Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Brain Res ; 1614: 1-13, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-25881894

ABSTRACT

Prenatal alcohol exposure is associated with microencephaly, cognitive and behavioral deficits, and growth retardation. Some of the mechanisms of ethanol-induced injury, such as high level oxidative stress and overexpression of pro-apoptotic genes, can increase the sensitivity of fetal neurons towards hypoxic/ischemic stress associated with normal labor. Thus, alcohol-induced sequelae may be the cumulative result of direct ethanol toxicity and increased neuronal vulnerability towards metabolic stressors, including hypoxia. We examined the effects of ethanol exposure on the fetal cerebellar granular neurons' susceptibility to hypoxic/hypoglycemic damage. A chronic ethanol exposure covered the entire prenatal period and 5 days postpartum through breastfeeding, a time interval partially extending into the third-trimester equivalent in humans. After a binge-like alcohol exposure at postnatal day 5, glutamatergic cerebellar granule neurons were cultured and grown for 7 days in vitro, then exposed to a 3-h oxygen-glucose deprivation to mimic a hypoxic/ischemic condition. Cellular viability was monitored by dynamic recording of propidium iodide fluorescence over 20 h reoxygenation. We explored differentially expressed genes on microarray data from a mouse embryonic ethanol-exposure model and validated these by real-time PCR on the present model. In the ethanol-treated cerebellar granule neurons we find an increased expression of genes related to apoptosis (Mapk8 and Bax), but also of genes previously described as neuroprotective (Dhcr24 and Bdnf), which might suggest an actively maintained viability. Our data suggest that neurons exposed to ethanol during development are more vulnerable to in vitro hypoxia/hypoglycemia and have higher intrinsic death susceptibility than unexposed neurons.


Subject(s)
Central Nervous System Depressants/toxicity , Cerebellum/pathology , Ethanol/toxicity , Neurons , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Glucose/deficiency , Hypoxia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Pregnancy , Rats, Wistar
2.
Biomed Res ; 34(6): 329-42, 2013.
Article in English | MEDLINE | ID: mdl-24389409

ABSTRACT

Adult rat dorsal root ganglion (DRG) neurons cultured in the presence of 100-ng/mL NGF were reported to show spontaneous action potentials in the cell-attached recording. In this study, underlying mechanisms were examined in the whole-cell and outside-out voltage clamp recording. In 75% neurons with on-cell firing, transient inward current spikes were repetitively recorded in the voltage clamp mode at -50 mV in the whole-cell configuration (named "Isp"). Isp with stable amplitudes occurred in an all-or-none fashion, and was abolished by TTX (< 100 nM), lidocaine (< 1 mM) and a reduction of extracellular Na(+) (154 to 100 mM) in an all-or-none fashion, suggesting that Isp reflects spontaneous dicharges occurring at the loosely voltage-clamped regions. Isp was also observed in the excised outside-out patches and the kinetics and the sensitivity to TTX and lidocaine resembled those in the whole-cell. Spontaneous action potentials were also recorded in the current clamp mode. Small subthreshold spikes often preceded the action potentials. When the localized discharge affected a whole-somatic membrane potential to overcome a threshold, the action potential generated. These results indicate that the triggering sources of the action potential exist in the somatic membrane itself in NGF-treated DRG neurons.


Subject(s)
Ganglia, Spinal/drug effects , Nerve Growth Factor/pharmacology , Neurons/drug effects , Action Potentials , Animals , Cations, Monovalent , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Lidocaine/pharmacology , Male , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Tetrodotoxin/pharmacology
3.
Brain Res ; 1483: 1-12, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-22975133

ABSTRACT

Arginine-vasopressin (AVP) is a nonapeptide of hypothalamic origin that has been shown to exert many important cognitive and physiological functions in neurons and terminals of both the central and peripheral nervous system (CNS and PNS). Here we report for the first time that AVP induced an increase in intracellular Ca²âº concentration ([Ca²âº](i)) in non-neuronal cells isolated from the rat dorsal root ganglion (DRG) and cultured in vitro. The ratiometric [Ca²âº](i) measurements showed that AVP evoked [Ca²âº](i) responses in the non-neuronal cells and these concentration-dependent (100 pM to 1 µM) responses increased with days in vitro in culture, reaching a maximum amplitude after 4-5 day. Immunostaining by anti-S-100 antibody revealed that more than 70% of S-100 positive cells were AVP-responsive, indicating that glial cells responded to AVP and increased their [Ca²âº](i). The responses were inhibited by depletion of the intracellular Ca²âº stores or in the presence of inhibitors of phospholipase C, indicating a metabotropic response involving inositol trisphosphate, and were mediated by the V1 subclass of AVP receptors, as evidenced by the use of the specific blockers for V1 and OT receptors, (d(CH2)5¹,Tyr(Me)²,Arg8)-Vasopressin and (d(CH2)5¹,Tyr(Me)²,Thr4,Orn8,des-Gly-NH29)-Vasotocin, respectively. V(1a) but not V(1b) receptor mRNA was expressed sustainably through the culture period in cultured DRG cells. These results suggest that AVP modulates the activity of DRG glial cells via activation of V(1a) receptor.


Subject(s)
Calcium/metabolism , Ganglia, Spinal/cytology , Intracellular Fluid/metabolism , Neuroglia/drug effects , Vasoconstrictor Agents/pharmacology , Vasopressins/pharmacology , Animals , Arginine Vasopressin/analogs & derivatives , Arginine Vasopressin/pharmacology , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Intracellular Fluid/drug effects , Male , Potassium Chloride/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , S100 Proteins/metabolism , Time Factors
4.
PLoS One ; 7(6): e39302, 2012.
Article in English | MEDLINE | ID: mdl-22768071

ABSTRACT

BACKGROUND: Neurogenesis persists throughout life in the adult mammalian brain. Because neurogenesis can only be assessed in postmortem tissue, its functional significance remains undetermined, and identifying an in vivo correlate of neurogenesis has become an important goal. By studying pentylenetetrazole-induced brain stimulation in a rat model of kindling we accidentally discovered that 25±1 days periodic stimulation of Sprague-Dawley rats led to a highly efficient increase in seizure susceptibility. METHODOLOGY/PRINCIPAL FINDINGS: By EEG, RT-PCR, western blotting and immunohistochemistry, we show that repeated convulsive seizures with a periodicity of 25±1 days led to an enrichment of newly generated neurons, that were BrdU-positive in the dentate gyrus at day 25±1 post-seizure. At the same time, there was a massive increase in the number of neurons expressing the migratory marker, doublecortin, at the boundary between the granule cell layer and the polymorphic layer in the dorsal hippocampus. Some of these migrating neurons were also positive for NeuN, a marker for adult neurons. CONCLUSION/SIGNIFICANCE: Our results suggest that the increased susceptibility to seizure at day 25±1 post-treatment is coincident with a critical time required for newborn neurons to differentiate and integrate into the existing hippocampal network, and outlines the importance of the dorsal hippocampus for seizure-related neurogenesis. This model can be used as an in vivo correlate of neurogenesis to study basic questions related to neurogenesis and to the neurogenic mechanisms that contribute to the development of epilepsy.


Subject(s)
Hippocampus/metabolism , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Pentylenetetrazole/administration & dosage , Pentylenetetrazole/pharmacology , Animals , Biomarkers/metabolism , Bromodeoxyuridine/metabolism , Cell Count , Cell Lineage/drug effects , Disease Susceptibility , Dose-Response Relationship, Drug , Doublecortin Domain Proteins , Doublecortin Protein , Drug Administration Schedule , Electroencephalography , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Kindling, Neurologic , Male , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/pharmacology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phenotype , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Seizures/pathology , Seizures/physiopathology
6.
Neurobiol Aging ; 33(11): 2692-703, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22405041

ABSTRACT

Oscillations in hippocampal neuronal networks in the gamma frequency band have been implicated in various cognitive tasks and we showed previously that aging reduces the power of such oscillations. Here, using submerged hippocampal slices allowing simultaneous electrophysiological recordings and imaging, we studied the correlation between the kainate-evoked gamma oscillation and mitochondrial activity, as monitored by rhodamine 123. We show that the initiation of kainate-evoked gamma oscillations induces mitochondrial depolarization, indicating a metabolic response. Aging had an opposite effect on these parameters: while depressing the gamma oscillation strength, it increases mitochondrial depolarization. Also, in the aged neurons, kainate induced significantly larger Ca2+ signals. In younger slices, acute mitochondrial depolarization induced by low concentrations of mitochondrial protonophores strongly, but reversibly, inhibits gamma oscillations. These data indicating that the complex network activity required by the maintenance of gamma activity is susceptible to changes and modulations in mitochondrial status.


Subject(s)
Aging , Hippocampus/drug effects , Kainic Acid/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Animals , Calcium/metabolism , Electrophysiological Phenomena/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology
7.
Cell Calcium ; 51(3-4): 207-11, 2012.
Article in English | MEDLINE | ID: mdl-22385835

ABSTRACT

This special issue on Ca(2+) signalling in neuroendocrine cells is an opportunity to assess, through a range of first-class review articles, the complex world of endocrine signalling, a complexity that is probably best captured by calling it "diversity in unity". The unity comes from the fact that all the endocrine cells are excitable cells, able to generate action potentials and are using Ca(2+) as an essential informational molecule, coupling cell stimulation with the activation of secretion, through the exocytotic process. The 'diversity' element, illustrated by almost all the reviews, stems from the modalities employed to achieve the increase in cytosolic Ca(2+) signal, the balance between the participation of Ca(2+) entry through the plasma membrane voltage-operated Ca(2+) channels and the release of Ca(2+) from intracellular Ca(2+) stores, and the cross-talk between the Ca(2+) and cyclic AMP signalling pathways.


Subject(s)
Calcium Signaling , Cyclic AMP/metabolism , Neuroendocrine Cells/physiology , Action Potentials , Animals , Calcium Channels/metabolism , Humans , Receptor Cross-Talk , Secretory Pathway
8.
Cell Calcium ; 51(3-4): 293-9, 2012.
Article in English | MEDLINE | ID: mdl-22386684

ABSTRACT

Every cell or neuronal type utilizes its own specific organization of its Ca(2+) homeostasis depending on its specific function and its physiological needs. The magnocellular neurones, with their somata situated in the supraoptic and paraventricular nuclei of the hypothalamus and their nerve terminals populating the posterior hypophysis (neural lobe) are a typical and classical example of a neuroendocrine system, and an important experimental model for attempting to understand the characteristics of the neuronal organization of Ca(2+) homeostasis. The magnocellular neurones synthesize, in a cell specific manner, two neurohormones: arginine-vasopressin (AVP) and oxytocin (OT), which can be released, in a strict Ca(2+)-dependent manner, both at the axonal terminals, in the neural lobe, and at the somatodendritic level. The two types of neurones show also distinct type of bioelectrical activity, associated with specific secretory patterns. In these neurones, the Ca(2+) homeostatic pathways such as the Na(+)/Ca(2+) exchanger (NCX), the endoplasmic reticulum (ER) Ca(2+) pump, the plasmalemmal Ca(2+) pump (PMCA) and the mitochondria are acting in a complementary fashion in clearing Ca(2+) loads that follow neuronal stimulation. The somatodendritic AVP and OT release closely correlates with intracellular Ca(2+) dynamics. More importantly, the ER Ca(2+) stores play a major role in Ca(2+) homeostatic mechanism in identified OT neurones. The balance between the Ca(2+) homeostatic systems that are in the supraoptic neurones differ from those active in the terminals, in which mainly Ca(2+) extrusion through the Ca(2+) pump in the plasma membrane and uptake by mitochondria are active. In both AVP and OT nerve terminals, no functional ER Ca(2+) stores can be evidenced experimentally. We conclude that the physiological significance of the complexity of Ca(2+) homeostatic mechanisms in the somatodendritic region of supraoptic neurones and their terminals can be multifaceted, attributable, in major part, to their specialized electrical activity and Ca(2+)-dependent neurohormone release.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Nerve Endings/metabolism , Pituitary Gland/physiology , Presynaptic Terminals/metabolism , Animals , Homeostasis , Humans , Neurosecretion , Oxytocin/metabolism , Receptor Cross-Talk , Vasopressins/metabolism
9.
Neurobiol Aging ; 32(5): 956-65, 2011 May.
Article in English | MEDLINE | ID: mdl-19523715

ABSTRACT

Normal ageing-associated spatial memory impairment has been linked to subtle changes in the hippocampal network. Here we test whether the age-dependent reduction in gamma oscillations can be explained by the changes in intrinsic properties of hippocampal interneurons. Kainate-induced gamma oscillations, but not spontaneous gamma oscillations, were reduced in slices from aged mice. CA3 interneurons were recorded in slices from young and aged mice using Fura-2-filled pipettes. Passive membrane properties, firing properties, medium- and slow-afterhyperpolarisation amplitudes, basal [Ca(2+)](i) and firing-induced [Ca(2+)](i) transients were not different with ageing. Kainate caused a larger depolarisation and increase in [Ca(2+)](i) signal in aged interneurons than in young ones. In contrast to young interneurons, kainate increased the medium- and slow-afterhyperpolarisation and underlying [Ca(2+)](i) transient in aged interneurons. Modulating the slow-afterhyperpolarisation by modulating L-type calcium channels with BAY K 8644 and nimodipine suppressed and potentiated, respectively, kainate-induced gamma oscillations in young slices. The age-dependent and stimulation-dependent increase in basal [Ca(2+)](i), firing-induced [Ca(2+)](i) transient and associated afterhyperpolarisation may reduce interneuron excitability and contribute to an age-dependent impairment of hippocampal gamma oscillations.


Subject(s)
Aging/physiology , CA3 Region, Hippocampal/physiology , Interneurons/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Aging/drug effects , Animals , CA3 Region, Hippocampal/drug effects , Calcium/physiology , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/pharmacology , Chelating Agents/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Fura-2/pharmacology , Interneurons/drug effects , Kainic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Nimodipine/pharmacology , Periodicity
10.
Neurobiol Learn Mem ; 95(3): 221-30, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21093596

ABSTRACT

Neuronal synchronisation at gamma frequencies (30-100 Hz) has been implicated in cognition and memory. Gamma oscillations can be studied in various in vitro models, but their in vivo validity and their relationship with reference memory remains to be proven. By using the natural variation of wild type C57bl/6J mice, we assessed the relationships between reference memory and gamma oscillations recorded in hippocampal area CA3 in vivo and in vitro. Local field potentials (LFPs) were recorded from area CA3 in behaviourally-characterised freely moving mice, after which hippocampal slices were prepared for recordings in vitro of spontaneous gamma oscillations and kainate-induced gamma oscillations in CA3. The gamma-band power of spontaneous oscillations in vitro correlated with that of CA3 LFP oscillations during inactive behavioural states. The gamma-band power of kainate-induced oscillations correlated with the activity-dependent increase in CA3 LFP gamma-band power in vivo. Kainate-induced gamma-band power correlated with Barnes circular platform performance and object location recognition, but not with object novelty recognition. Kainate-induced gamma-band power was larger in mice that recognised the aversive context, but did not correlate with passive avoidance delay. The correlations between behavioural and electrophysiological measures obtained from the same animals show that the gamma-generating capacity of the CA3 network in vitro is a useful index of in vivo gamma strength and supports an important role of CA3 gamma oscillations in spatial reference memory.


Subject(s)
CA3 Region, Hippocampal/physiology , Discrimination Learning/physiology , Evoked Potentials/physiology , Memory/physiology , Spatial Behavior/physiology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , CA3 Region, Hippocampal/drug effects , Discrimination Learning/drug effects , Electroencephalography Phase Synchronization , Evoked Potentials/drug effects , Excitatory Amino Acid Agonists/pharmacology , In Vitro Techniques , Kainic Acid/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Mice , Mice, Inbred C57BL , Spatial Behavior/drug effects , Statistics, Nonparametric
11.
Cell Calcium ; 47(2): 158-64, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20045187

ABSTRACT

Normal brain ageing is associated with a varying degree of cognitive impairment. Although ageing is a complex, multifactorial process, and no single process could explain the ageing phenotype, a number of processes and homeostatic systems, due to their central roles in cellular physiology, have been identified as playing important roles in the process of normal ageing. In this review we revisit the basic tenets of the Ca2+ hypothesis of neuronal ageing and stress the major conceptual changes that occurred between the time of its original proposal and now, in particular in respect to the extent of neuronal loss in normal ageing. We provide a general overview of the most important ageing-associated changes in neuronal Ca2+ homeostasis and then discuss in some detail how such homeostatic changes are affecting basic neuronal properties, such as intrinsic excitability and how, by extension, such changes could lead to significant perturbations in the activity of whole neuronal network ensembles. Since some of these network activities, such as the synchronisation of neuronal activity in the gamma frequency range, have been linked to learning and cognition, understanding the metabolic substrates and homeostatic dysregulation that underpin them could provide a novel basis for attempts at counteracting the cognitive decline of older individuals.


Subject(s)
Aging/metabolism , Calcium Signaling , Calcium/metabolism , Cellular Senescence , Neurons/metabolism , Aging/pathology , Animals , Brain/pathology , Electron Transport , Humans , Nerve Net/metabolism , Neurons/pathology
12.
J Neurol Sci ; 283(1-2): 99-106, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19264328

ABSTRACT

The current pathogenic scenarios of different types of dementia are based on a number of common mechanisms of neurodegeneration, such as accumulation of abnormal proteins (within or outside cells), mitochondrial dysfunction and oxidative stress, calcium homeostasis dysregulation, early synaptic disconnection and late apoptotic cell death. Ageing itself is associated with mild cognitive deterioration, probably due to subtle multifactorial changes resulting in a global decrease of a functional brain reserve. Increased age is a risk factor for neurodegeneration and key pathological features of dementia can also be found in aged brains. One of the underexplored brain structures in ageing and dementia is the blood-brain barrier (BBB), a complex cellular gate which regulates tightly the transport of molecules into and from the central nervous system. Disruption of this barrier is now increasingly documented not only in brain vascular disease but also in ageing and neurodegenerative disorders. To date, such evidence points mainly at an association between various dementia forms and disruption of the BBB. But, in reviewing such results, and taking into account the exquisite sensitivity of neuronal function to the composition of the interstitial brain fluid (IBF), which is regulated by the BBB, we would like to propose the existence of a possible causal link between alterations of BBB and conditions associated with cognitive decline.


Subject(s)
Aging/physiology , Alzheimer Disease/physiopathology , Blood-Brain Barrier/physiopathology , Dementia, Vascular/physiopathology , Animals , Cerebrovascular Disorders/physiopathology , Humans , Models, Neurological , Parkinson Disease/physiopathology
13.
J Neurosci ; 28(15): 3877-86, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18400886

ABSTRACT

Prion protein (PrP) is a normal component of neurons, which confers susceptibility to prion diseases. Despite its evolutionary conservation, its normal function remains controversial. PrP-deficient (Prnp(0/0)) mice have weaker afterhyperpolarizations (AHPs) in cerebellar and hippocampal neurons. Here we show that the AHP impairment in hippocampal CA1 pyramidal cells is selective for the slow AHP, and is not caused by an impairment of either voltage-gated Ca(2+) channels or Ca(2+)-activated K(+) channels. Instead, Prnp(0/0) neurons have twofold to threefold stronger Ca(2+) buffering and double the Ca(2+) extrusion rate. In Prnp(0/0) neurons thapsigargin abolished the stronger Ca(2+) buffering and extrusion, and thapsigargin or cyclopiazonic acid abolished the weakening of the slow AHPs. These data implicate sarcoplasmic/endoplasmic reticulum calcium ATPase in the enhanced Ca(2+) buffering, and extrusion into the endoplasmic reticulum, which contains substantial amounts of PrP in wild-type mice. Altered Ca(2+) homeostasis can explain several phenotypes identified in Prnp(0/0) mice.


Subject(s)
Calcium/metabolism , Hippocampus/physiology , Pregnancy Proteins/deficiency , Pyramidal Cells , Action Potentials , Animals , Buffers , Calcium Signaling , Electrophysiology , Enzyme Inhibitors/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Homeostasis , In Vitro Techniques , Indoles/pharmacology , Mice , Mice, Knockout , Patch-Clamp Techniques , Photolysis , Reaction Time , Thapsigargin/pharmacology
15.
Aging Cell ; 6(3): 267-73, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17517038

ABSTRACT

Aging is a complex, multifactorial process. One of the features of normal aging of the brain is a decline in cognitive functions and much experimental attention has been devoted to understanding this process. Evidence accumulated in the last decade indicates that such functional changes are not due to gross morphological alterations, but to subtle functional modification of synaptic connectivity and intracellular signalling and metabolism. Such synaptic modifications are compatible with a normal level of activity and allow the maintenance of a certain degree of functional reserve. This is in contrast to the changes in various neurodegenerative diseases, characterized by significant neuronal loss and dramatic and irreversible functional deficit. This whole special issue has been initiated with the intention of focusing on the processes of normal brain aging. In this review, we present data that shows how subtle changes in Ca(2+) homeostasis or in the state of various Ca(2+)-dependent processes or molecules, which occur in aging can have significant functional consequences.


Subject(s)
Aging , Calcium/metabolism , Cognition Disorders/pathology , Cognition , Homeostasis , Aged , Calcium Signaling , Cognition Disorders/metabolism , Humans , Models, Biological , Neurodegenerative Diseases/metabolism , Signal Transduction , Synaptic Transmission
16.
Biochem J ; 398(3): 547-56, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16753042

ABSTRACT

S1P (sphingosine 1-phosphate) receptor expression and the effects of S1P on migration were studied in one papillary (NPA), two follicular (ML-1, WRO) and two anaplastic (FRO, ARO) thyroid cancer cell lines, as well as in human thyroid cells in primary culture. Additionally, the effects of S1P on proliferation, adhesion and calcium signalling were addressed in ML-1 and FRO cells. All cell types expressed multiple S1P receptors. S1P evoked intracellular calcium signalling in primary cultures, ML-1 cells and FRO cells. Neither proliferation nor migration was affected in primary cultures, whereas S1P partly inhibited proliferation in ML-1 and FRO cells. Low nanomolar concentrations of S1P inhibited migration in FRO, WRO and ARO cells, but stimulated ML-1 cell migration. Consistently, S1P1 and S1P3, which mediate migratory responses, were strongly expressed in ML-1 cells, and S1P2, which inhibits migration, was the dominating receptor in the other cell lines. The migratory effect in ML-1 cells was mediated by G(i) and phosphatidylinositol 3-kinase. Both S1P and the S1P1-specific agonist SEW-2871 induced Akt phosphorylation at Ser473. However, SEW-2871 failed to stimulate migration, whereas the S1P1/S1P3 antagonist VPC 23019 inhibited S1P-induced migration. The results suggest that aberrant S1P receptor expression may enhance thyroid cancer cell migration and thus contribute to the metastatic behaviour of some thyroid tumours.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Receptors, Lysosphingolipid/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Calcium/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Thyroid Gland/cytology , Thyroid Neoplasms/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
17.
Philos Trans R Soc Lond B Biol Sci ; 360(1464): 2347-54, 2005 Dec 29.
Article in English | MEDLINE | ID: mdl-16321805

ABSTRACT

Normal ageing is associated with a degree of decline in a number of cognitive functions. Apart from the issues raised by the current attempts to expand the lifespan, understanding the mechanisms and the detailed metabolic interactions involved in the process of normal neuronal ageing continues to be a challenge. One model, supported by a significant amount of experimental evidence, views the cellular ageing as a metabolic state characterized by an altered function of the metabolic triad: mitochondria-reactive oxygen species (ROS)-intracellular Ca2+. The perturbation in the relationship between the members of this metabolic triad generate a state of decreased homeostatic reserve, in which the aged neurons could maintain adequate function during normal activity, as demonstrated by the fact that normal ageing is not associated with widespread neuronal loss, but become increasingly vulnerable to the effects of excessive metabolic loads, usually associated with trauma, ischaemia or neurodegenerative processes. This review will concentrate on some of the evidence showing altered mitochondrial function with ageing and also discuss some of the functional consequences that would result from such events, such as alterations in mitochondrial Ca2+ homeostasis, ATP production and generation of ROS.


Subject(s)
Aging/physiology , Brain/physiology , Homeostasis/physiology , Mitochondria/physiology , Models, Biological , Neurons/physiology , Adenosine Triphosphate/biosynthesis , Calcium/metabolism , Humans , Mitochondria/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Reactive Oxygen Species/metabolism
20.
Trends Neurosci ; 27(10): 614-20, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15374673

ABSTRACT

Understanding the cellular mechanisms that characterize the functional changes of the aged brain is an ongoing and formidable challenge for the neuroscience community. Evidence now links changes in Ca(2+) influx and homeostasis with perturbations induced by the aging process in the function of the main intracellular organelles involved in Ca(2+) regulation: the endoplasmic reticulum and mitochondria. New perspectives are also offered by recent gene microarray studies, illustrating the multifactorial nature of the aging process.


Subject(s)
Aging/physiology , Brain/physiology , Calcium Signaling/physiology , Calcium/metabolism , Gene Expression Regulation/physiology , Neurons/physiology , Animals , Endoplasmic Reticulum/physiology , Homeostasis/physiology , Humans , Mitochondria/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...