Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36233268

ABSTRACT

A crucial etiological component in fetal programming is early nutrition. Indeed, early undernutrition may cause a chronic increase in blood pressure and cardiovascular diseases, including stroke and heart failure. In this regard, current evidence has sustained several pathological mechanisms involving changes in central and peripheral targets. In the present review, we summarize the neuroendocrine and neuroplastic modifications that underlie maladaptive mechanisms related to chronic hypertension programming after early undernutrition. First, we analyzed the role of glucocorticoids on the mechanism of long-term programming of hypertension. Secondly, we discussed the pathological plastic changes at the paraventricular nucleus of the hypothalamus that contribute to the development of chronic hypertension in animal models of prenatal undernutrition, dissecting the neural network that reciprocally communicates this nucleus with the locus coeruleus. Finally, we propose an integrated and updated view of the main neuroendocrine and central circuital alterations that support the occurrence of chronic increases of blood pressure in prenatally undernourished animals.


Subject(s)
Hypertension , Malnutrition , Prenatal Exposure Delayed Effects , Animals , Blood Pressure , Female , Glucocorticoids/physiology , Humans , Malnutrition/pathology , Paraventricular Hypothalamic Nucleus , Pregnancy , Prenatal Exposure Delayed Effects/pathology
2.
Int J Mol Sci ; 23(17)2022 Sep 03.
Article in English | MEDLINE | ID: mdl-36077493

ABSTRACT

A loss of neuroplastic control on nucleus accumbens (NAc) neuronal activity exerted by the medial prefrontal cortex (mPFC) through long-term depression (LTD) is involved in triggering drug-seeking behavior and relapse on several substances of abuse due to impaired glutamate homeostasis in tripartite synapses of the nucleus accumbens (NAc) core. To test whether this maladaptive neuroplastic mechanism underlies the addiction-like behavior induced in young mice by a high-fat diet (HFD), we utilized 28-days-old male mice fed HFD ad-libitum over 2 weeks, followed by 5 days of HFD abstinence. Control groups were fed a regular diet. HFD fed mice showed increased ΔFosB levels in the NAc core region, whereas LTD triggered from the mPFC became suppressed. Interestingly, LTD suppression was prevented by an i.p. injection of 100 mg/kg N-acetylcysteine 2.5 h before inducing LTD from the mPFC. In addition, excessive weight gain due to HFD feeding was diminished by adding 2mg/mL N-acetylcysteine in drinking water. Those results show a loss of neuroplastic mPFC control over NAc core activity induced by HFD consumption in young subjects. In conclusion, ad libitum consumption of HFD can lead to neuroplastic changes an addiction-like behavior that can be prevented by N-acetylcysteine, helping to decrease the rate of excessive weight gain.


Subject(s)
Diet, High-Fat , Nucleus Accumbens , Acetylcysteine/pharmacology , Animals , Diet, High-Fat/adverse effects , Humans , Male , Mice , Obesity/drug therapy , Obesity/etiology , Obesity/prevention & control , Prefrontal Cortex , Weight Gain
3.
Molecules ; 26(12)2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34207980

ABSTRACT

Prenatally malnourished rats develop hypertension in adulthood, in part through increased α1-adrenoceptor-mediated outflow from the paraventricular nucleus (PVN) to the sympathetic system. We studied whether both α1-adrenoceptor-mediated noradrenergic excitatory pathways from the locus coeruleus (LC) to the PVN and their reciprocal excitatory CRFergic connections contribute to prenatal undernutrition-induced hypertension. For that purpose, we microinjected either α1-adrenoceptor or CRH receptor agonists and/or antagonists in the PVN or the LC, respectively. We also determined the α1-adrenoceptor density in whole hypothalamus and the expression levels of α1A-adrenoceptor mRNA in the PVN. The results showed that: (i) agonists microinjection increased systolic blood pressure and heart rate in normotensive eutrophic rats, but not in prenatally malnourished subjects; (ii) antagonists microinjection reduced hypertension and tachycardia in undernourished rats, but not in eutrophic controls; (iii) in undernourished animals, antagonist administration to one nuclei allowed the agonists recover full efficacy in the complementary nucleus, inducing hypertension and tachycardia; (iv) early undernutrition did not modify the number of α1-adrenoceptor binding sites in hypothalamus, but reduced the number of cells expressing α1A-adrenoceptor mRNA in the PVN. These results support the hypothesis that systolic pressure and heart rate are increased by tonic reciprocal paraventricular-coerulear excitatory interactions in prenatally undernourished young-adult rats.


Subject(s)
Hypertension/pathology , Hypothalamus/metabolism , Malnutrition/complications , Paraventricular Hypothalamic Nucleus/physiopathology , Prenatal Exposure Delayed Effects/pathology , Animals , Blood Pressure , Disease Models, Animal , Female , Heart Rate , Hypertension/etiology , Hypertension/physiopathology , Male , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Rats
4.
Behav Brain Res ; 363: 182-190, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30721762

ABSTRACT

Moderate reduction of dietary protein (from 25% to 8% casein) in pregnant rats, calorically compensated by carbohydrates, gives rise to 'hidden prenatal malnutrition' (HPM) in the offspring since it does not alter body and brain weights of pups at birth. However, this dietary treatment leads to decreased ß-adrenoceptor signaling and brain derived neurotrophic factor (BDNF) levels in the pup' brain, altogether with defective cortical long-term potentiation (LTP) and lowered visuospatial memory performance. Since early postnatal environmental enrichment (EE) has been shown to exert plastic effects on the developing brain and neuroprotection both on cognition and on structural properties of the neocortex, in the present study we addressed the question of whether early postnatal EE during the lactation period could exert compensatory changes in the expression of ®-adrenergic receptors and BDNF in the neocortex of HPM rats, and if these effects are associated with an improvement or even a restore of both neocortical LTP in vivo and cognitive performance induced by HPM. The results obtained show that EE restored ß-adrenoceptor density, BDNF expression and the ability to support LTP at prefrontal and occipital cortices of HPM rats. Besides, EE improved learning performance in visuospatial and operant conditioning tasks. The latter support the notion that adequate maternal protein nutrition during pregnancy is required for proper brain development and function. Further, the results highlight the role of environmental enrichment during early postnatal life in increasing later brain plasticity and exerting neuroprotection against brain deficits induced by prenatal malnutrition.


Subject(s)
Cerebral Cortex/physiology , Learning/physiology , Postnatal Care/methods , Animals , Animals, Newborn/psychology , Brain-Derived Neurotrophic Factor/metabolism , Cognition/physiology , Female , Long-Term Potentiation/physiology , Male , Malnutrition/physiopathology , Memory/physiology , Neocortex/physiopathology , Neuronal Plasticity/physiology , Occipital Lobe/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/metabolism
5.
Nutr Rev ; 77(2): 65-80, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30445479

ABSTRACT

Studies in rats have shown that a decrease in either protein content or total dietary calories results in molecular, structural, and functional changes in the cerebral cortex and hippocampus, among other brain regions, which lead to behavioral disturbances, including learning and memory deficits. The neurobiological bases underlying those effects depend at least in part on fetal programming of the developing brain, which in turn relies on epigenetic regulation of specific genes via stable and heritable modifications of chromatin. Prenatal malnutrition also leads to epigenetic programming of obesity, and obesity on its own can lead to poor cognitive performance in humans and experimental animals, complicating understanding of the factors involved in the fetal programming of neuroplasticity deficits. This review focuses on the role of epigenetic mechanisms involved in prenatal malnutrition-induced brain disturbances, which are apparent at a later postnatal age, through either a direct effect of fetal programming on brain plasticity or an indirect effect on the brain mediated by the postnatal development of obesity.


Subject(s)
Epigenesis, Genetic , Fetal Development , Fetal Nutrition Disorders , Prenatal Exposure Delayed Effects , Animals , Brain/growth & development , Female , Humans , Metabolic Syndrome , Neuronal Plasticity , Obesity , Pregnancy
6.
Neurobiol Learn Mem ; 119: 1-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25464009

ABSTRACT

Moderate reduction in dietary protein composition of pregnant rats from 25% to 8% casein, calorically compensated by carbohydrates, has been described as a "hidden malnutrition" because it does not alter body and brain weights of pups at birth. However, this dietary treatment leads to altered central noradrenergic systems, impaired cortical long-term potentiation (LTP) and worsened visuo-spatial memory performance. Given the increasing interest on the role played by ß2-adrenoceptors (ß2-ARs) on brain plasticity, the present study aimed to address the following in hidden-malnourished and eutrophic control rats: (i) the expression levels of ß2-ARs in the frontal cortex determined by immunohistochemistry, and (ii) the effect of the ß2 selective agonist clenbuterol on both LTP elicited in vivo in the prefrontal cortex and visuospatial performance measured in an eight-arm radial maze. Our results showed that, prenatally malnourished rats exhibited a significant reduction of neocortical ß2-AR expression in adulthood. Concomitantly, they were unable to elicit and maintain prefrontal cortex LTP and exhibited lower visuospatial learning performance. Administration of clenbuterol (0.019, 0.038 and 0.075 mg/kg i.p.) enhanced LTP in malnourished and control animals and restored visuospatial learning performance in malnourished but not in normal rats, in a dose-dependent manner. The results suggest that decreased density of neocortical ß2-ARs during postnatal life, subsequent to hidden prenatal malnutrition might affect some synaptic networks required to elicit neocortical LTP and form visuospatial memory, since those neuroplastic deficits were counteracted by ß2-AR stimulation.


Subject(s)
Frontal Lobe/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Protein-Energy Malnutrition/physiopathology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/administration & dosage , Animals , Clenbuterol/administration & dosage , Female , Frontal Lobe/metabolism , Long-Term Potentiation/drug effects , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Protein-Energy Malnutrition/metabolism , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley , Spatial Memory/drug effects , Spatial Memory/physiology
7.
Neurobiol Learn Mem ; 107: 13-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24211700

ABSTRACT

Obesity is a worldwide epidemic that is increasing at an alarming rate. One of its causes is the increased availability and consumption of diets rich in fat. In the present study, we investigated the effects of short-term consumption of a high fat diet (HFD) on dietary preferences in Swiss CD1 mice and its relation in time to specific metabolic effects. Mice that were weaned 21days postpartum and fed a chow diet for one week were afterward subjected to a diet preference test for 5days, exposed to both a regular diet (RD) and HFD. We found that mice did not show any preferences. In a second experiment, two groups of mice that were weaned 21days postpartum and subjected to a chow diet for one week were fed either RD or HFD for 18days, and a diet preference test was performed for 5days. After this short-term consumption of HFD, mice preferred HFD, while mice subjected to RD did not show any preference. Importantly, no differences in blood glucose levels were found between the groups prior to and after the experiments. The results support our hypothesis that the preference for HFD is not a spontaneous behavior in CD1 mice, but it can be observed after short-term consumption; additionally, this preference develops before metabolic effects appear. Finally, this preference for HFD could not be observed when the mice were i.p. injected daily with low doses of the NMDA receptor antagonists, ketamine, ifenprodil or MK-801 during the HFD feeding period. These data suggest that acquisition of dietary preference for HFD is a NMDA receptor-dependent learning process.


Subject(s)
Diet, High-Fat , Food Preferences/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Blood Glucose/analysis , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Male , Mice , Mice, Inbred Strains , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
8.
Neurobiol Learn Mem ; 98(3): 228-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22892388

ABSTRACT

Moderate reduction in the protein content of the mother's diet calorically compensated by carbohydrates (the so-called "hidden" prenatal malnutrition) leads to increased neocortical expression of the α(2C)-adrenoceptor subtype, together with decreased cortical release of noradrenaline and impaired long-term potentiation (LTP) and visuospatial memory performance during the rat postnatal life. In order to study whether overexpression of the α(2C)-adrenoceptor subtype is causally related to the decreased indices of neocortical plasticity found in prenatally malnourished rats, we evaluated the effect of intracortical (occipital cortex) administration of an antisense oligodeoxynucleotide (ODN) raised against the α(2C)-adrenoceptor mRNA on the LTP elicited in vivo in the occipital cortex of hidden prenatally malnourished rats. In addition, we compare the effect of the antisense ODN to that produced by systemical administration of the subtype-nonselective α(2)-adrenoceptor antagonist atipamezole. Prenatal protein malnutrition led to impaired occipital cortex LTP together with increased expression of α(2C)-adrenoceptors (about twice Bmax) in the same cortical region. [(3)H]-rauwolscine binding assay showed that a 7-day intracortical antisense ODN treatment in the malnourished rats resulted in 50% knockdown of α(2C)-adrenoceptor expression and, in addition, completely rescued the ability of the occipital cortex to develop and maintain long-term potentiation. Atipamezole (0.3 mg/kg i.p.) also led to full recovery of neocortical LTP in malnourished rats. The present results argue in favor of our original hypothesis that the deleterious effect of prenatal malnutrition on neocortical plasticity in the adult progeny is in part consequence of increased neocortical α(2C)-adrenoceptor expression. This receptor subtype is known to be involved in the presynaptic control of noradrenaline release from central neurons, a neurotransmitter that critically influences LTP and memory formation.


Subject(s)
Long-Term Potentiation/physiology , Malnutrition/metabolism , Occipital Lobe/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prenatal Nutritional Physiological Phenomena/physiology , Receptors, Adrenergic, alpha-2/genetics , Animals , Female , Imidazoles/pharmacology , Long-Term Potentiation/drug effects , Male , Malnutrition/genetics , Malnutrition/physiopathology , Neurons/drug effects , Neurons/metabolism , Occipital Lobe/drug effects , Occipital Lobe/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/metabolism
9.
Biol Res ; 45(1): 87-92, 2012.
Article in English | MEDLINE | ID: mdl-22688989

ABSTRACT

The effect of prenatal malnutrition on the anatomy of the corpus callosum was assessed in adult rats (45-52 days old). In the prenatally malnourished animals we observed a significant reduction of the corpus callosum total area, partial areas, and perimeter, as compared with normal animals. In addition, the splenium of corpus callosum (posterior fifth) showed a significant decrease of fiber diameters in the myelinated fibers without changing density. There was also a significant decrease in diameter and a significant increase in density of unmyelinated fibers. Measurements of perimeter's fractal dimensions from sagittal sections of the brain and corpus callosum did not show significant differences between malnourished and control animals. These findings indicate that cortico-cortical connections are vulnerable to the prenatal malnutrition, and suggest this may affect interhemispheric conduction velocity, particularly in visual connections (splenium).


Subject(s)
Corpus Callosum/anatomy & histology , Malnutrition/pathology , Nerve Fibers/ultrastructure , Prenatal Exposure Delayed Effects/pathology , Animals , Body Weight/physiology , Control Groups , Corpus Callosum/physiology , Female , Male , Malnutrition/physiopathology , Nerve Fibers, Myelinated/ultrastructure , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley
10.
Biol. Res ; 45(1): 87-92, 2012. ilus, tab
Article in English | LILACS | ID: lil-626752

ABSTRACT

The effect of prenatal malnutrition on the anatomy of the corpus callosum was assessed in adult rats (45-52 days old). In the prenatally malnourished animals we observed a significant reduction of the corpus callosum total area, partial areas, and perimeter, as compared with normal animals. In addition, the splenium of corpus callosum (posterior fifth) showed a significant decrease of fiber diameters in the myelinated fibers without changing density. There was also a significant decrease in diameter and a significant increase in density of unmyelinated fibers. Measurements of perimeter's fractal dimensions from sagittal sections of the brain and corpus callosum did not show significant differences between malnourished and control animals. These findings indicate that cortico-cortical connections are vulnerable to the prenatal malnutrition, and suggest this may affect interhemispheric conduction velocity, particulary in visual connections (splenium).


Subject(s)
Animals , Female , Male , Pregnancy , Rats , Corpus Callosum/anatomy & histology , Malnutrition/pathology , Nerve Fibers/ultrastructure , Prenatal Exposure Delayed Effects/pathology , Body Weight/physiology , Control Groups , Corpus Callosum/physiology , Malnutrition/physiopathology , Nerve Fibers, Myelinated/ultrastructure , Prenatal Exposure Delayed Effects/physiopathology , Rats, Sprague-Dawley
11.
J Neurochem ; 119(2): 314-23, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21848869

ABSTRACT

Moderate reduction in the protein content of the mother's diet (hidden malnutrition) does not alter body and brain weights of rat pups at birth, but leads to dysfunction of neocortical noradrenaline systems together with impaired long-term potentiation and visuo-spatial memory performance. As ß1-adrenoceptors and downstream protein kinase signaling are critically involved in synaptic long-term potentiation and memory formation, we evaluated the ß1-adrenoceptor density and the expression of cyclic-AMP dependent protein kinase, calcium/calmodulin-dependent protein kinase and protein kinase Fyn, in the frontal cortex of prenatally malnourished adult rats. In addition, we also studied if ß1-adrenoceptor activation with the selective ß1 agonist dobutamine could improve deficits of prefrontal cortex long-term potentiation presenting these animals. Prenatally malnourished rats exhibited half of ß1-adrenoceptor binding, together with a 51% and 65% reduction of cyclic AMP-dependent protein kinase α and calcium/calmodulin-dependent protein kinase α expression, respectively, as compared with eutrophic animals. Administration of the selective ß1 agonist dobutamine prior to tetanization completely rescued the ability of the prefrontal cortex to develop and maintain long-term potentiation in the malnourished rats. Results suggest that under-expression of neocortical ß1-adrenoceptors and protein kinase signaling in hidden malnourished rats functionally affects the synaptic networks subserving prefrontal cortex long-term potentiation. ß1-adrenoceptor activation was sufficient to fully recover neocortical plasticity in the PKA- and calcium/calmodulin-dependent protein kinase II-deficient undernourished rats, possibly by producing extra amounts of cAMP and/or by recruiting alternative signaling cascades.


Subject(s)
Fetal Nutrition Disorders/physiopathology , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Receptors, Adrenergic, beta-1/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Body Weight/physiology , Brain/drug effects , Brain/embryology , Brain/growth & development , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet , Dobutamine/pharmacology , Electrophysiological Phenomena , Female , In Vitro Techniques , Long-Term Potentiation/drug effects , Male , Membranes/metabolism , Organ Size/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/metabolism , src-Family Kinases/metabolism
12.
Brain Res Bull ; 83(5): 238-44, 2010 Oct 30.
Article in English | MEDLINE | ID: mdl-20800665

ABSTRACT

Modafinil is a novel wake-promoting agent whose effects on cognitive performance have begun to be addressed at both preclinical and clinical level. The present study was designed to investigate in rats the effects of chronic modafinil administration on cognitive performance by evaluating: (i) working and reference memories in an Olton 4×4 maze, and (ii) learning of a complex operant conditioning task in a Skinner box. In addition, the effect of modafinil on the ability of the rat frontal cortex to develop long-term potentiation (LTP) was also studied. Chronic modafinil did not significantly modify working memory errors but decreased long-term memory errors on the Olton 4×4 maze, meaning that the drug may have a favourable profile on performance of visuo-spatial tasks (typically, a hippocampus-dependent task) when chronically administered. On the other hand, chronic modafinil resulted in a marked decrease of successful responses in a complex operant conditioning learning, which means that repeated administration of the drug influences negatively problem-solving abilities when confronting the rat to a sequencing task (typically, a prefrontal cortex-dependent task). In addition, in vivo electrophysiology showed that modafinil resulted in impaired capacity of the rat prefrontal cortex to develop LTP following tetanization. It is concluded that modafinil can improve the performance of spatial tasks that depend almost exclusively on hippocampal functioning, but not the performance in tasks including a temporal factor where the prefrontal cortex plays an important role. The fact that modafinil together with preventing operant conditioning learning was also able to block LTP induction in the prefrontal cortex, suggests that the drug could interfere some critical component required for LTP can be developed, thereby altering neuroplastic capabilities of the prefrontal cortex.


Subject(s)
Benzhydryl Compounds/pharmacology , Central Nervous System Stimulants/pharmacology , Learning/drug effects , Long-Term Potentiation/drug effects , Neocortex/drug effects , Neocortex/physiology , Animals , Conditioning, Operant/drug effects , Male , Memory, Short-Term/drug effects , Modafinil , Rats , Rats, Sprague-Dawley
13.
Neurosci Lett ; 483(1): 41-6, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20674672

ABSTRACT

Prenatal undernutrition induces hypertension later in life, possibly by disturbing the hypothalamo-pituitary-adrenal axis through programming decreased expression of hypothalamic glucocorticoid receptors. We examined the systolic blood pressure, heart rate and plasma corticosterone response to intra-paraventricular dexamethasone, mifepristone and corticosterone in eutrophic and prenatally undernourished young rats. Undernutrition was induced during fetal life by restricting the diet of pregnant mothers to 10 g daily (40% of diet consumed by well-nourished controls). At day 40 of postnatal life (i) intra-paraventricular administration of dexamethasone significantly reduced at least for 24h both the systolic pressure (-11.6%), the heart rate (-20.8%) and the plasma corticosterone (-40.0%) in normal animals, while producing lower effects (-5.5, -8.7, and -22.3%, respectively) on undernourished rats; (ii) intra-paraventricular administration of the antiglucocorticoid receptor ligand mifepristone to normal rats produced opposite effects (8.2, 20.3, and 48.0% increase, respectively) to those induced by dexamethasone, being these not significant in undernourished animals; (iii) intra-paraventricular corticosterone did not exert any significant effect. Results suggest that the low sensitivity of paraventricular neurons to glucocorticoid receptor ligands observed in prenatally undernourished rats could be due to the already reported glucocorticoid receptor expression, found in the hypothalamus of undernourished animals.


Subject(s)
Hypertension/etiology , Hypothalamus/metabolism , Malnutrition/complications , Prenatal Nutritional Physiological Phenomena/physiology , Receptors, Glucocorticoid/metabolism , Animals , Area Under Curve , Corticosterone/blood , Female , Heart Rate/physiology , Hypertension/metabolism , Hypertension/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/physiopathology , Male , Malnutrition/metabolism , Malnutrition/physiopathology , Neurons/metabolism , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Pregnancy , Rats , Rats, Wistar
14.
Brain Res Bull ; 82(5-6): 284-8, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20510341

ABSTRACT

beta-Adrenergic receptor stimulation can significantly facilitate synaptic potentiation in the hippocampus and enhance memory processes, but its effect on neocortical plastic mechanisms is less conclusive. In the present study we determined the effect of propranolol, a beta-adrenoceptor antagonist, on long-term potentiation (LTP) induced in vivo in rat occipital cortex by tetanizing stimulation of corpus callosum and observed a dose-dependent inhibition of LTP. We further administered propranolol through mini-osmotic pumps during 3 days, and observed the performance of rats in a complex operant conditioning learning paradigm and assessed the expression of brain-derived neurotrophic factor (BDNF) in the occipital cortex. Propranolol exposure depressed both the number of reinforced responses in the operant conditioning task and BDNF expression in occipital cortex. Taken together, our results suggest that propranolol impairs memory formation by inhibiting cortical LTP induction and associated BDNF expression.


Subject(s)
Gene Expression Regulation/physiology , Neocortex/physiology , Neuronal Plasticity/physiology , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Antagonists/pharmacology , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/metabolism , Conditioning, Operant/physiology , Corpus Callosum/physiology , Dose-Response Relationship, Drug , Electric Stimulation/methods , Gene Expression Regulation/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Neocortex/drug effects , Neuronal Plasticity/drug effects , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
15.
Biochem Biophys Res Commun ; 382(4): 740-4, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19318086

ABSTRACT

Amyloid precursor protein (APP) is a transmembrane glycoprotein widely expressed in mammalian tissues and plays a central role in Alzheimer's disease. However, its physiological function remains elusive. Cu(2+) binding and reduction activities have been described in the extracellular APP135-156 region, which might be relevant for cellular copper uptake and homeostasis. Here, we assessed Cu(2+) reduction and (64)Cu uptake in two human HEK293 cell lines overexpressing APP. Our results indicate that Cu(2+) reduction increased and cells accumulated larger levels of copper, maintaining cell viability at supra-physiological levels of Cu(2+) ions. Moreover, wild-type cells exposed to both Cu(2+) ions and APP135-155 synthetic peptides increased copper reduction and uptake. Complementation of function studies in human APP751 transformed Fre1 defective Saccharomyces cerevisiae cells rescued low Cu(2+) reductase activity and increased (64)Cu uptake. We conclude that Cu(2+) reduction activity of APP facilitates copper uptake and may represent an early step in cellular copper homeostasis.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Copper/metabolism , Copper/toxicity , Cell Line , Copper/analysis , Homeostasis/drug effects , Humans , Oxidation-Reduction/drug effects , Peptide Fragments/pharmacology
16.
Cell ; 129(3): 537-48, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17482547

ABSTRACT

Metabolic and inflammatory pathways crosstalk at many levels, and, while required for homeostasis, interaction between these pathways can also lead to metabolic dysregulation under conditions of chronic stress. Thus, we hypothesized that mechanisms might exist to prevent overt inflammatory responses during physiological fluctuations in nutrients or under nutrient-rich conditions, and we identified the six-transmembrane protein STAMP2 as a critical modulator of this integrated response system of inflammation and metabolism in adipocytes. Lack of STAMP2 in adipocytes results in aberrant inflammatory responses to both nutrients and acute inflammatory stimuli. Similarly, in whole animals, visceral adipose tissue of STAMP2(-/-) mice exhibits overt inflammation, and these mice develop spontaneous metabolic disease on a regular diet, manifesting insulin resistance, glucose intolerance, mild hyperglycemia, dyslipidemia, and fatty liver disease. We conclude that STAMP2 participates in integrating inflammatory and metabolic responses and thus plays a key role in systemic metabolic homeostasis.


Subject(s)
Adipocytes/metabolism , Food , Inflammation/metabolism , Membrane Proteins/metabolism , Metabolic Networks and Pathways , 3T3-L1 Cells , Adipose Tissue/metabolism , Animals , Cells, Cultured , Female , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Male , Membrane Proteins/genetics , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Mutation
17.
Am J Pathol ; 164(6): 2163-74, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161650

ABSTRACT

Neuropathological changes generated by human amyloid-beta peptide (Abeta) fibrils and Abeta-acetylcholinesterase (Abeta-AChE) complexes were compared in rat hippocampus in vivo. Results showed that Abeta-AChE complexes trigger a more dramatic response in situ than Abeta fibrils alone as characterized by the following features observed 8 weeks after treatment: 1). amyloid deposits were larger than those produced in the absence of AChE. In fact, AChE strongly stimulates rat Abeta aggregation in vitro as shown by turbidity measurements, Congo Red binding, as well as electron microscopy, suggesting that Abeta-AChE deposits observed in vivo probably recruited endogenous Abeta peptide; 2). the appearance of laminin expressing neurons surrounding Abeta-AChE deposits (such deposits are resistant to disaggregation by laminin in vitro); 3). an extensive astrocytosis revealed by both glial fibrillary acidic protein immunoreactivity and number counting of reactive hypertrophic astrocytes; and 4). a stronger neuronal cell loss in comparison with Abeta-injected animals. We conclude that the hippocampal injection of Abeta-AChE complexes results in the appearance of some features reminiscent of Alzheimer-like lesions in rat brain. Our studies are consistent with the notion that Abeta-AChE complexes are more toxic than Abeta fibrils and that AChE triggered some of the neurodegenerative changes observed in Alzheimer's disease brains.


Subject(s)
Acetylcholinesterase/toxicity , Amyloid beta-Protein Precursor/toxicity , Astrocytes/pathology , Gliosis/pathology , Hippocampus/pathology , Laminin/genetics , Neurons/pathology , Animals , Astrocytes/drug effects , Cattle , Cell Survival/drug effects , Glial Fibrillary Acidic Protein/analysis , Gliosis/chemically induced , Hippocampus/drug effects , Humans , Image Processing, Computer-Assisted , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence
18.
Prog Neurobiol ; 74(6): 323-49, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15649580

ABSTRACT

This review is focused on the structure and function of Alzheimer's amyloid deposits. Amyloid formation is a process in which normal well-folded cellular proteins undergo a self-assembly process that leads to the formation of large and ordered protein structures. Amyloid deposition, oligomerization, and higher order polymerization, and the structure adopted by these assemblies, as well as their functional relationship with cell biology are underscored. Numerous efforts have been directed to elucidate these issues and their relation with senile dementia. Significant advances made in the last decade in amyloid structure, dynamics and cell biology are summarized and discussed. The mechanism of amyloid neurotoxicity is discussed with emphasis on the Wnt signaling pathway. This review is focused on Alzheimer's amyloid fibrils in general and has been divided into two parts dealing with the structure and function of amyloid.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloid/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/physiopathology , Amyloid/toxicity , Animals , Apoptosis/physiology , Humans , Models, Biological , Models, Molecular , Necrosis/metabolism , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Signal Transduction/physiology
19.
Peptides ; 23(7): 1229-40, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12128080

ABSTRACT

Amyloid deposition in Alzheimer fibrils forms neurotoxic senile plaques in a process that may be modulated by associated proteins. In this work we demonstrate the ability of laminin-1 and laminin-2 to inhibit fibril formation and toxicity on cultured rat hippocampal neurons. We confirm that the laminin-1-derived peptide YFQRYLI inhibits efficiently both fibril formation and neurotoxicity and show that the IKVAV peptide inhibits amyloid neurotoxicity despite its slight inhibition of fibril formation. On other hand, laminin-1 induces disaggregation of preformed fibrils in vitro, characterized as a progressive disassembly of fibrils into protofibrils and further clearance of these latter species, leading to a continual inhibition of amyloid neurotoxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Laminin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Peptide Fragments/toxicity , Polymers/metabolism , Amyloid beta-Peptides/ultrastructure , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/cytology , Humans , Kinetics , Mice , Neurofibrils/ultrastructure , Neurons/cytology , Neurotoxins/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...