Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 21(1): 31, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38263116

ABSTRACT

BACKGROUND AND PURPOSE: The only validated treatment to prevent brain damage associated with hypoxia-ischemia (HI) encephalopathy of the newborn is controlled hypothermia with limited benefits. Additional putative neuroprotective drug candidates include sildenafil citrate, a phosphodiesterase-type 5 inhibitor. The main objective of this preclinical study is to assess its ability to reduce HI-induced neuroinflammation, in particular through its potential effect on microglial activation. METHODS: HI was induced in P10 Sprague-Dawley rats by unilateral carotid permanent artery occlusion and hypoxia (HI) and treated by either hypothermia (HT) alone, Sildenafil (Sild) alone or combined treatment (SildHT). Lesion size and glial activation were analyzed by immunohistochemistry, qRT-PCR, and proteomic analyses performed at P13. RESULTS: None of the treatments was associated with a significant early reduction in lesion size 72h after HI, despite significant changes in tissue loss distribution. Significant reductions in both Iba1 + (within the ipsilateral hemisphere) and GFAP + cells (within the ipsilateral hippocampus) were observed in SildHT group, but not in the other treatment groups. In microglia-sorted cells, pro-inflammatory markers, i.e. Il1b, Il6, Nos2, and CD86 were significantly downregulated in SildHT treatment group only. These changes were restricted to the ipsilateral hemisphere, were not evidenced in sorted astrocytes, and were not sex dependent. Proteomic analyses in sorted microglia refined the pro-inflammatory effect of HI and confirmed a biologically relevant impact of SildHT on specific molecular pathways including genes related to neutrophilic functions. CONCLUSIONS: Our findings suggest that Sildenafil combined with controlled hypothermia produces maximum effect in mitigating microglial activation induced by HI through complex proteomic regulation. The reduction of neuroinflammation induced by Sildenafil may represent an interesting therapeutic strategy for neonatal neuroprotection.


Subject(s)
Hypothermia , Hypoxia-Ischemia, Brain , Rats , Animals , Sildenafil Citrate , Microglia , Rats, Sprague-Dawley , Neuroinflammatory Diseases , Proteomics , Ischemia , Hypoxia
2.
Neurotox Res ; 41(6): 526-545, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37378827

ABSTRACT

Neonatal hypoxia-ischemia (HI) is one of the main causes of tissue damage, cell death, and imbalance between neuronal excitation and inhibition and synaptic loss in newborns. GABA, the major inhibitory neurotransmitter of the central nervous system (CNS) in adults, is excitatory at the onset of neurodevelopment and its action depends on the chloride (Cl-) cotransporters NKCC1 (imports Cl-) and KCC2 (exports Cl-) expression. Under basal conditions, the NKCC1/KCC2 ratio decreases over neurodevelopment. Thus, changes in this ratio caused by HI may be related to neurological disorders. The present study evaluated the effects of bumetanide (NKCC cotransporters inhibitor) on HI impairments in two neurodevelopmental periods. Male Wistar rat pups, 3 (PND3) and 11 (PND11) days old, were submitted to the Rice-Vannucci model. Animals were divided into 3 groups: SHAM, HI-SAL, and HI-BUM, considering each age. Bumetanide was administered intraperitoneally at 1, 24, 48, and 72 h after HI. NKCC1, KCC2, PSD-95, and synaptophysin proteins were analyzed after the last injection by western blot. Negative geotaxis, righting reflex, open field, object recognition test, and Morris water maze task were performed to assess neurological reflexes, locomotion, and memory function. Tissue atrophy and cell death were evaluated by histology. Bumetanide prevented neurodevelopmental delay, hyperactivity, and declarative and spatial memory deficits. Furthermore, bumetanide reversed HI-induced brain tissue damage, reduced neuronal death and controlled GABAergic tone, maintained the NKCC1/KCC2 ratio, and synaptogenesis close to normality. Thereby, bumetanide appears to play an important therapeutic role in the CNS, protecting the animals against HI damage and improving functional performance.


Subject(s)
Bumetanide , Hypoxia-Ischemia, Brain , Rats , Animals , Male , Bumetanide/pharmacology , Bumetanide/therapeutic use , Rats, Wistar , Solute Carrier Family 12, Member 2/metabolism , Ischemia/drug therapy , Hypoxia/drug therapy , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/drug therapy , Brain/metabolism , Cognition , Animals, Newborn
3.
Behav Brain Res ; 430: 113935, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35605797

ABSTRACT

Chronic cerebral hypoperfusion leads to neuronal loss in the hippocampus and spatial memory impairments. Physical exercise is known to prevent cognitive deficits in animal models; and there is evidence of sex differences in behavioral neuroprotective approaches. The aim of present study was to investigate the effects of acrobatic training in male and female rats submitted to chronic cerebral hypoperfusion. Males and females rats underwent 2VO (two-vessel occlusion) surgery and were randomly allocated into 4 groups of males and 4 groups of females, as follows: 2VO acrobatic, 2VO sedentary, Sham acrobatic and Sham sedentary. The acrobatic training started 45 days after surgery and lasted 4 weeks; animals were then submitted to object recognition and water maze testing. Brain samples were collected for histological and morphological assessment and flow cytometry. 2VO causes cognitive impairments and acrobatic training prevented spatial memory deficits assessed in the water maze, mainly for females. Morphological analysis showed that 2VO animals had less NeuN labeling and acrobatic training prevented it. Increased number of GFAP positive cells was observerd in females; moreover, males had more branched astrocytes and acrobatic training prevented the branching after 2VO. Flow cytometry showed higher mitochondrial potential in trained animals and more reactive oxygen species production in males. Acrobatic training promoted neuronal survival and improved mitochondrial function in both sexes, and influenced the glial scar in a sex-dependent manner, associated to greater cognitive benefit to females after chronic cerebral hypoperfusion.


Subject(s)
Brain Ischemia , Spatial Memory , Animals , Female , Male , Rats , Astrocytes/pathology , Brain Ischemia/pathology , Cicatrix/pathology , Disease Models, Animal , Hippocampus , Maze Learning , Spatial Memory/physiology
4.
Brain Res ; 1756: 147334, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33539794

ABSTRACT

Acute seizures may cause permanent brain damage depending on the severity. The pilocarpine animal model has been broadly used to study the acute effects of seizures on neurogenesis and plasticity processes and the resulting epileptogenesis. Likewise, zebrafish is a good model to study neurogenesis and plasticity processes even in adulthood. Thus, the aim of this study is to evaluate the effects of pilocarpine-induced acute seizures-like behavior on neuroplasticity and long-term behavior in adult zebrafish. To address this issue, adult zebrafish were injected with Pilocarpine (350 mg/Kg, i.p; PILO group) or Saline (control group). Experiments were performed at 1, 2, 3, 10 or 30 days after injection. We evaluated behavior using the Light/Dark preference, Open Tank and aggressiveness tests. Flow cytometry and BrdU were carried out to detect changes in cell death and proliferation, while Western blotting was used to verify different proliferative, synaptic and neural markers in the adult zebrafish telencephalon. We identified an increased aggressive behavior and increase in cell death in the PILO group, with increased levels of cleaved caspase 3 and PARP1 1 day after seizure-like behavior induction. In addition, there were decreased levels of PSD95 and SNAP25 and increased BrdU positive cells 3 days after seizure-like behavior induction. Although most synaptic and cell death markers levels seemed normal by 30 days after seizures-like behavior, persistent aggressive and anxiolytic-like behaviors were still detected as long-term effects. These findings might indicate that acute severe seizures induce short-term biochemical alterations that ultimately reflects in a long-term altered phenotype.


Subject(s)
Behavior, Animal/drug effects , Cell Proliferation/drug effects , Neuronal Plasticity/physiology , Pilocarpine/pharmacology , Seizures/drug therapy , Animals , Cell Proliferation/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Neurogenesis/physiology , Neuronal Plasticity/drug effects , Seizures/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Time , Zebrafish
5.
Mol Neurobiol ; 58(5): 2297-2308, 2021 May.
Article in English | MEDLINE | ID: mdl-33417220

ABSTRACT

Neonatal hypoxia-ischemia (HI) is among the main causes of mortality and morbidity in newborns. Experimental studies show that the immature rat brain is less susceptible to HI injury, suggesting that changes that occur during the first days of life drastically alter its susceptibility. Among the main developmental changes observed is the mitochondrial function, namely, the tricarboxylic acid (TCA) cycle and respiratory complex (RC) activities. Therefore, in the present study, we investigated the influence of neonatal HI on mitochondrial functions, redox homeostasis, and cell damage at different postnatal ages in the hippocampus of neonate rats. For this purpose, animals were divided into four groups: sham postnatal day 3 (ShP3), HIP3, ShP11, and HIP11. We initially observed increased apoptosis in the HIP11 group only, indicating a higher susceptibility of these animals to brain injury. Mitochondrial damage, as determined by flow cytometry showing mitochondrial swelling and loss of mitochondrial membrane potential, was also demonstrated only in the HIP11 group. This was consistent with the decreased mitochondrial oxygen consumption, reduced TCA cycle enzymes, and RC activities and induction of oxidative stress in this group of animals. Considering that HIP3 and the sham animals showed no alteration of mitochondrial functions, redox homeostasis, and showed no apoptosis, our data suggest an age-dependent vulnerability of the hippocampus to hypoxia-ischemia. The present results highlight age-dependent metabolic differences in the brain of neonate rats submitted to HI indicating that different treatments might be needed for HI newborns with different gestational ages.


Subject(s)
Apoptosis/physiology , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Age Factors , Animals , Disease Models, Animal , Female , Homeostasis/physiology , Oxidation-Reduction , Oxygen Consumption/physiology , Rats , Rats, Wistar
6.
J Neurochem ; 157(6): 1911-1929, 2021 06.
Article in English | MEDLINE | ID: mdl-33098090

ABSTRACT

Prenatal and early postnatal periods are important for brain development and neural function. Neonatal insults such as hypoxia-ischemia (HI) causes prolonged neural and metabolic dysregulation, affecting central nervous system maturation. There is evidence that brain hypometabolism could increase the risk of adult-onset neurodegenerative diseases. However, the impact of non-pharmacologic strategies to attenuate HI-induced brain glucose dysfunction is still underexplored. This study investigated the long-term effects of early environmental enrichment in metabolic, cell, and functional responses after neonatal HI. Thereby, male Wistar rats were divided according to surgical procedure, sham, and HI (performed at postnatal day 3), and the allocation to standard (SC) or enriched condition (EC) during gestation and lactation periods. In-vivo cerebral metabolism was assessed by means of [18 F]-FDG micro-positron emission tomography, and cognitive, biochemical, and histological analyses were performed in adulthood. Our findings reveal that HI causes a reduction in glucose metabolism and glucose transporter levels as well as hyposynchronicity in metabolic brain networks. However, EC during prenatal or early postnatal period attenuated these metabolic disturbances. A positive correlation was observed between [18 F]-FDG values and volume ratios in adulthood, indicating that preserved tissue by EC is metabolically active. EC promotes better cognitive scores, as well as down-regulation of amyloid precursor protein in the parietal cortex and hippocampus of HI animals. Furthermore, growth-associated protein 43 was up-regulated in the cortex of EC animals. Altogether, results presented support that EC during gestation and lactation period can reduce HI-induced impairments that may contribute to functional decline and progressive late neurodegeneration.


Subject(s)
Brain/metabolism , Environment , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , Neuronal Plasticity/physiology , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn , Female , Hypoxia-Ischemia, Brain/psychology , Lactation/metabolism , Lactation/psychology , Male , Maze Learning/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Neurodegenerative Diseases/psychology , Positron-Emission Tomography/methods , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Wistar
7.
Neurobiol Learn Mem ; 171: 107207, 2020 05.
Article in English | MEDLINE | ID: mdl-32147586

ABSTRACT

BACKGROUND AND PURPOSE: Hypoxia and cerebral ischemia (HI) events are capable of triggering important changes in brain metabolism, including glucose metabolism abnormalities, which may be related to the severity of the insult. Using positron emission microtomography (microPET) with [18F]fluorodeoxyglucose (18F-FDG), this study proposes to assess abnormalities of brain glucose metabolism in adult rats previously submitted to the neonatal HI model. We hypothesize that cerebral metabolic outcomes will be associated with cognitive deficits and magnitude of brain injury. METHODS: Seven-day-old rats were subjected to an HI model, induced by permanent occlusion of the right common carotid artery and systemic hypoxia. 18F-FDG-microPET was used to assess regional and whole brain glucose metabolism in rats at 60 postnatal days (PND 60). An interregional cross-correlation matrix was utilized to construct metabolic brain networks (MBN). Rats were also subjected to the Morris Water Maze (MWM) to evaluate spatial memory and their brains were processed for volumetric evaluation. RESULTS: Brain glucose metabolism changes were observed in adult rats after neonatal HI insult, limited to the right brain hemisphere. However, not all HI animals exhibited significant cerebral hypometabolism. Hippocampal glucose metabolism was used to stratify HI animals into HI hypometabolic (HI-h) and HI non-hypometabolic (HI non-h) groups. The HI-h group had drastic MBN disturbance, cognitive deficit, and brain tissue loss, concomitantly. Conversely, HI non-h rats had normal brain glucose metabolism and brain tissue preserved, but also presented MBN changes and spatial memory impairment. Furthermore, data showed that brain glucose metabolism correlated with cognitive deficits and brain volume outcomes. CONCLUSIONS: Our findings demonstrated that long-term changes in MBN drive memory impairments in adult rats subjected to neonatal hypoxic ischemia, using in vivo imaging microPET-FDG. The MBN analyses identified glucose metabolism abnormalities in HI non-h animals, which were not detected by conventional 18F-FDG standardized uptake value (SUVr) measurements. These animals exhibited a metabolic brain signature that may explain the cognitive deficit even with no identifiable brain damage.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Memory Disorders/metabolism , Nerve Net/metabolism , Animals , Brain/diagnostic imaging , Disease Models, Animal , Glucose/metabolism , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/diagnostic imaging , Male , Memory Disorders/diagnostic imaging , Memory Disorders/etiology , Nerve Net/diagnostic imaging , Positron-Emission Tomography , Rats , Rats, Wistar
8.
Int J Dev Neurosci ; 79: 86-95, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31693927

ABSTRACT

INTRODUCTION: Neonatal Hypoxia-Ischemia (HI) is a major cause of morbidity and mortality, and is frequently associated with short and long-term neurologic and cognitive impairments. The HI injury causes mitochondrial damage leading to increased production of reactive oxygen species (ROS). Phytoestrogens are non-steroidal plant substances structurally and functionally similar to estrogen. Coumestrol is a potent isoflavonoid with a protective effect against ischemic brain damage in adult rats. Our aim was to determine if coumestrol treatment following neonatal HI attenuates the long-term cognitive deficits induced by neonatal HI, as well as to investigate one possible mechanism underlying its potential effect. METHODS: On the 7th postnatal day, male Wistar rats were submitted to the Levine-Rice HI model. Intraperitoneal injections of 20 mg/kg of coumestrol, or vehicle, were administered immediately pre-hypoxia or 3 h post-hypoxia. At 12 h after HI the mitochondrial status and ROS levels were determined. At 60th postnatal day the cognitive deficits were revealed in the Morris water maze reference and working spatial memories. Following behavioral analysis, histological assessment was performed and reactive astrogliosis was measured by GFAP expression. RESULTS: Results demonstrate that both pre- and post-HI administration of coumestrol were able to counteract the long-term cognitive and morphological impairments caused by HI, as well as to block the late reactive astrogliosis. The pre-HI administration of coumestrol was able to prevent the early mitochondrial dysfunction in the hippocampus of injured rat pups. CONCLUSION: Present data suggest that coumestrol exerts protection against experimental neonatal brain hypoxia-ischemia through, at least in part, early modulation of mitochondrial function.


Subject(s)
Brain/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Coumestrol/pharmacology , Hypoxia-Ischemia, Brain/drug therapy , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Cognitive Dysfunction/metabolism , Coumestrol/therapeutic use , Hypoxia-Ischemia, Brain/metabolism , Male , Maze Learning/drug effects , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Phytoestrogens/pharmacology , Phytoestrogens/therapeutic use , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
9.
Mol Neurobiol ; 56(1): 761-762, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29909452

ABSTRACT

The authors hereby declare that the Figure 4 in page eight of the paper "Stem cells from human exfoliated deciduous teeth modulate early astrocyte response after spinal cord contusion" authored by Fabrício Nicola and colleagues (DOI: 10.1007/s12035-018-1127-4) was mistakenly included.

10.
Mol Neurobiol ; 56(1): 748-760, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29796991

ABSTRACT

The transplantation of stem cells from human exfoliated deciduous teeth (SHED) has been studied as a possible treatment strategy for spinal cord injuries (SCIs) due to its potential for promoting tissue protection and functional recovery. The aim of the present study was to investigate the effects of the early transplantation of SHED on glial scar formation and astrocytic reaction after an experimental model of SCI. Wistar rats were spinalized using the NYU Impactor. Animals were randomly distributed into three groups: control (naive) (animal with no manipulation); SCI (receiving laminectomy followed by SCI and treated with vehicle), and SHED (SCI rat treated with intraspinal SHED transplantation, 1 h after SCI). In vitro investigation demonstrated that SHED were able to express mesenchymal stem cells, vimentin and S100B markers, related with neural progenitor and glial cells, respectively. The acute SHED transplantation promoted functional recovery, measured as from the first week after spinal cord contusion by Basso, Beattie, and Bresnahan scale. Twenty-four and 48 h after lesion, flow cytometry revealed a spinal cord vimentin+ cells increment in the SHED group. The increase of vimentin+ cells was confirmed by immunofluorescence. Moreover, the bioavailability of astrocytic proteins such as S100B and Kir4.1 shown to be increased in the spinal cord of SHED group, whereas there was a glial scar reduction, as indicated by ELISA and Western blot techniques. The presented results support that SHED act as a neuroprotector agent after transplantation, probably through paracrine signaling to reduce glial scar formation, inducing tissue plasticity and functional recovery.


Subject(s)
Astrocytes/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Tooth Exfoliation/pathology , Tooth, Deciduous/cytology , Animals , Aquaporin 4/metabolism , Astrocytes/metabolism , Cells, Cultured , Glial Fibrillary Acidic Protein/metabolism , Hindlimb/physiopathology , Humans , Male , Potassium Channels, Inwardly Rectifying/metabolism , Rats, Wistar , S100 Calcium Binding Protein beta Subunit/metabolism , Spinal Cord Injuries/metabolism , Vimentin/metabolism
11.
Neurochem Res ; 43(12): 2268-2276, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30255215

ABSTRACT

Neonatal hypoxia-ischemia (HI) is associated to cognitive and motor impairments and until the moment there is no proven treatment. The underlying neuroprotective mechanisms of stem cells are partially understood and include decrease in excitotoxicity, apoptosis and inflammation suppression. This study was conducted in order to test the effects of intracardiac transplantation of human dental pulp stem cells (hDPSCs) for treating HI damage. Seven-day-old Wistar rats were divided into four groups: sham-saline, sham-hDPSCs, HI-saline, and HI-hDPSCs. Motor and cognitive tasks were performed from postnatal day 30. HI-induced cognitive deficits in the novel-object recognition test and in spatial reference memory impairment which were prevented by hDPSCs. No motor impairments were observed in HI animals. Immunofluorescence analysis showed human-positive nuclei in hDPSC-treated animals closely associated with anti-GFAP staining in the lesion scar tissue, suggesting that these cells were able to migrate to the injury site and could be providing support to CNS cells. Our study evidence novel evidence that hDPSC can contribute to the recovery following hypoxia-ischemia and highlight the need of further investigation in order to better understand the exact mechanisms underlying its neuroprotective effects.


Subject(s)
Cognitive Dysfunction/prevention & control , Dental Pulp/transplantation , Hypoxia-Ischemia, Brain/therapy , Stem Cell Transplantation/methods , Animals , Animals, Newborn , Cells, Cultured , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Dental Pulp/cytology , Dental Pulp/physiology , Female , Heart Ventricles , Humans , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/pathology , Injections , Male , Maze Learning/physiology , Pregnancy , Random Allocation , Rats , Rats, Wistar , Stem Cells/physiology
12.
J Perinat Med ; 46(4): 433-439, 2018 May 24.
Article in English | MEDLINE | ID: mdl-28841577

ABSTRACT

Neonatal hypoxia ischemia (HI) is the main cause of mortality and morbidity in newborns. The mechanisms involved in its progression start immediately and persist for several days. Oxidative stress and inflammation are determinant factors of the severity of the final lesion. The spleen plays a major part in the inflammatory response to HI. This study assessed the temporal progression of HI-induced alterations in oxidative stress parameters in the hippocampus, the most affected brain structure, and in the spleen. HI was induced in Wistar rat pups in post-natal day 7. Production of reactive oxygen species (ROS), and the activity of the anti oxidant enzyme superoxide dismutase and catalase were assessed 24 h, 96 h and 38 days post-HI. Interestingly, both structures showed a similar pattern, with few alterations in the production of ROS species up to 96 h often combined with an increased activity of the anti oxidant enzymes. However, 38 days after the injury, ROS were at the highest in both structures, coupled with a decrease in the activity of the enzymes. Altogether, present results suggest that HI causes long lasting alterations in the hippocampus as well as in the spleen, suggesting a possible target for delayed treatments for HI.


Subject(s)
Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Oxidative Stress , Spleen/metabolism , Animals , Animals, Newborn , Catalase/metabolism , Disease Models, Animal , Female , Hippocampus/pathology , Hypoxia-Ischemia, Brain/pathology , Rats, Wistar , Reactive Oxygen Species/metabolism , Spleen/pathology , Superoxide Dismutase/metabolism
13.
Brain Res ; 1663: 95-105, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28322752

ABSTRACT

Stem cells from human exfoliated deciduous teeth (SHED) transplants have been investigated as a possible treatment strategy for spinal cord injuries (SCI) due to their potential for promoting functional recovery. The aim of present study was to investigate the effects of SHED on neuronal death after an experimental model of SCI. METHODS: Wistar rats were spinalized using NYU impactor®. Animals were randomly distributed into 4 groups: Control (Naive) or Surgical control, Sham (laminectomy with no SCI); SCI (laminectomy followed by SCI, treated with vehicle); SHED (SCI treated with intraspinal transplantation of 3×105 SHED, 1h after SCI). Functional evaluations and morphological analysis were performed to confirm the spinal injury and the benefit of SHED transplantation on behavior, tissue protection and motor neuron survival. Flow cytometry of neurons, astrocytes, macrophages/microglia and T cells of spinal cord tissue were run at six, twenty-four, forty-eight and seventy-two hours after lesion. Six hours after SCI, ELISA and Western Blot were run to assess pro- and anti-apoptotic factors. The SHED group showed a significant functional improvement in comparison to the SCI animals, as from the first week until the end of the experiment. This behavioral protection was associated with less tissue impairment and greater motor neuron preservation. SHED reduced neuronal loss over time, as well as the overexpression of pro-apoptotic factor TNF-α, while maintained basal levels of the anti-apoptotic BCL-XL six hours after lesion. Data here presented show that SHED transplantation one hour after SCI interferes with the balance between pro- and anti-apoptotic factors and reduces early neuronal apoptosis, what contributes to tissue and motor neuron preservation and hind limbs functional recovery.


Subject(s)
Adult Stem Cells/transplantation , Spinal Cord Injuries/therapy , Tooth, Deciduous/transplantation , Adult Stem Cells/pathology , Animals , Apoptosis , Astrocytes/pathology , Cell Survival , Cells, Cultured , Humans , Male , Neurons/physiology , Neuroprotective Agents/metabolism , Rats , Rats, Wistar , Recovery of Function/physiology , Spinal Cord/pathology , Spinal Cord Injuries/physiopathology , Stem Cells/pathology , Tooth, Deciduous/metabolism
14.
Neurochem Res ; 42(5): 1422-1429, 2017 May.
Article in English | MEDLINE | ID: mdl-28210957

ABSTRACT

Regular physical activity has shown to improve the quality of life and to prevent age-related memory deficits. Memory processing requires proper regulation of several enzymes such as sodium-potassium adenosine triphosphatase (Na+, K+-ATPase) and acetylcholinesterase (AChE), which have a pivotal role in neuronal transmission. The present study investigated the effects of a treadmill running protocol in young (3 months), mature (6 months) and aged (22 months) Wistar rats, on: (a) cognitive function, as assessed in the Water maze spatial tasks; (b) Na+, K+-ATPase and AChE activities in the hippocampus following cognitive training alone or treadmill running combined with cognitive training. Animals of all ages were assigned to naïve (with no behavioral or exercise training), sedentary (non-exercised, with cognitive training) and exercised (20 min of daily running sessions, 3 times per week for 4 weeks and with cognitive training) groups. Cognition was assessed by reference and working memory tasks run in the Morris Water maze; 24 h after last session of behavioral testing, hippocampi were collected for biochemical analysis. Results demonstrated that: (a) a moderate treadmill running exercise prevented spatial learning and memory deficits in aged rats; (b) training in the Water maze increased both Na+, K+-ATPase and AChE activities in the hippocampus of mature and aged rats; (c) aged exercised rats displayed an even further increase of Na+, K+-ATPase activity in the hippocampus, (d) enzyme activity correlated with memory performance in aged rats. It is suggested that exercise prevents spatial memory deficits in aged rats probably through the activation of Na+, K+-ATPase in the hippocampus.


Subject(s)
Aging/metabolism , Hippocampus/enzymology , Memory Disorders/enzymology , Physical Conditioning, Animal/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Spatial Memory/physiology , Animals , Enzyme Activation/physiology , Exercise Test/methods , Exercise Test/psychology , Male , Maze Learning/physiology , Memory Disorders/prevention & control , Physical Conditioning, Animal/methods , Physical Conditioning, Animal/psychology , Random Allocation , Rats , Rats, Wistar
15.
J Neurosci Res ; 95(1-2): 409-421, 2017 01 02.
Article in English | MEDLINE | ID: mdl-27870406

ABSTRACT

Neonatal hypoxia-ischemia (HI) is an important cause of neurological deficits in humans, and the Levine-Rice model of experimental HI in the rat mimics the human brain lesion and the following sensory motor deficits and cognitive disabilities. With the growing evidence that sex influences all levels of brain functions, this Mini-Review highlights studies in which sex was a controlled variable and that provided evidence of sexual dimorphism in behavioral outcome, extension of brain damage, mechanisms of lesion, and treatment efficacy in the rat neonatal HI model. It was shown that 1) females have greater memory deficits; 2) cell death is dependent mainly on caspase activation in females; 3) males are more susceptible to oxidative stress; and 4) treatments acting on distinct cell death pathways afford sex-dependent neuroprotection. These tentative conclusions, along with growing evidence from other fields of neurobiology, support the need for scientists to design their experiments considering sex as an important variable; otherwise, important knowledge will continue to be missed. It is conceivable that sex can influence the development of efficacious therapeutic tools to treat neonates suffering from brain HI. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/pathology , Developmental Disabilities/etiology , Hypoxia-Ischemia, Brain/complications , Sex Characteristics , Animals , Animals, Newborn , Disease Models, Animal , Female , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...