IFNγ-induced NO production by astrocytes. Our results suggest that SRs expressed by astrocytes participate in the modulation of inflammatory activation.


Subject(s)
Astrocytes/metabolism , Interleukin-1beta/metabolism , Scavenger Receptors, Class A/biosynthesis , Animals , Astrocytes/drug effects , Cells, Cultured , Interferon-gamma/pharmacology , Ligands , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/physiology , Mice , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Rats , Rats, Sprague-Dawley
6.
Med. intensiva (Madr., Ed. impr.) ; 36(1): 37-44, ene.-feb. 2012. ilus, tab
Article in Spanish | IBECS | ID: ibc-98900

ABSTRACT

El traumatismo encéfalo craneano (TEC) es un problema de salud de distribución mundial, y es especialmente prevalente en la población adulta joven. Es característica la presencia de uno o más focos de daño, que luego progresan hacia áreas inicialmente no lesionadas, mediante cascadas de respuesta inflamatoria, excitotoxicidad, condiciones de falla energética, y la participación de la glía amplificando la respuesta tisular al daño inicial. Esta progresión es, en teoría, susceptible de una intervención terapéutica. Sin embargo, hasta ahora todos los estudios con fármacos neuroprotectores han fracasado, no existiendo un tratamiento especí-fico efectivo. Los resultados negativos se explican en parte por el empleo de una estrategia centrada solo en las neuronas, sin considerar otras células participantes, u otros mecanismos patogénicos. Para cambiar este panorama, es necesario re-enfocar el problema a través de una mejor comprensión de los mecanismos que determinan la progresión del daño. En esta revisión discutiremos los principales mecanismos biológicos involucrados en la progresión del daño tisular post-trauma. Se aborda la fisiopatología general de los tipos de traumatismos, mecanismos celulares del daño secundario incluyendo inflamación, apoptosis, tumefacción celular, excitoxicidad, y participación de la glía en la propagación del daño. Se destaca el papel de laglía en cada uno de los mecanismos celulares mencionados. Se incluyen algunas aproximaciones terapéuticas relacionadas con los mecanismos descritos. Se finaliza con un diagrama general que resume los principales aspectos discutido (AU)


Traumatic brain injury (TBI) is a worldwide health problem that is especially prevalent in young adults. It is characterized by one or more primary injury foci, with secondary spread to initially not compromised areas via cascades of inflammatory response, excitotoxicity, energy failure conditions, and amplification of the original tissue injury by glia. In theory, such progression of injury should be amenable to management. However, all neuroprotective drug trials have failed, and specific treatments remain lacking. These negative results can be explained by a neuron centered approach, excluding the participation of other cell types and pathogenic mechanisms. To change this situation, it is necessary to secure a better understanding of the biological mechanisms determining damage progression or spread. We discuss the biological mechanisms involved in the progression of post-trauma tissue damage, including the general physiopathology of TBI and cellular mechanisms of secondary damage such as inflammation, apoptosis, cell tumefaction, excitotoxicity, and the role of glia in damage propagation. We highlight the role of glia in each cellular mechanism discussed. Therapeutic approaches related to the described mechanisms have been included. The discussion is completed with a working model showing the convergence of the main topics (AU)


Subject(s)
Humans , Craniocerebral Trauma/physiopathology , Brain Injury, Chronic/physiopathology , Apoptosis , Neurotoxins/pharmacokinetics , Neuroglia , Risk Factors , Disease Progression
7.
Neurotox Res ; 22(1): 69-78, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22237943

ABSTRACT

Alzheimer disease (AD) is a neurodegenerative disorder characterized by the accumulation of ß amyloid (Aß) aggregates. Aß induces the inflammatory activation of glia, inducing secretion of Interleukin 1ß (IL1ß), nitric oxide (NO) and superoxide radicals. The specific receptor responsible for the induction of inflammatory activation by Aß, is still an open question. We propose that scavenger receptors (SR) participate in the activation of glia by Aß. We assessed production of NO, synthesis of IL1ß and activation of ERK, JNK and NF-κB signaling pathways by Western blot, in primary rat glial cultures exposed to SR ligands (fucoidan and Poly I), LPS + IFNγ (LI), and Aß. Poly I but not fucoidan nor fibrillar Aß increased threefold NO production by astrocytes in a time-dependent manner. Fucoidan and Poly I increased 5.5- and 3.5-fold NO production by microglia, and co-stimulation with Aß increased an additional 60% NO induced by SR ligands. Potentiation by Aß was observed later for astrocytes than for microglia. In astrocytes, co-stimulation with Aß potentiated ERK and JNK activation in response to Fucoidan and Poly I, whereas it reduced induction of JNK activation by LI and left unaffected NF-κB activation induced by LI. Levels of pro-IL1ß in astrocytes increased with Aß, SR ligands and LI, and were potentiated by co-stimulation with Aß. Our results suggest that SRs play a role on inflammatory activation, inducing production of NO and IL1ß, and show potentiation by Aß. Potentiation of the inflammatory response of Aß could be meaningful for the activation of glia observed in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Inflammation/metabolism , Microglia/metabolism , Receptors, Scavenger/metabolism , Signal Transduction , Alzheimer Disease/immunology , Animals , Cells, Cultured , Interferon-gamma/pharmacology , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide/metabolism , Poly I/pharmacology , Polysaccharides/pharmacology , Rats , Receptors, Scavenger/agonists , Superoxides/metabolism
8.
Med Intensiva ; 36(1): 37-44, 2012.
Article in Spanish | MEDLINE | ID: mdl-21903299

ABSTRACT

Traumatic brain injury (TBI) is a worldwide health problem that is especially prevalent in young adults. It is characterized by one or more primary injury foci, with secondary spread to initially not compromised areas via cascades of inflammatory response, excitotoxicity, energy failure conditions, and amplification of the original tissue injury by glia. In theory, such progression of injury should be amenable to management. However, all neuroprotective drug trials have failed, and specific treatments remain lacking. These negative results can be explained by a neuron centered approach, excluding the participation of other cell types and pathogenic mechanisms. To change this situation, it is necessary to secure a better understanding of the biological mechanisms determining damage progression or spread. We discuss the biological mechanisms involved in the progression of post-trauma tissue damage, including the general physiopathology of TBI and cellular mechanisms of secondary damage such as inflammation, apoptosis, cell tumefaction, excitotoxicity, and the role of glia in damage propagation. We highlight the role of glia in each cellular mechanism discussed. Therapeutic approaches related to the described mechanisms have been included. The discussion is completed with a working model showing the convergence of the main topics.


Subject(s)
Brain Injuries/physiopathology , Animals , Apoptosis , Brain Death , Brain Edema/etiology , Brain Injuries/complications , Brain Injuries/metabolism , Cell Size , Diffuse Axonal Injury/etiology , Diffuse Axonal Injury/pathology , Diffuse Axonal Injury/physiopathology , Glutamic Acid/metabolism , Humans , Hyperglycemia/etiology , Inflammation , Intracranial Hypertension/etiology , Intracranial Hypertension/physiopathology , Ion Transport , Models, Neurological , Neuroglia/physiology , Neurons/pathology
9.
Rev. neurol. (Ed. impr.) ; 52(7): 426-434, 1 abr., 2011. tab, ilus
Article in Spanish | IBECS | ID: ibc-87346

ABSTRACT

Introducción. La esclerosis lateral amiotrófica (ELA) es una enfermedad neurodegenerativa para la cual no existe tratamiento curativo. En sus mecanismos patogénicos y de progresión participaría el daño oxidativo, acumulación de agregados intracelulares, disfunción mitocondrial, defectos en el transporte axonal, disminución de factores tróficos, alteraciones gliales, metabolismo aberrante del ARN y excitotoxicidad. Objetivo. Evaluar los resultados terapéuticos de las células troncales adultas como terapia en la ELA. Desarrollo. Las células troncales son una posible estrategia terapéutica, puesto que sus mecanismos de acción permitirían revertir varios de los mecanismos patogénicos descritos para la ELA. Entre las células troncales adultas destacan las células mesenquimales obtenidas de la médula ósea. Estas células son capaces de diferenciarse en todas las células del sistema nervioso central y potencialmente reemplazarlas. Además, poseen efectos inmunomoduladores, caracterizándose por secretar, especialmente en ambientes neuroinflamatorios, factores neurotróficos y antiinflamatorios. Estudios en modelos murinos de ELA muestran disminución de la inflamación y progresión de la enfermedad, y aumento de la supervivencia. Hay varios ensayos clínicos publicados, muy heterogéneos entre sí, que sugieren que el trasplante de células troncales sería seguro, pero no mejoraría en la evolución clínica de los pacientes. Conclusión. Se necesitan estudios preclínicos adicionales para refinar esta aproximación terapéutica, evaluando la supervivencia a largo plazo, diferenciación de células mesenquimales, dosificación, actividad biológica y seguridad, para continuar con estudios en pacientes (AU)


Introduction. Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease. At present, there are not curative therapies for ALS. Pathogenic and progression mechanisms suggest the existence of oxidative stress, abnormal intracellular protein aggregation, mitochondrial dysfunction, axonal transport impairment, impairment of trophic support, altered glial cell function, and glutamate excitoxicity. Aim. To evaluate therapeutic results with adult stem cell for ALS treatment. Development. Stem cells represent a potential therapeutic strategy, because their biological mechanisms could act on several of the pathogenic mechanisms proposed for ALS. Bone marrow mesenchymal stem cells are especially interesting among adult stem cells. Mesenchymal stem cells can differentiate in all central nervous system cells and potentially replace them. Furthermore, they have immunomodulatory effects, secreting, especially in neuroinflammatory environments, neurotrophic and antiinflammatory factors. Studies in murine models of ALS show decrease of inflammation and disease progression, and increase on animal highly heterogeneous, suggest that mesenchymal stem cells transplant in ALS appears to be safe. However, they fail showing clinical improvement of patients. Conclusion. Additional preclinical studies are necessary to refine this therapeutic approach, to assess long term survival and differentiation of mesenchymal stem cells, dosing, biological activity and safety should be conducted before any planning further human testing occurs (AU)


Subject(s)
Humans , Amyotrophic Lateral Sclerosis/surgery , Stem Cell Transplantation , Regenerative Medicine/trends , Disease Progression , Disease-Free Survival , Inflammation/physiopathology , Immunologic Factors
10.
Neuroscience ; 158(4): 1338-47, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19041694

ABSTRACT

Progression of Alzheimer's disease (AD) is associated with chronic inflammation and microvascular alterations, which can induce impairment of brain perfusion because of vascular pathology and local acidosis. Acidosis can promote amyloidogenesis, which could further contribute to neurodegenerative changes. Nevertheless, there is also evidence that acidosis has neuroprotective effects in hypoxia models. Here we studied the effect of moderate acidosis on beta-amyloid (Abeta)-mediated neurotoxicity. We evaluated morphological changes, cell death, nitrite production and reductive metabolism of hippocampal cultures from Sprague-Dawley rats exposed to Abeta under physiological (pH 7.4) or moderate acidosis (pH 7.15-7.05). In addition, because transforming growth factor beta (TGFbeta) 1 is neuroprotective and is induced by several pathophysiological conditions, we assessed its presence at the different pHs. The exposure of hippocampal cells to Abeta induced a conspicuous reduction of neurites' arborization, as well as increased neuronal death and nitric oxide production. However, Abeta neurotoxicity was significantly attenuated when hippocampal cultures were kept at pH 7.15-7.05, showing a 68% reduction on lactate dehydrogenase release compared with cultures exposed to Abeta at pH 7.4 (P<0.01). Similarly, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide reduction increased 3.5-fold (P<0.05), and Abeta-induced nitrite production was reduced by 65% when exposed to moderate acidosis compared with basal pH media (P<0.05). At the same time, moderate acidosis decreased intracellular TGFbeta1 precursor (latency associated protein-TGFbeta1) and increased up to fourfold TGFbeta1 bioactivity, detecting a 43% increase in the active TGFbeta levels in cultures exposed to Abeta and moderate acidosis. Inhibition of TGFbeta signaling abolished the neuroprotective effect of moderate acidosis. Our results show that moderate acidosis protected hippocampal cells from Abeta-mediated neurotoxicity through the increased activation and signaling potentiation of TGFbeta.


Subject(s)
Acidosis/metabolism , Amyloid beta-Peptides/toxicity , Hippocampus/cytology , Neurons/drug effects , Peptide Fragments/toxicity , Transforming Growth Factor beta/metabolism , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Azides , Benzamides/pharmacology , Cell Size/drug effects , Dioxoles/pharmacology , Embryo, Mammalian , Female , Hydrogen-Ion Concentration , L-Lactate Dehydrogenase/metabolism , Nitric Oxide/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Tubulin/metabolism
11.
Neuroimmunomodulation ; 14(3-4): 168-74, 2007.
Article in English | MEDLINE | ID: mdl-18073510

ABSTRACT

The strong inflammatory response observed in neurodegenerative diseases can depend on the impairment of the endogenous control of microglial activation, triggering the release of potentially detrimental factors such as cytokines, nitric oxide (NO) and superoxide anion (O(2)(-)). Our aim was to study the activation of microglial cells and the transduction pathways involved in their modulation by IL-1beta and TNF-alpha. Microglial and mixed glial cell cultures from neonatal rats were exposed to IFN-gamma and/or IL-1beta and TNF-alpha. We analyzed NO secretion and the activation of ERK and STAT1. We found that astrocytes modulated microglial cell activation, decreasing production of NO. IFN-gamma induced an 18- to 25-fold increase in NO, associated to a 3- to 5-fold increase in ERK phosphorylation in microglial cultures. IL-1beta, but not TNF-alpha, inhibited IFN-gamma-induced production of NO in microglia by 87%. It also reduced IFN-gamma-induced phosphoERK (pERK) by 40%, without affecting phosphoSTAT1 (pSTAT1). In contrast, in microglial cultures exposed to media conditioned by astrocytes, IL-1beta did not inhibit pERK, whereas it reduced activation of STAT1. Inducible NO synthase expression induced by IFN-gamma in microglial cultures was reduced when the activation of ERK was prevented. We propose that IL-1beta modulates IFN-gamma-induced production of oxidative molecules through cross talk between STAT1 and MAPK pathways, regulating the amplitude and duration of microglial activation. Modulation of ERK was observed at 30 min, whereas inhibition of pSTAT was observed later (at 4 h), indicating that it was an early and transient phenomenon.


Subject(s)
Astrocytes/immunology , Encephalitis/immunology , Gliosis/immunology , Interleukin-1beta/immunology , Microglia/immunology , Signal Transduction/immunology , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Encephalitis/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gliosis/physiopathology , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Microglia/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor/drug effects , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
12.
Rev Neurol ; 44(9): 519-23, 2007.
Article in Spanish | MEDLINE | ID: mdl-17492608

ABSTRACT

INTRODUCTION: Attention-deficit/hyperactivity disorder (ADHD) is the main neurological diagnosis in Chilean children. Its profile and evolution in adults has not been appropriately studied, despise its personal and social impact. AIM: To describe the characteristics of adults with ADHD evaluated in a memory unit, verifying the existence of differences depending on gender. PATIENTS AND METHODS: A demographic and symptomatic evaluation protocol was applied to all patients diagnosed with ADHD who consulted at the Memory Unit of the Pontificia Universidad Católica de Chile, during the year 2004. RESULTS: Eighty six patients were included. Average age was 37, being 53% male. Most patients were diagnosed for the first time in adulthood, corresponding to an ADHD of combined type. The main patients' complaints were forgetfulness and distraction. A stressing factor capable of worsening the symptoms was identified in 59% of patients. Depression was the principal comorbidity, with a significantly higher incidence in women. CONCLUSIONS: The limitations of ADHD diagnostic criteria available for adult patients are discussed. Differences depending on gender were analyzed, describing a predominantly disruptive profile in men and depressive profile in women. There is a clear under-diagnosis of female children with ADHD, with a potential negative impact on their neuropsychological development. The differential diagnosis with mild cognitive impairment, in patients complaining of recent memory decline is discussed.


Subject(s)
Attention Deficit Disorder with Hyperactivity/diagnosis , Memory/physiology , Adult , Attention Deficit Disorder with Hyperactivity/physiopathology , Child , Chile , Comorbidity , Depression/physiopathology , Female , Humans , Male , Memory Disorders/diagnosis , Memory Disorders/etiology , Middle Aged , Neuropsychological Tests , Sex Factors
13.
Neuroscience ; 141(1): 223-31, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16675136

ABSTRACT

In newborn and adult mammals, chemosensory drive exerted by CO(2) and H(+) provides an essential tonic input: without it the rhythm of respiration is abolished. It is not known, however, whether this chemosensory drive and the respiratory rhythm appear simultaneously during development. In isolated brainstem-spinal cord preparations from fetal mice, we determined at what stage of fetal life the respiratory rhythm appeared in third to fifth cervical ventral roots (phrenic motoneurons) and whether this fetal rhythm was sensitive to chemosensory inputs. A respiratory-like rhythm consisting of short duration bursts of discharges recurring at 2-16 min(-1) was detected in two of nine embryonic day 13 fetuses; it was abolished by transection of the spinal cord between the first to second cervical segments and was phase-related to rhythmic activity from medullary units of the ventral respiratory group. At embryonic day 13, it coexisted with a slow rhythm (0.1-2.0 min(-1)) of long duration bursts of action potentials which was generated by the spinal cord. At later fetal stages, the respiratory-like rhythm became more robust and of higher frequency, while the spinal cord rhythm became less obvious. At all fetal stages, acidification of the superfusion medium from pH 7.5-7.2 or 7.4-7.3 or 7.4 to 7.2 increased the frequency of both the respiratory-like and the spinal cord rhythms. In addition, acidification reduced the amplitude of the integrated burst activity of the spinal cord rhythm of embryonic day 13-embryonic day 16 fetuses and the respiratory-like rhythm of embryonic day 17 and older fetuses. Our results indicate that the rhythms transmitted by phrenic motoneurons during fetal development are chemosensitive from early fetal stages. Through its effects on induction and patterning of the rhythm, chemosensory drive may play a role in activity-dependent formation of respiratory neural networks.


Subject(s)
Motor Neurons/physiology , Periodicity , Respiratory Center/physiology , Spinal Cord/embryology , Action Potentials/physiology , Age Factors , Animals , Animals, Newborn , Brain Stem/cytology , Brain Stem/embryology , Embryo, Mammalian , Hydrogen-Ion Concentration , In Vitro Techniques , Mice , Spinal Cord/cytology , Spinal Cord Injuries/physiopathology , Statistics, Nonparametric , Stimulation, Chemical
14.
Neurotox Res ; 8(3-4): 277-87, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16371322

ABSTRACT

Pro-inflammatory molecules induce glial activation and the release of potentially detrimental factors capable of generating oxidative damage, such as nitric oxide (NO) and superoxide anion (O2.-). Activated glial cells (astrocytes and microglia) are associated to the inflammatory process in neurodegenerative diseases. A strong inflammatory response could escape endogenous control becoming toxic to neurons and contributing to the course of the disease. We evaluated in a hippocampal cells-microglia co-culture model, if the pro-inflammatory condition induced by lipopolysaccharide + interferon-gamma (LPS+IFN-gamma) promoted damage directly or if damage was secondary to glial activation. In addition, we explored the effect of the anti-inflammatory cytokine transforming growth factor-beta1 (TGF-beta1), and pro-inflammatory cytokines, interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha) on the regulation of the inflammatory response of microglia. We found that LPS+IFN-gamma-induced damage on hippocampal cultures was dependent on the presence of microglial cells. In hippocampal cultures exposed to LPS+IFN-gamma, TGF-beta1 was induced whereas in microglial cell cultures LPS+IFN-gamma induced the secretion of IL-1beta. TGF-beta1 and IL-1beta but not TNF-alpha decreased the NO production by 70-90%. PD98059, an inhibitor of MAP kinase (MEK), reduced the IFN-gamma-induced NO production by 40%. TGF-beta and IL-1beta reduced the IFN-gamma induced phosphorylation of ERK1,2 by 60% and 40%, respectively. However, the effect of IL-1beta was observed at 30 min and that of TGF-beta1 only after 24 h of exposure. We propose that acting with different timing, TGF-beta1 and IL-1beta can modulate the extracellular signal-regulated kinase ERK1,2, as a common element for different transduction pathways, regulating the amplitude and duration of glial activation in response to LPS+IFN-gamma. Cross-talk among brain cells may be key for the understanding of inflammatory mechanisms involved in pathogenesis of neurodegenerative diseases.


Subject(s)
Cytokines/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neuroglia/drug effects , Signal Transduction/drug effects , Analysis of Variance , Animals , Animals, Newborn , Blotting, Western/methods , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Ethidium , Flavonoids/pharmacology , Hippocampus/cytology , Interleukin-1/metabolism , Neurons/drug effects , Nitrites/metabolism , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , Tetrazolium Salts , Thiazoles , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
15.
Biol Res ; 34(2): 123-8, 2001.
Article in English | MEDLINE | ID: mdl-11715204

ABSTRACT

Brain glial cells secrete several molecules that can modulate the survival of neurons after various types of damage to the CNS. Activated microglia and astrocytes closely associate to amyloid plaques in Alzheimer Disease (AD). They could have a role in the neurotoxicity observed in AD because of the inflammatory reaction they generate. There is controversy regarding the individual part played by the different glial cells, and the interrelationships between them. Both astrocytes and microglia produce several cytokines involved in the inflammatory reaction. Moreover, the same cytokines may have different effects, depending on their concentration and the type of cells in the vicinity. In turn, the events occurring in response to injury may lead to changes in the nature and relative concentration of the various factors involved. To learn about these putative glial interrelationships, we examined some effects of astrocytes on microglial activation.


Subject(s)
Alzheimer Disease/pathology , Astrocytes/physiology , Cytokines/metabolism , Microglia/physiology , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , Astrocytes/metabolism , Cell Communication , Cell Culture Techniques , Microglia/metabolism , Rats
16.
Eur J Neurosci ; 14(6): 946-56, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11595033

ABSTRACT

Astrocytes and microglia are closely associated with amyloid plaques in Alzheimer's disease (AD). Microglia constitute the first barrier surrounding plaques, although they seem to be unable to remove them efficiently. We evaluated the reaction of microglial cells from neonatal rats and mice to plaque mimetics. The C-terminal part of the amyloid precursor protein (APP) or amyloid peptide (A beta) was immobilized to either 60-microm or 2.8-microm beads and incubated with microglial cells. Beads of 60 microm, having approximately the size of senile plaques, were not phagocytosed, in contrast to 2.8-microm beads, which were phagocytosed by microglia but not by astrocytes. Once taken up by the cells, proteins immobilized to the beads were degraded rapidly, as confirmed by mass spectrometry and immunofluorescence with an antibody against beta-amyloid. On the other hand, no protein degradation was observed with 60-microm beads. Also, probably as a reaction to its incapability to phagocytose the beads, glia organized around the beads and started to proliferate. Cell proliferation was more pronounced when the beads contained the A beta epitope compared with the beads with an inert surface. This in vitro effect could be exploited to set up a screening assay for compounds that ameliorate the adverse reaction of microglia supposed to contribute to the pathogenesis of AD.


Subject(s)
Amyloid beta-Protein Precursor , Neuroglia/drug effects , Amyloid beta-Peptides/pharmacology , Cells, Cultured , Cloning, Molecular , Endocytosis/drug effects , Endocytosis/physiology , Endopeptidases/metabolism , Fluorescent Antibody Technique , Humans , Indicators and Reagents , Mass Spectrometry , Microspheres , Nitrites/metabolism
17.
J Neurosci Res ; 60(3): 401-11, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10797543

ABSTRACT

Low density lipoprotein receptor-related protein (LRP) participates in the uptake and degradation of several ligands implicated in neuronal pathophysiology including apolipoprotein E (apoE), activated alpha(2) -macroglobulin (alpha(2)M*) and beta-amyloid precursor protein (APP). The receptor is expressed in a variety of tissues. In the brain LRP is present in pyramidal-type neurons in cortical and hippocampal regions and in astrocytes that are activated as a result of injury or neoplasmic transformation. As LRP is expressed in the monocyte/macrophage cell system, we were interested in examining whether LRP is expressed in microglia. We isolated glial cells from the brain of neonatal rats and LRP was immunodetected both in microglial cells and in astrocytes expressing glial fibrillar acidic protein (GFAP). Microglial cells were able to bind and internalize LRP-specific ligand, alpha(2)M*. The internalization was inhibitable by RAP, with a Kd of 1.7 nM. The expression of LRP was up-regulated by dexamethasone, and down-regulated by lipopolysaccharide (LPS), gamma interferon (IFN-gamma) or a combination of both. LRP was less sensitive to dexamethasone in activated astrocytes than in microglia. We provided the first analysis of LRP expression and regulation in microglia. Our results open the possibility that microglial cells could be related to the participation of LRP and its ligands in different pathophysiological states in brain.


Subject(s)
Microglia/metabolism , Receptors, Immunologic/biosynthesis , Receptors, LDL/biosynthesis , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Dexamethasone/pharmacology , Down-Regulation/drug effects , Fluorescent Antibody Technique , Glucocorticoids/pharmacology , Immunohistochemistry , Interferon-gamma/pharmacology , Kinetics , Ligands , Lipopolysaccharides/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1 , Neuroglia/metabolism , Precipitin Tests , Rats , Up-Regulation/drug effects
18.
J Neurosci Res ; 58(3): 387-95, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10518112

ABSTRACT

We studied the expression of the mannose receptor (ManR) in rat microglial cells. Microglial cells are the central nervous system resident macrophages, key participants of the innate immune response. ManR is a differentiation marker and a relevant glycoprotein for the phagocytic and endocytic function of macrophages. Because there is evidence suggesting that ManR could mediate some of the nonenzymatic effects of acetilcholinesterase (AchE) and the enzyme seems to be involved in Alzheimer's disease (AD), we looked for ManR in microglia, evaluating the functionality of the receptor. We isolated microglial cells from the brain of 2-day-old neonatal rats. Microglial cells, identified by their specific staining with the lectin Griffonia simplicifolia, expressed ManR, being detected by immunocytochemistry, Western blot, and immunoprecipitation. Microglial ManR was downregulated by lipopolysaccharide (LPS) and upregulated by dexamethasone, as described for peripheral macrophages. Microglial ManR was functional and able to internalize horseradish peroxidase (HRP), a known ManR ligand, in a mannan-inhibitable manner. The presence of a functional ManR in microglia opens the possibility that ManR could participate in multiple physiologic and pathologic conditions in the central nervous system (CNS), including inflammation, ischaemia, and neurodegenerative diseases such as AD.


Subject(s)
Lectins, C-Type , Mannose-Binding Lectins , Microglia/metabolism , Plant Lectins , Receptors, Cell Surface/metabolism , Acetylcholinesterase/metabolism , Animals , Animals, Newborn , Antibodies, Monoclonal , Brain/cytology , Brain/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Down-Regulation/drug effects , Horseradish Peroxidase/pharmacokinetics , Humans , Lectins , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mannose Receptor , Microglia/cytology , Rats , Receptors, Cell Surface/genetics , Up-Regulation/drug effects
19.
Dev Neurosci ; 21(6): 423-35, 1999.
Article in English | MEDLINE | ID: mdl-10640861

ABSTRACT

The pathway a growing axon follows is determined by a number of cues, including differential adhesion to surface molecules on axons and the matrix of the fascicles along which they grow. We have characterized the differential expression of an extracellular antigen and the effects of a monoclonal antibody against this molecule on the development of the grasshopper central nervous system (CNS). The 5C1 monoclonal antibody was generated against ganglion chains of grasshopper embryos; it labels cell bodies of newly differentiated neurons and their axons as they extend. Electron microscopy of embryos at 42% of development reveals that 5C1 labels neuronal filopodia, axons and somata, and areas of glial membrane in apposition to neurite fascicles. After 70% of development, labeling is lost from axon bundles, but remains on cell bodies. 5C1 also cross-reacts with an epitope expressed in Drosophila CNS during embryonic development. Enzymatic digestion suggests that the antigen recognized by the antibody is likely to be a glycolipid. In embryos exposed to 5C1 during early stages of development of the CNS, at the time when the first axons begin to extend, the formation of axon pathways is blocked or greatly delayed. Our results suggest that the 5C1 antigen participates in the formation of the axon scaffold and may play a functional role in the initiation and maintenance of axon outgrowth during early development of the CNS.


Subject(s)
Antibodies, Monoclonal/pharmacology , Axons/chemistry , Nerve Tissue Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Axons/immunology , Axons/ultrastructure , Drosophila , Embryo, Nonmammalian/chemistry , Epitopes , Glycolipids , Grasshoppers , Insect Proteins/analysis , Insect Proteins/immunology , Microscopy, Immunoelectron , Nerve Tissue Proteins/analysis , Nervous System/chemistry , Nervous System/growth & development
20.
Mol Psychiatry ; 3(3): 247-55, 1998 May.
Article in English | MEDLINE | ID: mdl-9672900

ABSTRACT

Acetylcholinesterase (AChE), the enzyme involved in the hydrolysis of the neurotransmitter acetylcholine, has been implicated in non-cholinergic actions which may play a role in neurodegenerative diseases such as Alzheimer's disease. To study the potential cytotoxicity of brain AChE, the effects of affinity purified AChE were analyzed on neuronal (Neuro 2a) and glial-like (B12) cells. LDH release and MTT reduction assays showed that AChE was toxic; the toxicity was dependent on the enzyme concentration, time of incubation and cellular density. The toxic effect of AChE was not related to its catalytic activity, since the anti-cholinesterase drug BW284C51 and heat inactivation were unable to block the effects of the enzyme. When cells were incubated at 4 degrees C, toxicity was completely blocked, in contrast to cells incubated at 37 degrees C. The presence of serum in the culture medium inhibited the toxic effects of AChE. Cytoplasmic shrinkage, condensation and fragmentation of nucleus as well as DNA strand breaks detected with the TUNEL technique indicated that apoptotic cell death is involved in the effect of AChE. Considering that we have previously shown that AChE promotes the assembly of beta-amyloid peptide into neurotoxic amyloid fibrils, it is conceivable that the neurotoxicity of AChE shown here may play a role in the neuronal degeneration observed in Alzheimer's disease.


Subject(s)
Acetylcholinesterase/toxicity , Neuroglia/drug effects , Neurons/drug effects , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Animals , Biotin , Blood Proteins/pharmacology , Cattle , Cell Count , Cold Temperature , DNA Fragmentation , Deoxyuracil Nucleotides , Enzyme Activation/physiology , Mice , Neuroblastoma , Neurofibrillary Tangles/enzymology , Neuroglia/cytology , Neurons/cytology , Staining and Labeling , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...