Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Brain Sci ; 14(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38672009

ABSTRACT

Stress-related mental disorders have become increasingly prevalent, thus endangering mental health worldwide. Exploring stress-associated brain alterations is vital for understanding the possible neurobiological mechanisms underlying these changes. Based on existing evidence, the brain endogenous cannabinoid system (ECS) plays a significant role in the stress response, and disruptions in its function are associated with the neurobiology of various stress-related disorders. This study primarily focuses on investigating the impact of chronic unpredictable stress (CUS) on the expression of hippocampal cannabinoid type 1 (CB1) receptors, part of the ECS, in adult male and female Wistar rats. Additionally, it explores whether environmental enrichment (EE) initiated during adolescence could mitigate the CUS-associated alterations in CB1 expression. Wistar rats, shortly after weaning, were placed in either standard housing (SH) or EE conditions for a duration of 10 weeks. On postnatal day 66, specific subgroups of SH or EE animals underwent a 4-week CUS protocol. Western blot (WB) analysis was conducted in the whole hippocampus of the left brain hemisphere to assess total CB1 protein expression, while immunohistochemistry (IHC) was performed on the right hemisphere to estimate the expression of CB1 receptors in certain hippocampal areas (i.e., CA1, CA3 and dentate gyrus-DG). The WB analysis revealed no statistically significant differences in total CB1 protein levels among the groups; however, reduced CB1 expression was found in specific hippocampal sub-regions using IHC. Specifically, CUS significantly decreased CB1 receptor expression in the CA1 and DG of both sexes, whereas in CA3 the CUS-associated decrease was limited to SH males. Interestingly, EE housing proved protective against these reductions. These findings suggest a region and sex-specific endocannabinoid response to chronic stress, emphasizing the role of positive early experiences in the protection of the adolescent brain against adverse conditions later in life.

2.
Dev Psychobiol ; 65(7): e22422, 2023 11.
Article in English | MEDLINE | ID: mdl-37796476

ABSTRACT

This study aims at investigating whether environmental enrichment (EE) initiated in adolescence can alter chronic unpredictable stress (CUS)-associated changes in astroglial and synaptic plasticity markers in male and female rats. To this end, we studied possible alterations in hippocampal glial fibrillary acidic protein (GFAP) and synaptophysin (SYN) in CUS rats previously housed in EE. Wistar rats on postnatal day (PND) 23 were housed for 10 weeks in standard housing (SH) or enriched conditions. On PND 66, animals were exposed to CUS for 4 weeks. SYN and GFAP expressions were evaluated in CA1 and CA3 subfields and dentate gyrus (DG). CUS reduced the expression of SYN in all hippocampal areas, whereas lower GFAP expression was evident only in CA1 and CA3. The reduced expression of SYN in DG and CA3 was evident to male SH/CUS rats, whereas the reduced GFAP expression in CA1 and CA3 was limited to SH/CUS females. EE housing increased the hippocampal expression of both markers and protected against CUS-associated decreases. Our findings indicate that the decreases in the expression of SYN and GFAP following CUS are region and sex-specific and underline the neuroprotective role of EE against these CUS-associated changes.


Subject(s)
Hippocampus , Rats , Male , Female , Animals , Rats, Wistar , Synaptophysin/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism
3.
Eur J Neurosci ; 58(4): 3003-3025, 2023 08.
Article in English | MEDLINE | ID: mdl-37461295

ABSTRACT

Environmental factors interact with biological and genetic factors influencing the development and well-being of an organism. The interest in better understanding the role of environment on behavior and physiology led to the development of animal models of environmental manipulations. Environmental enrichment (EE), an environmental condition that allows cognitive and sensory stimulation as well as social interaction, improves cognitive function, reduces anxiety and depressive-like behavior and promotes neuroplasticity. In addition, it exerts protection against neurodegenerative disorders, cognitive aging and deficits aggravated by stressful experiences. Given the beneficial effects of EE on the brain and behavior, preclinical studies have focused on its protective role as an alternative, non-invasive manipulation, to help an organism to cope better with stress. A valid, reliable and effective animal model of chronic stress that enhances anxiety and depression-like behavior is the chronic unpredictable mild stress (CUMS). The variety of stressors and the unpredictability in the time and sequence of exposure to prevent habituation, render CUMS an ethologically relevant model. CUMS has been associated with dysregulation of the hypothalamic-pituitary-adrenal axis, elevation in the basal levels of stress hormones, reduction in brain volume, dendritic atrophy and alterations in markers of synaptic plasticity. Although numerous studies have underlined the compensatory role of EE against the negative effects of various chronic stress regimens (e.g. restraint and social isolation), research concerning the interaction between EE and CUMS is sparse. The purpose of the current systematic review is to present up-to-date research findings regarding the protective role of EE against the negative effects of CUMS.


Subject(s)
Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Animals , Depression , Anxiety , Anxiety Disorders , Stress, Psychological/psychology , Disease Models, Animal , Hippocampus
4.
Int J Mol Sci ; 23(24)2022 Dec 19.
Article in English | MEDLINE | ID: mdl-36555816

ABSTRACT

Hypoxic ischemic (HI) brain injury that occurs during neonatal period has been correlated with severe neuronal damage, behavioral deficits and infant mortality. Previous evidence indicates that N-acetylcysteine (NAC), a compound with antioxidant action, exerts a potential neuroprotective effect in various neurological disorders including injury induced by brain ischemia. The aim of the present study was to investigate the role of NAC as a potential therapeutic agent in a rat model of neonatal HI brain injury and explore its long-term behavioral effects. To this end, NAC (50 mg/kg/dose, i.p.) was administered prior to and instantly after HI, in order to evaluate hippocampal and cerebral cortex damage as well as long-term functional outcome. Immunohistochemistry was used to detect inducible nitric oxide synthase (iNOS) expression. The results revealed that NAC significantly alleviated sensorimotor deficits and this effect was maintained up to adulthood. These improvements in functional outcome were associated with a significant decrease in the severity of brain damage. Moreover, NAC decreased the short-term expression of iNOS, a finding implying that iNOS activity may be suppressed and that through this action NAC may exert its therapeutic action against neonatal HI brain injury.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Neuroprotective Agents , Animals , Rats , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Acetylcysteine/metabolism , Animals, Newborn , Rats, Sprague-Dawley , Hypoxia-Ischemia, Brain/metabolism , Brain Injuries/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/metabolism , Brain/metabolism
5.
Behav Processes ; 200: 104707, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35842198

ABSTRACT

Environmental Enrichment (EE) improves cognitive function and enhances brain plasticity, while chronic stress increases emotionality, impairs learning and memory, and has adverse effects on brain anatomy and biochemistry. We explored the beneficial role of environmental enrichment initiated in adolescence against the negative outcomes of Chronic Unpredictable Stress (CUS) during adulthood on emotional behavior, cognitive function, as well as somatic and neuroendocrine markers in both sexes. Adolescent Wistar rats housed either in enriched or standard housing conditions for 10 weeks. On postnatal day 66, a subgroup from each housing condition was daily exposed to a 4-week stress protocol. Following stress, adult rats underwent behavioral testing to evaluate anxiety, exploration/locomotor activity, depressive-like behavior and spatial learning/memory. Upon completion of behavioral testing, animals were exposed to a 10-m stressful event to test the neuroendocrine response to acute stress. CUS decreased body weight gain and increased adrenal weight. Some stress-induced behavioral adverse effects were sex-specific since learning impairments were limited to males while depressive-like behavior to females. EE housing protected against CUS-related behavioral deficits and body weight loss. Exposure to CUS affected the neuroendocrine response of males to acute stress as revealed by the increased corticosterone levels. Our findings highlight the significant role of EE in adolescence as a protective factor against the negative effects of stress and underline the importance of inclusion of both sexes in animal studies.


Subject(s)
Environment , Stress, Psychological , Animals , Anxiety , Corticosterone , Female , Male , Rats , Rats, Wistar , Spatial Learning , Stress, Psychological/psychology
6.
Dev Psychobiol ; 63(2): 173-182, 2021 03.
Article in English | MEDLINE | ID: mdl-32623722

ABSTRACT

This study aims at investigating whether early stress interacts with brain injury due to neonatal hypoxia-ischemia (HI). To this end, we examined possible changes in synaptophysin (SYN) and brain-derived neurotrophic factor (BDNF) expression in the medial prefrontal cortex (mPFC) of maternally separated rats that were subsequently exposed to a HI episode. Rat pups (n = 11) were maternally separated during postnatal days 1 to 6 (3hr/day), while another group was left undisturbed (n = 11). On postnatal day 7, a subgroup (n = 12) from each postnatal manipulation was exposed to HI. Synaptophysin and BDNF expression was estimated in mPFC prelimbic and anterior cingulate subregions of the ipsilateral and contralateral to the occluded common carotid artery hemispheres. Maternally separated rats expressed significantly less BDNF and SYN in both hemispheres. Neonatal HI significantly reduced BDNF and SYN expression in the ipsilateral mPFC only and this reduction was not further altered by early stress. Our findings indicate the enduring negative effect of a short period of maternal separation on the expression of mPFC SYN and BDNF. They, also, reveal that the HI-associated decreases in these markers are limited to the ipsilateral mPFC and are not exacerbated by early stress. These decreases may have important functional implications given the role of prefrontal area in high-order cognition.


Subject(s)
Brain-Derived Neurotrophic Factor , Prefrontal Cortex , Stress, Psychological , Synaptophysin , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Hypoxia , Ischemia , Maternal Deprivation , Prefrontal Cortex/metabolism , Rats , Synaptophysin/metabolism
7.
Int J Dev Neurosci ; 67: 19-32, 2018 06.
Article in English | MEDLINE | ID: mdl-29545098

ABSTRACT

Exposure to environmental enrichment can beneficially influence the behavior and enhance synaptic plasticity. The aim of the present study was to investigate the mediated effects of environmental enrichment on postnatal stress-associated impact with regard to behavior, stress reactivity as well as synaptic plasticity changes in the dorsal hippocampus. Wistar rat pups were submitted to a 3 h maternal separation (MS) protocol during postnatal days 1-21, while another group was left undisturbed. On postnatal day 23, a subgroup from each rearing condition (maternal separation, no-maternal separation) was housed in enriched environmental conditions until postnatal day 65 (6 weeks duration). At approximately three months of age, adult rats underwent behavioral testing to evaluate anxiety (Elevated Plus Maze), locomotion (Open Field Test), spatial learning and memory (Morris Water Maze) as well as non-spatial recognition memory (Novel Object Recognition Test). After completion of behavioral testing, blood samples were taken for evaluation of stress-induced plasma corticosterone using an enzyme-linked immunosorbent assay (ELISA), while immunofluorescence was applied to evaluate hippocampal BDNF and synaptophysin expression in dorsal hippocampus. We found that environmental enrichment protected against the effects of maternal separation as indicated by the lower anxiety levels and the reversal of spatial memory deficits compared to animals housed in standard conditions. These changes were associated with increased BDNF and synaptophysin expression in the hippocampus. Regarding the neuroendocrine response to stress, while exposure to an acute stressor potentiated corticosterone increases in maternally-separated rats, environmental enrichment of these rats prevented this effect. The current study aimed at investigating the compensatory role of enriched environment against the negative outcomes of adverse experiences early in life concurrently on emotional and cognitive behaviors, HPA function and neuroplasticity markers.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Environment , Gene Expression Regulation/physiology , Hippocampus/metabolism , Stress, Psychological/nursing , Synaptophysin/metabolism , Animals , Animals, Newborn , Body Weight/physiology , Corticosterone/blood , Exploratory Behavior/physiology , Female , Male , Maternal Deprivation , Maze Learning/physiology , Rats , Rats, Wistar , Recognition, Psychology , Stress, Psychological/blood , Stress, Psychological/pathology , Stress, Psychological/physiopathology
8.
Brain Res ; 1667: 55-67, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28495306

ABSTRACT

Increasing evidence shows that exposure to an enriched environment (EE) is neuroprotective in adult and neonatal animal models of brain ischemia. However, the mechanisms underlying this effect remain unclear. The aim of the current study was to investigate whether post-weaning EE would be effective in preventing functional deficits and brain damage by affecting markers of synaptic plasticity in a neonatal rat model of hypoxia-ischemia (HI). We also examined the possibility that granulocyte-colony stimulating factor (G-CSF), a growth factor with known neuroprotective effects in a variety of experimental brain injury models, combined with EE stimulation could enhance the potential beneficial effect of EE. Seven-day-old Wistar rats of either sex were subjected to permanent ligation of the left common carotid artery followed by 60min of hypoxia (8% O2) and immediately after weaning (postnatal day 21) were housed in enriched conditions for 4weeks. A group of enriched-housed rats had been treated with G-CSF immediately after HI for 5 consecutive days (50µg/kg/day). Behavioral examination took place approximately at three months of age and included assessments of learning and memory (Morris water maze) as well as motor coordination (Rota-Rod). Infarct size and hippocampal area were estimated following behavioral assessment. Synaptic plasticity was evaluated based on BDNF and synaptophysin expression in the dorsal hippocampus. EE resulted in recovery of post-HI motor deficits and partial improvement of memory impairments which was not accompanied by reduced brain damage. Increased synaptophysin expression was observed in the contralateral to carotid ligation hemisphere. Hypoxia-ischemia alone or followed by enriched conditions did not affect BDNF expression which was increased only in enriched-housed normal rats. The combined therapy of G-CSF and EE further enhanced cognitive function compared to EE provided as monotherapy and prevented HI-induced brain damage by altering synaptic plasticity as reflected by increased synaptophysin expression. The above findings demonstrate that combination of neuroprotective treatments may result in increased protection and it might be a more effective strategy for the treatment of neonatal hypoxic-ischemic brain injury.


Subject(s)
Environment , Granulocyte Colony-Stimulating Factor/pharmacology , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/therapy , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Asphyxia Neonatorum/therapy , Brain-Derived Neurotrophic Factor/metabolism , Combined Modality Therapy , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Housing, Animal , Hypoxia-Ischemia, Brain/pathology , Maze Learning/drug effects , Maze Learning/physiology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Motor Activity/drug effects , Motor Activity/physiology , Random Allocation , Rats, Wistar , Spatial Memory/drug effects , Spatial Memory/physiology , Synaptophysin/metabolism
9.
Int J Dev Neurosci ; 52: 1-12, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27165447

ABSTRACT

Exposure to early-life stress is associated with long-term alterations in brain and behavior, and may aggravate the outcome of neurological insults. This study aimed at investigating the possible interaction between maternal separation, a model of early stress, and subsequent neonatal hypoxia-ischemia on emotional behavior and markers of synaptic plasticity in hippocampus. Therefore, rat pups (N=60) were maternally separated for a prolonged (MS 180min) or a brief (MS 15min) period during the first six postnatal days, while a control group was left undisturbed. Hypoxia-ischemia was applied to a subgroup of each rearing condition on postnatal day 7. Emotional behavior was examined at three months of age and included assessments of anxiety (elevated plus maze), depression-like behavior (forced swimming) and spontaneous exploration (open field). Synaptic plasticity was evaluated based on BDNF and synaptophysin expression in CA3 and dentate gyrus hippocampal regions. We found that neonatal hypoxia-ischemia caused increased levels of anxiety, depression-like behavior and locomotor activity (ambulation). Higher anxiety levels were also seen in maternally separated rats (MS180min) compared to non-maternally separated rats, but prolonged maternal separation prior to HI did not potentiate the HI-associated effect. No differences among the three rearing conditions were found regarding depression-like behavior or ambulation. Immunohistochemical evaluation of synaptophysin revealed that both prolonged maternal separation (MS180min) and neonatal hypoxia-ischemia significantly reduced its expression in the CA3 and dentate gyrus. Decreases in synaptophysin expression in these areas were not exacerbated in rats that were maternally separated for a prolonged period prior to HI. Regarding BDNF expression, we found a significant decrease in immunoreactivity only in the hypoxic-ischemic rats that were subjected to the prolonged maternal separation paradigm. The above findings suggest that early-life stress prior to neonatal hypoxia-ischemia leads to significant alterations in synaptic plasticity of the dorsal hippocampus during adulthood, but does not exacerbate HI-related changes in emotional behavior.


Subject(s)
Hippocampus/physiopathology , Hypoxia/complications , Maternal Deprivation , Neuronal Plasticity/physiology , Stress, Psychological/etiology , Stress, Psychological/pathology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Exploratory Behavior/physiology , Female , Hippocampus/pathology , Male , Maze Learning/physiology , Rats , Rats, Wistar , Swimming/psychology , Synaptophysin/metabolism , Time Factors
10.
Behav Brain Res ; 280: 51-61, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25433094

ABSTRACT

Animal studies suggest that maternal separation, a widely used paradigm to study the effects of early life adversity, exerts a profound and life-long impact on both brain and behavior. The aim of the current study was to investigate whether adverse early life experiences interact with neonatal hypoxia-ischemia, affecting the outcome of this neurological insult at both functional and structural levels during adulthood. Rat pups were separated from their mothers during postnatal days 1-6, for either a short (15 min) or prolonged (180 min) period, while another group was left undisturbed. On postnatal day 7, a subgroup from each of the three postnatal manipulations was exposed to a hypoxic-ischemic episode. Behavioral examination took place approximately at three months of age and included tests of learning and memory (Morris water maze, novel object and novel place recognition), as well as motor coordination (rota-rod). We found that both prolonged maternal separation and neonatal hypoxia-ischemia impaired the animals' spatial learning and reference memory. Deficits in spatial but not visual recognition memory were detected only in hypoxic-ischemic rats. Interestingly, prolonged maternal separation prior to neonatal hypoxia-ischemia augmented the reference memory impairments. Histological analysis of infarct size, hippocampal area and thickness of corpus callosum did not reveal any exacerbation of damage in hypoxic-ischemic rats that were maternally separated for a prolonged period. These are the first data suggesting that an adverse postnatal environmental manipulation of just 6 days causes long-term effects on spatial learning and memory and may render the organism more vulnerable to a subsequent insult.


Subject(s)
Brain/pathology , Brain/physiopathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Maternal Deprivation , Animals , Animals, Newborn , Brain/growth & development , Disease Models, Animal , Female , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/psychology , Male , Memory Disorders/etiology , Memory Disorders/pathology , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Neuropsychological Tests , Psychomotor Performance/physiology , Random Allocation , Rats, Wistar , Recognition, Psychology/physiology , Rotarod Performance Test , Spatial Learning/physiology , Spatial Memory/physiology , Stress, Psychological/pathology , Stress, Psychological/physiopathology , Visual Perception/physiology
11.
Exp Toxicol Pathol ; 66(9-10): 469-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25238695

ABSTRACT

INTRODUCTION: Decalcification of osseous specimens is required for histological analysis; this however may cause tissue damage. In rodent models of allergic rhinitis (AR), epithelial histologic assessment necessitates prior decalcification of the nasal osseous structures. However, respiratory epithelium is highly susceptible to damage, and rat nasal architecture is elaborate and its sectioning is challenging. Nevertheless, decalcification is not standardized in experimental AR. We therefore undertook this task, in order to reduce experimental bias. METHODS: Six-to-eight week-old Wistar rats underwent an AR protocol. Subsequently, nasal structures were decalcified in the following mediums: (i) formic acid 10% for 5 and 20 days; (ii) formic acid 15% for 5 and 15 days; (iii) Morse Solution for 5 and 20 days and (iv) EDTA for 20 and 40 days. Decalcification efficiency/speed was evaluated via radiographic analysis. Furthermore, specimens were stained with hematoxylin and eosin and assessed for preservation of epithelial features. RESULTS: Specimens were appropriately decalcified in 5 days in the formic acid-based mediums and in 20 days in EDTA with minimal epithelial damage. EDTA for 40 days had no unacceptable adverse effects; conversely, 15 and/or 20 days in acid-based agents provided no extra benefit for decalcification and were detrimental to the epithelium. CONCLUSIONS: EDTA treatment for 20 days is appropriate for decalcification of nasal structures in rat models of allergic rhinitis; further incubation preserves epithelial integrity but is not required. When urgency is a factor, formic-acid-based decalcification for 5 days yields acceptable results.


Subject(s)
Decalcification Technique/standards , Rhinitis, Allergic/pathology , Animals , Bone and Bones , Disease Models, Animal , Male , Nose , Rats , Rats, Wistar
12.
Int Forum Allergy Rhinol ; 3(7): 543-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23307410

ABSTRACT

BACKGROUND: The pathophysiologic mechanism of allergy is dependent on the action of many redox-sensitive proinflammatory mediators. However, even though redox disturbances are believed to be a hallmark of inflammation, little is known of the effect of redox imbalance to the pathophysiology of allergic rhinitis. We thus opted to investigate the relation of oxidative stress and allergic rhinitis, through the utilization of a potent antioxidant substance (N-acetylcysteine [NAC]) in a rat model of allergic rhinitis and the evaluation of its action on specific markers of inflammation. METHODS: NAC (50 mg/kg and 250 mg/kg) was intraperitoneally administered to ovalbumin (OVA)-sensitized rats prior to intranasal challenge with OVA. Mucosal congregation of inflammatory cells (eosinophils and mast cells), mucosal expression of redox-sensitive enzymes (inducible nitric oxide synthase [iNOS] and cyclooxygenase 2 [COX-2]), and the blood levels of a key proinflammatory mediator (tumor necrosis factor-α [TNF-α]) were evaluated. RESULTS: Intranasal OVA challenges lead to mucosal inflammation, induction of the mucosal expression of iNOS and COX-2 and elevation of TNF-α blood levels. NAC significantly inhibited accumulation of inflammatory cells and downregulated iNOS expression and TNF-α serum levels. The role of COX-2 appeared to be 2-fold and its expression was divergently modulated by NAC. CONCLUSION: Our findings suggest that redox balance is involved in the pathophysiology of allergic rhinitis in rats and that NAC can potentially suppress the allergen-induced nasal inflammatory cascade. The investigation of the role of oxidative stress in atopy could help in the evaluation of the therapeutic potential of antioxidant substances in allergic diseases.


Subject(s)
Acetylcysteine/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Rhinitis, Allergic, Perennial/drug therapy , Acetylcysteine/pharmacology , Administration, Intranasal , Allergens/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cyclooxygenase 2/immunology , Disease Models, Animal , Male , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Nitric Oxide Synthase Type II/immunology , Ovalbumin/administration & dosage , Rats , Rats, Sprague-Dawley , Rhinitis, Allergic , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/pathology , Tumor Necrosis Factor-alpha/blood
13.
J Neuropathol Exp Neurol ; 71(10): 907-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22964785

ABSTRACT

Inhibition of the myelin-associated neurite outgrowth inhibitor Nogo-A has been found to be beneficial in experimental autoimmune encephalomyelitis (EAE), but there are little data on its expression dynamics during the disease course. We analyzed Nogo-A mRNA and protein during the course of EAE in 27 C57BL/6 mice and in 8 controls. Histopathologic and molecular analyses were performed on Day 0 (naive), preclinical (Day 10), acute (Days 18-22) and chronic (Day 50) time points. In situ hybridization and real-time polymerase chain reaction analyses revealed reduced Nogo-A mRNA expression at preclinical (p < 0.0001) and acute phases (p < 0.0001), followed by upregulation during the chronic phase (p < 0.0001). Nogo-A mRNA was expressed in neurons and oligodendrocytes. By immunohistochemistry and Western blot, there was increased Nogo-A protein expression (p < 0.001) in the chronic phase. Moreover, spatial differences were observed within EAE lesions. The pattern of Nogo-A protein expression inversely correlated with axonal regeneration growth-associated protein 43-positive axons (60% of which were Nogo-A contact-free during the acute phase) and axonal injury (ß-amyloid precursor protein-positive axons). Cortical Nogo-66 receptor protein and mRNA levels increased during the chronic phase. The results indicate that Nogo-A and Nogo receptor are actively regulated in EAE lesions; this may indicate a specific time window for localized axonal regeneration in the acute phase of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , Myelin Proteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Acute Disease , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Mice , Mice, Inbred C57BL , Myelin Proteins/genetics , Neurons/metabolism , Nogo Proteins , Nogo Receptor 1 , Oligodendroglia/metabolism , Receptors, Cell Surface/genetics , Time Factors
14.
Brain Res ; 1425: 98-110, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-22018692

ABSTRACT

Hypoxia-ischemia (HI) induced injury of the neonatal brain accounts for behavioral deficits concerning mainly neurological reflexes, sensorimotor functions and learning/memory disabilities that may evolve throughout development. The positive biological effects of resveratrol, a natural compound with anti-oxidant/anti-inflammatory properties found mainly in red wine have been indicated recently. Aim of this study was to investigate the delayed outcome of early administration of resveratrol in an experimental model of hypoxic-ischemic encephalopathy, by means of behavioral analysis and late neuropathological examination. Seven-day-old (P7) rats were separated into 3 groups: Group 1 underwent HI and treated with resveratrol. Group 2 (HI-treated) was subjected to HI and received same volume of saline. Group 3 (sham-operated) was the control group. A battery of behavioral tests was performed from days P8-P66, during which early reflexes (righting reflex, gait, geotaxis), sensorimotor (rope suspension, beam walking, rotarod) and learning/memory function (passive avoidance, Morris water-maze) were examined. Significant difference among the groups was observed in righting reflex, rotarod and water maze tests in which the resveratrol group almost reached the performance of the control animals. The other behavioral tests showed that control and resveratrol groups were better compared to HI, although not significant. Neuropathology study revealed a remarkable reduction of the infarct and preservation of myelination after resveratrol treatment, which was in most cases correlated with the better performance of the resveratrol group. These findings indicate that long-term neuroprotective effect of resveratrol on neonatal HI-induced gray and white matter damage might be associated with the preservation of behavioral functions.


Subject(s)
Brain Injuries/prevention & control , Hypoxia-Ischemia, Brain/prevention & control , Maze Learning/drug effects , Neuroprotective Agents/therapeutic use , Stilbenes/therapeutic use , Animals , Animals, Newborn , Brain Injuries/pathology , Female , Hypoxia-Ischemia, Brain/pathology , Male , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Reflex, Righting/drug effects , Reflex, Righting/physiology , Resveratrol , Stilbenes/pharmacology
15.
Exp Neurol ; 230(1): 78-89, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440544

ABSTRACT

Autologous bone marrow stromal cells (BMSCs) offer significant practical advantages for potential clinical applications in multiple sclerosis (MS). Based on recent experimental data, a number of clinical trials have been designed for the intravenous (IV) and/or intrathecal (ITH) administration of BMSCs in MS patients. Delivery of BMSCs in the cerebrospinal fluid via intracerebroventricular (ICV) transplantation is a useful tool to identify mechanisms underlying the migration and function of these cells. In the current study, BMSCs were ICV administered in severe and mild EAE, as well as naive animals; neural precursor cells (NPCs) served as cellular controls. Our data indicated that ICV-transplanted BMSCs significantly ameliorated mild though not severe EAE. Moreover, BMSCs exerted significant anti-inflammatory effect on spinal cord with concomitant reduced axonopathy only in the mild EAE model. BMSCs migrated into the brain parenchyma and, depending on their cellular density, within brain parenchyma formed cellular masses characterized by focal inflammation, demyelination, axonal loss and increased collagen-fibronectin deposition. These masses were present in 64% of ICV BMASC-transplanted severe EAE animals whereas neither BMSCs transplanted in mild EAE cases nor the NPCs exhibited similar behavior. BMSCs possibly exerted their fibrogenic effect via both paracrine and autocrine manner, at least partly due to up-regulation of connective tissue growth factor (CTGF) under the trigger of TGFb1. Our findings are of substantial relevance for clinical trials in MS, particularly regarding the possibility that ICV transplanted BMSCs entering the inflamed central nervous system may exhibit - under conditions - a local pathology of yet unknown consequences.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/surgery , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/physiology , Animals , Area Under Curve , Brain/pathology , Cell Survival/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/mortality , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Injections, Intraventricular/adverse effects , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Severity of Illness Index , Spinal Cord/pathology , Transforming Growth Factor beta/metabolism , Transplantation, Autologous/adverse effects , Tumor Necrosis Factor-alpha/metabolism
16.
Neonatology ; 95(1): 33-40, 2009.
Article in English | MEDLINE | ID: mdl-18787335

ABSTRACT

Perinatal asphyxia (PA) is a major determinant for long-term sensorimotor and locomotor deficits. The model of neonatal hypoxia-ischemia (HI) in 7-day-old rats produces sensorimotor cortex, thalamus and striatum injury, which are all critical for the maintenance of sensory motor function. The aim of this study was to evaluate the long-term neurodevelopmental disturbances in the above experimental model and to assess the neuroprotective effect of MgSO(4) in terms of long-term behavioral and morphological changes. Seven-day-old rats were separated into three groups: A (control), neither ligated nor exposed to hypoxia; B (HI/MgSO(4)) ligated, exposed to hypoxia and treated with MgSO(4) (2 g/kg b.w., i.p.), and C (HI) ligated and exposed to hypoxia. At the age of 42 days, the behavior of the rats was evaluated using 5 sensorimotor tests. Muscle power, motor coordination, reflexes, and limb placing were tested to different sensory stimuli. The study was completed with the histopathological evaluation of brain tissue damage. In all individual tests the HI-treated rats performed significantly worse than the control and MgSO(4)-treated rats and this difference was more pronounced in the limb placing tests. Additionally, neonatal HI resulted in extensive neuronal damage that was limited after MgSO(4) administration. Behavioral alterations represent a useful endpoint for studying the consequences of a perinatal HI insult and the efficacy of potential neuroprotective treatments. MgSO(4) administration resulted in prevention of HI-induced sensorimotor deficits and brain injury.


Subject(s)
Gait Disorders, Neurologic/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Magnesium Sulfate/pharmacology , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/prevention & control , Motor Activity/drug effects , Motor Activity/physiology , Movement Disorders/drug therapy , Movement Disorders/physiopathology , Muscle Strength/drug effects , Muscle Strength/physiology , Proprioception/drug effects , Proprioception/physiology , Rats , Rotarod Performance Test
17.
Neuro Endocrinol Lett ; 29(6): 999-1006, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19112389

ABSTRACT

OBJECTIVES: This study is an attempt to determine, the in vivo action of leptin on this hypophysiotropic hypothalamic area, by evaluating the concentrations of luteinizing hormone (LH) and follicular stimulating hormone (FSH) in the serum. METHODS: The experiments were done by stereotaxic injection of recombinant rat leptin (rrleptin) into the third cerebral ventricle (V3) of adult female Wistar rats. Subjects were divided into five groups. Group A included normal intact animals. In Group B, the rats were stereotaxically administered with rrleptin in the V3. The rats, in Groups C, D and E, were subjected to electrolytic lesion of the arcuate nucleus (ARC), of the ventromedial nucleus (VMH) and of both of these hypothalamic nuclei, respectively. Immediately after the electric lesion, they were intracranially injected with rrleptin. Blood samplings for serum LH and FSH levels estimation were performed three times: 1) just before any stereotaxic procedure, 2) six hours, and 3) twenty-four hours after leptin administration. RESULTS: The results showed that serum LH levels increased dramatically in group B, six hours after leptin administration. The LH levels in Groups C, D and E presented the same pattern with a lower peak. The FSH levels were doubled six hours after leptin administration in all groups without any exception. Both LH and FSH serum levels reverted to the initial basic levels after 24 hours. DISCUSSION: The significant conclusion derived from this study is that ARC and VMH, which are responsible for controlling the tonic secretion of gonadotropins, respond in a different way for the FSH and LH secretion. This also suggests that some other mechanism(s) or factor(s) may additionally participate in the control of the tonic component of FSH secretion.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Follicle Stimulating Hormone/blood , Leptin/physiology , Luteinizing Hormone/blood , Ventromedial Hypothalamic Nucleus/metabolism , Analysis of Variance , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Female , Follicle Stimulating Hormone/metabolism , Injections, Intraventricular , Leptin/administration & dosage , Luteinizing Hormone/metabolism , Rats , Rats, Wistar , Statistics, Nonparametric , Third Ventricle , Ventromedial Hypothalamic Nucleus/drug effects
18.
J Neurotrauma ; 25(2): 112-23, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18260794

ABSTRACT

Both heat acclimation (HA) and post-injury treatment with recombinant human erythropoietin (Epo, rhEpo, exogenous Epo) are neuroprotective against traumatic brain injury (TBI). Our previous data demonstrated that HA-induced neuroprotection includes improved functional recovery and reduced cerebral edema formation. Additionally, in earlier Western-blot analyses, we found that HA mice display increased expression of the specific erythropoietin receptor (EpoR) and of hypoxia-inducible factor-1 alpha (HIF-1 alpha), the inducible subunit of the transcription factor, which regulates Epo gene expression, but not of Epo itself. In light of this, the aim of the current study was threefold: (1) to assess Epo expression in the trauma area and hippocampus following HA, rhEpo administration, or combined HA-rhEpo treatment, using immunohistochemical methods that offer enhanced anatomical resolution; (2) to examine the effects of endogenous and exogenous Epo on edema formation in normothermic (NT) mice; and (3) to evaluate the effects of exogenous Epo administration on neuroprotective outcome measures in HA animals. HA induced enhanced expression of endogenous Epo in the trauma area and the hippocampus. Treatment with anti-Epo antibody given to NT mice increased edema formation, whereas rhEpo induced no beneficial effect. Cognitive performance testing and immunohistochemical findings reinforced HA and rhEpo as separate protective interventions but showed no advantage to combining the two strategies. We therefore suggest that HA-induced neuroprotection is shaped by pre-existing mediators but cannot be modified by post-injury treatment aimed at increasing the levels of neuroprotective agents.


Subject(s)
Brain Injuries/drug therapy , Erythropoietin/pharmacology , Erythropoietin/physiology , Neuroprotective Agents , Acclimatization , Animals , Brain Edema/etiology , Brain Edema/prevention & control , Brain Injuries/pathology , Cognition/physiology , Erythropoietin/biosynthesis , Erythropoietin/therapeutic use , Fluoresceins , Fluorescent Dyes , Hippocampus/pathology , Hot Temperature , Humans , Immunohistochemistry , Immunotherapy , Male , Mice , Nerve Degeneration/pathology , Organic Chemicals , Recognition, Psychology/physiology , Recombinant Proteins
19.
Reprod Sci ; 14(7): 667-77, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18000228

ABSTRACT

Previous studies have shown contradictory results regarding magnesium-mediated neuroprotection in animal models of perinatal asphyxia. The aim of this study is to investigate the effects of MgSO(4) postasphyxial treatment on hypoxia-ischemia (HI)-induced brain injury in neonatal rats and the possibility that this effect is related to the severity of brain damage. Seven-day-old rats underwent unilateral carotid artery ligation followed by 1 or 2 hours of hypoxia (8% O(2)) and MgSO(4) administration. Adenosine triphosphate/phosphocreatine and glutamate/glutamine measurements and neuropathological evaluation of the hippocampus were used to assess the effects of HI and MgSO(4). HI caused time-dependent changes in energy stores, amino acid concentrations, and brain damage. Administration of MgSO(4) after 1 hour but not after 2 hours of hypoxia resulted in significant prevention of HI-induced brain injury. MgSO(4) administration results in a significant protection against moderate HI-induced brain damage, whereas it fails to offer a similar effect against severe brain damage.


Subject(s)
Hypoxia, Brain/prevention & control , Hypoxia-Ischemia, Brain/drug therapy , Magnesium Sulfate/pharmacology , Neuroprotective Agents/pharmacology , Adenosine Triphosphate/metabolism , Age Factors , Amino Acids/metabolism , Animals , Animals, Newborn , Female , Hippocampus/pathology , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Male , Phosphocreatine/metabolism , Random Allocation , Rats , Rats, Wistar
20.
Nephrol Dial Transplant ; 21(2): 330-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16221709

ABSTRACT

BACKGROUND: Erythropoietin (EPO), originally identified for its critical role in promoting erythrocyte survival and differentiation, has been shown to exert multiple paracrine/autocrine functions. Protective effects of EPO have been demonstrated in various tissues and experimental models of ischaemia-induced injury. In the present study, we investigated the effect of EPO on an in vivo rat model of renal ischaemia/reperfusion (I/R) injury and the possible mechanisms implicated in the EPO-mediated anti-apoptotic action. METHODS: Male Wistar rats, subjected to renal ischaemia for 45 min, were administered either saline or EPO (500 U/kg, i.p.) 20 min prior to I/R. A sham-operated group served as the control. At 48 h of reperfusion, the renal dysfunction and injury was assessed by measurement of serum biochemical markers (urea, creatinine) and histological grading. Apoptosis was assessed by the TUNEL method and morphological criteria. Expression of Bax and NF-kappaB (p65) was also evaluated. RESULTS: High levels of serum urea and creatinine were identified at 48 h after ischaemia. The EPO-treated group had significantly lower serum and creatinine levels. Semi-quantitative assessment of the histological lesions showed that rats subjected to I/R developed marked structural damage, whereas significantly less tubular damage was observed in the EPO-treated group. I/R caused an increase in TUNEL-positive cells that was accompanied by morphological evidence of apoptosis. In the EPO-treated rats only a few scattered TUNEL-positive cells were observed. Up-regulation of Bax in the tubular epithelial cells and increased expression of NF-kappaB was observed in the I/R-treated rats, while diminished expression of Bax and positive immunostaining of NF-kappaB was observed in the EPO-treated rats. CONCLUSION: Administration of EPO as a single dose before the onset of ischaemia produced a significant reduction in tubular injury, which was accompanied by a marked amelioration of renal functional impairment. The cytoprotective action of EPO against I/R injury seems to be associated with its anti-apoptotic action. Moreover, transcription factor NF-kappaB is likely to play a pivotal role in the pathophysiology of I/R renal injury and might have a key role in EPO-mediated protective effects.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Disease Models, Animal , Erythropoietin/therapeutic use , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Acute Kidney Injury/pathology , Animals , Apoptosis , Male , NF-kappa B/physiology , Rats , Rats, Wistar , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...