Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Sci Rep ; 8(1): 16584, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30410110

ABSTRACT

The tetanus toxin C-fragment is a non-toxic peptide that can be transported from peripheral axons into spinal motoneurons. In in vitro experiments it has been shown that this peptide activates signaling pathways associated with Trk receptors, leading to cellular survival. Because motoneuron degeneration is the main pathological hallmark in motoneuron diseases, and excitotoxicity is an important mechanism of neuronal death in this type of disorders, in this work we tested whether the tetanus toxin C-fragment is able to protect MN in the spinal cord in vivo. For this purpose, we administered the peptide to rats subjected to excitotoxic motoneuron degeneration induced by the chronic infusion of AMPA in the rat lumbar spinal cord, a well-established model developed in our laboratory. Because the intraspinal infusion of the fragment was only weakly effective, whereas the i.m. administration was remarkably neuroprotective, and because the i.m. injection of an inhibitor of Trk receptors diminished the protection, we conclude that such effects require a retrograde signaling from the neuromuscular junction to the spinal motoneurons. The protection after a simple peripheral route of administration of the fragment suggests a potential therapeutic use of this peptide to target spinal MNs exposed to excitotoxic conditions in vivo.


Subject(s)
Motor Neuron Disease/prevention & control , Nerve Degeneration/prevention & control , Peptide Fragments/administration & dosage , Spinal Cord/pathology , Tetanus Toxin/administration & dosage , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/adverse effects , Animals , Disease Models, Animal , Injections, Intramuscular , Injections, Spinal , Male , Motor Neuron Disease/chemically induced , Motor Neuron Disease/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Peptide Fragments/pharmacology , Phosphorylation , Rats , Receptor, trkA/metabolism , Spinal Cord/metabolism , Tetanus Toxin/pharmacology
2.
Int. j. morphol ; 36(2): 584-591, jun. 2018. tab, graf
Article in English | LILACS | ID: biblio-954158

ABSTRACT

Among the neurodegenerative disorders, Parkinson disease (PD) is ranked as second most common. The pathological hallmark is selective degeneration of the dopaminergic neurons in the nigro-striatal regions of brain with appearance of the Lewy bodies. Present study explores the neuro-protective potential of polydatin in terms of amelioration of degeneration of dopaminergic neurons in nigro-striatal regions of brain and distorted neuromotor behavior in the rotenone model of Parkinson's disease. Thirty-six male Sprague Dawley rats were divided into three groups. Group A (control), Group B (rotenone treated) and Group C (rotenone+polydatin treated). Rotenone was administrated intraperitoneally (i.p) at a dose of 3 mg/kg/body weight while polydatin was given i.p. at a dose of 50 mg/ kg/body weight for four weeks. Then, animals were sacrificed; substantia nigra (SN) & striatum isolated from brain and five micron thick sections were prepared. Cresyl violet (CV), H&E and Immuno-histochemical staining using anti-TH antibody was done. Motor behavior was assessed weekly throughout the experiment using five different methods. Rotenone treated parkinsonian animals showed deterioration of motor behavior, weight loss, loss of dopaminergic neurons and diminished immune-reactivity in the sections from the nigrostriatal regions of these animals Polydatin+rotenone treatment showed contradicting effects to parkinsonism, with amelioration in weight loss, neuro-motor behavior, dopaminergic loss and immune-reactivity against dopaminergic neurons. Present study revealed a neuro-protective potential of polydatin in animal model of PD by ameliorating the neuro-motor abnormalities and degeneration of dopaminergic neurons in nigrostriatal regions.


Entre los trastornos neurodegenerativos, la enfermedad de Parkinson (EP) se clasifica como la segunda más común. El sello patológico es la degeneración selectiva de las neuronas dopaminérgicas en las regiones nigro-estriatales del cerebro, con la aparición de los cuerpos de Lewy. El presente estudio explora el potencial de protección neuronal de la polidatina en términos de la mejora de la degeneración de las neuronas dopaminérgicas en las regiones nigro-estriatales del cerebro y el comportamiento neuromotor distorsionado en el modelo de rotenona de la enfermedad de Parkinson. Treinta y seis ratas macho Sprague Dawley se dividieron en tres grupos: Grupo A (control), Grupo B (tratado con rotenona) y Grupo C (tratamiento con rotenona + polidatina). La rotenona se administró por vía intraperitoneal (i.p.) a una dosis de 3 mg/kg/peso corporal, mientras que la polidatina se administró i.p. a una dosis de 50 mg/kg/ peso corporal durante cuatro semanas. Posteriormente, los animales fueron sacrificados. Se aislaron la substantia nigra (SN) y cuerpo estriado de los cerebros y se realizaron secciones de cinco micras de espesor. Se realizó una tinción de violeta de cresilo (CV), H&E y tinción inmunohistoquímica usando anticuerpo anti-TH. El comportamiento motriz se evaluó semanalmente durante todo el experimento utilizando cinco métodos diferentes. Los animales parkinsonianos tratados con rotenona mostraron deterioro del comportamiento motriz, pérdida de peso, pérdida de neuronas dopaminérgicas y disminución de la reactividad inmune en las secciones de las regiones nigroestriadas. El tratamiento con polidatina + rotenona mostró efectos contrarios al parkinsonismo, con mejoría en la pérdida de peso, en el comportamiento motor, en la pérdida dopaminérgica y en la reactividad inmune contra las neuronas dopaminérgicas. El presente estudio reveló un potencial de protección neuronal de la polidatina en el modelo animal de la EP al mejorar las anomalías neuro-motoras y la degeneración de las neuronas dopaminérgicas en las regiones nigroestriatales.


Subject(s)
Animals , Male , Rats , Parkinson Disease/drug therapy , Stilbenes/administration & dosage , Glucosides/administration & dosage , Parkinson Disease/pathology , Rotenone/toxicity , Immunohistochemistry , Dopamine , Rats, Sprague-Dawley , Neuroprotective Agents , Disease Models, Animal , Movement Disorders/prevention & control , Nerve Degeneration/prevention & control
3.
Neurotoxicology ; 65: 85-97, 2018 03.
Article in English | MEDLINE | ID: mdl-29425760

ABSTRACT

Flavonoids are bioactive compounds that are known to be neuroprotective against glutamate-mediated excitotoxicity, one of the major causes of neurodegeneration. The mechanisms underlying these effects are unresolved, but recent evidence indicates flavonoids may modulate estrogen signaling, which can delay the onset and ameliorate the severity of neurodegenerative disorders. Furthermore, the roles played by glial cells in the neuroprotective effects of flavonoids are poorly understood. The aim of this study was to investigate the effects of the flavonoid agathisflavone (FAB) in primary neuron-glial co-cultures from postnatal rat cerebral cortex. Compared to controls, treatment with FAB significantly increased the number of neuronal progenitors and mature neurons, without increasing astrocytes or microglia. These pro-neuronal effects of FAB were suppressed by antagonists of estrogen receptors (ERα and ERß). In addition, treatment with FAB significantly reduced cell death induced by glutamate and this was associated with reduced expression levels of pro-inflammatory (M1) microglial cytokines, including TNFα, IL1ß and IL6, which are associated with neurotoxicity, and increased expression of IL10 and Arginase 1, which are associated with anti-inflammatory (M2) neuroprotective microglia. We also observed that FAB increased neuroprotective trophic factors, such as BDNF, NGF, NT4 and GDNF. The neuroprotective effects of FAB were also associated with increased expression of glutamate regulatory proteins in astrocytes, namely glutamine synthetase (GS) and Excitatory Amino Acid Transporter 1 (EAAT1). These findings indicate that FAB acting via estrogen signaling stimulates production of neurons in vitro and enhances the neuroprotective properties of microglia and astrocytes to significantly ameliorate glutamate-mediated neurotoxicity.


Subject(s)
Biflavonoids/pharmacology , Fabaceae , Glutamic Acid/adverse effects , Nerve Degeneration/prevention & control , Neurogenesis/drug effects , Animals , Astrocytes/drug effects , Biflavonoids/antagonists & inhibitors , Cell Death/drug effects , Cerebral Cortex , Coculture Techniques , Cytokines/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Fabaceae/chemistry , Glutamate-Ammonia Ligase/metabolism , Microglia/drug effects , Microglia/metabolism , Nerve Degeneration/chemically induced , Nerve Growth Factors/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats
4.
Nutr Neurosci ; 19(5): 224-30, 2016 Jun.
Article in English | MEDLINE | ID: mdl-25760797

ABSTRACT

OBJECTIVES: This study aimed to compare the effects of environmental enrichment in nourished (on a diet containing 16% protein) and malnourished (on a diet containing 6% protein) rats during the critical period of brain development, specifically focusing on the optic nerve. METHODS: By means of morphologic and morphometric assessment of the optic nerve, we analyzed the changes caused by diet and stimulation (environmental enrichment) on postnatal day 35, a time point ideal for such morphological analysis since developmental processes are considered complete at this age. RESULTS: Malnourished animals presented low body and brain weights and high body-to-brain weight ratio compared to well-nourished rats. Furthermore, malnourished animals showed morphological changes in the optic nerve such as edema and vacuolization characterized by increased interstitial space. The malnourished-stimulated group presented lesions characteristic of early protein malnutrition but were milder than lesions exhibited by malnourished-non-stimulated group. The morphometric analysis revealed no difference in glial cell density between groups, but there was significantly higher blood vessel density in the stimulated rats, independent of their nutritional condition. DISCUSSION: Our data indicate that protein malnutrition imposed during the critical period of brain development alters the cytoarchitecture of the optic nerve. In addition, we affirm that a 1-hour exposure to an enriched environment everyday was sufficient for tissue preservation in rats maintained on a low-protein diet. This protective effect might be related to angiogenesis, as confirmed by the increased vascular density observed in morphometric analyses.


Subject(s)
Disease Models, Animal , Lactation , Maternal Nutritional Physiological Phenomena , Nerve Degeneration/prevention & control , Optic Tract/blood supply , Photic Stimulation , Protein Deficiency/physiopathology , Animals , Animals, Newborn , Blood Vessels/pathology , Blood Vessels/physiopathology , Edema/etiology , Female , Male , Neovascularization, Physiologic , Nerve Degeneration/etiology , Optic Nerve/blood supply , Optic Nerve/pathology , Optic Nerve/physiopathology , Optic Tract/pathology , Optic Tract/physiopathology , Organ Size , Protein Deficiency/pathology , Random Allocation , Rats, Wistar , Vacuoles/pathology , Weight Gain
5.
J Cereb Blood Flow Metab ; 35(12): 1937-49, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26104286

ABSTRACT

Acute ischemic stroke is a cerebrovascular accident and it is the most common cause of physical disabilities around the globe. Patients may present with repeated ictuses, experiencing mental consequences, such as depression and cognitive disorders. Cyclin-dependent kinase 5 (CDK5) is a kinase that is involved in neurotransmission and plasticity, but its dysregulation contributes to cognitive disorders and dementia. Gene therapy targeting CDK5 was administered to the right hippocampus of ischemic rats during transient cerebral middle artery occlusion. Physiologic parameters (blood pressure, pH, pO2, and pCO2) were measured. The CDK5 downregulation resulted in neurologic and motor improvement during the first week after ischemia. Cyclin-dependent kinase 5 RNA interference (RNAi) prevented dysfunctions in learning, memory, and reversal learning at 1 month after ischemia. These observations were supported by the prevention of neuronal loss, the reduction of microtubule-associated protein 2 (MAP2) immunoreactivity, and a decrease in astroglial and microglia hyperreactivities and tauopathy. Additionally, CDK5 silencing led to an increase in the expression of brain-derived neurotrophic factor (BDNF), its Tropomyosin Receptor kinase B (TRKB) receptor, and activation of cyclic AMP response element-binding protein (CREB) and extracellular signal-regulated kinase (ERK), which are important targets in neuronal plasticity. Together, our findings suggest that gene therapy based on CDK5 silencing prevents cerebral ischemia-induced neurodegeneration and motor and cognitive deficits.


Subject(s)
Brain Ischemia/complications , Cognition Disorders/etiology , Cognition Disorders/prevention & control , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Hippocampus/pathology , Nerve Degeneration/genetics , Nerve Degeneration/prevention & control , Stroke/complications , Animals , Blood Pressure , Brain Chemistry/genetics , Brain Ischemia/pathology , Brain Ischemia/psychology , Carbon Dioxide/blood , Gene Knockdown Techniques , Hydrogen-Ion Concentration , Learning Disabilities/etiology , Learning Disabilities/prevention & control , Learning Disabilities/psychology , Male , Maze Learning , Memory Disorders/etiology , Memory Disorders/prevention & control , Memory Disorders/psychology , Movement Disorders/etiology , Movement Disorders/prevention & control , Movement Disorders/psychology , Nerve Degeneration/pathology , Oxygen/blood , RNA Interference , Rats , Reversal Learning/drug effects , Stroke/pathology , Stroke/psychology
6.
Rev. méd. Chile ; 143(1): 120-123, ene. 2015. ilus
Article in Spanish | LILACS | ID: lil-742561

ABSTRACT

We report a 37 years old male with a dermatomyositis treated with oral cyclophosphamide. He was admitted to the hospital due to a zone of skin necrosis with purulent exudate, located in the second left toe. A complete blood count showed a leukocyte count of 2,600 cells/mm³. A Chest CAT scan showed a pneumomediastinum with emphysema of adjacent soft tissue. Cyclophosphamide was discontinued and leukocyte count improved. The affected toe was amputated and a chest CAT scan showed a partial resolution of the pneumomediastinum. We discuss and review the pathogenesis, clinical presentation and management of pneumomediastinum and cutaneous necrosis in association with dermatomyositis.


Subject(s)
Animals , Female , Rats , Benzoxazines/therapeutic use , Cannabinoids/agonists , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Morpholines/therapeutic use , Naphthalenes/therapeutic use , Neurons/drug effects , Oligodendroglia/drug effects , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , /metabolism , Caspase 9/metabolism , Cell Count/methods , Central Nervous System/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Macrophages/drug effects , Nerve Degeneration/etiology , Nerve Degeneration/prevention & control , Neurologic Examination , Poly(ADP-ribose) Polymerases/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , T-Lymphocytes/drug effects , Time Factors
7.
Biomedica ; 34(3): 366-78, 2014.
Article in Spanish | MEDLINE | ID: mdl-25504124

ABSTRACT

INTRODUCTION: Stroke is the second leading cause of death and the first cause of disability in the world, with more than 85% of the cases having ischemic origin. OBJECTIVE: To evaluate in an embolism model of stroke the effect of atorvastatin and meloxicam on neurons, astrocytes and microglia. This evaluation was done administering each medication individually and in association. MATERIALS AND METHODS: Wistar rats were subjected to carotid arterial embolism and treatment with meloxicam and atorvastatin at 6, 24, 48 and 72 hours. Using immunohistochemistry, we evaluated the immunoreactivity of COX-2 protein, GFAP and OX-42 in neurons, astrocytes and microglia by densitometric and morphological studies. Data were evaluated by variance analysis and non-parametric multiple comparison. RESULTS: Cerebral ischemia by arterial embolism increased significantly the reactivity of microglia and astrocytes (p<0.001), whereas it was reduced by atorvastatin, meloxicam and their association. Ischemia produced astrocytic shortening, cellular thickening, protoplasmic rupture expansions (clasmatodendrosis) and microglial morphological changes characteristic of various activity stages. In perifocal areas, immunoreactivity of COX-2 was increased and in the ischemic focus it was reduced, while meloxicam and atorvastatin significantly reduced (p<0.001) perifocal immunoreactivity, restoring the marking of cyclooxygenase in the ischemic focus. CONCLUSION: These results suggest that the meloxicam-atorvastatin association attenuates astrocytic and microglial response in the inflammatory process after cerebral ischemia by arterial embolism, reducing neurodegeneration and restoring the morphological and functional balance of nervous tissue .


Subject(s)
Brain Ischemia/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Intracranial Embolism/complications , Nerve Degeneration/prevention & control , Pyrroles/therapeutic use , Thiazines/therapeutic use , Thiazoles/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/pathology , Atorvastatin , Biomarkers , Brain Ischemia/etiology , Brain Ischemia/pathology , CD11b Antigen/analysis , Carotid Stenosis/complications , Carotid Stenosis/pathology , Cyclooxygenase 2 , Cyclooxygenase Inhibitors/administration & dosage , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Glial Fibrillary Acidic Protein/analysis , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Inflammation , Intracranial Embolism/pathology , Meloxicam , Microglia/drug effects , Microglia/pathology , Nerve Tissue Proteins/analysis , Pyrroles/administration & dosage , Random Allocation , Rats , Rats, Wistar , Thiazines/administration & dosage , Thiazoles/administration & dosage
8.
Biomédica (Bogotá) ; Biomédica (Bogotá);34(3): 366-378, July-Sept. 2014. ilus
Article in Spanish | LILACS | ID: lil-726786

ABSTRACT

Introducción. El accidente cerebrovascular es la segunda causa de muerte y la primera de discapacidad en el mundo, y más de 85 % es de origen isquémico. Objetivo. Evaluar en un modelo de infarto cerebral por embolia arterial el efecto de la atorvastatina y el meloxicam, administrados por separado y de forma conjunta, sobre la respuesta neuronal, los astrocitos y la microglia. Materiales y métodos. Se sometieron ratas Wistar a embolia de la arteria carótida y a tratamiento con meloxicam y atorvastatina, administrados por separado y conjuntamente, a las 6, 24, 48 y 72 horas. Se evaluó la reacción de las proteínas COX-2, GFAP y OX-42 en las neuronas, los astrocitos y la microglia mediante inmunohistoquímica y estudios morfológicos y de densitometría. Los datos obtenidos se evaluaron por medio de un análisis de varianza y de pruebas no paramétricas de comparación múltiple. Resultados. La isquemia cerebral por embolia arterial incrementó significativamente (p<0,001) la reacción de los astrocitos y la microglia, en tanto que la atorvastatina y el meloxicam, administrados por separado y de forma conjunta, la redujeron. La isquemia produjo acortamiento de las proyecciones de los astrocitos, engrosamiento celular, ruptura de las expansiones protoplásmicas (clasmatodendrosis) y cambios morfológicos en la microglia propios de diversas etapas de actividad. En las zonas circundantes del foco se incrementó la reacción inmunológica de la COX-2 y se redujo en el foco isquémico, en tanto que el meloxicam y la atorvastatina redujeron significativamente (p<0,001) la reacción inmunológica en la zona circundante del foco, restableciendo la marcación de la ciclooxigenasa en el foco isquémico. Conclusión. La combinación de meloxicam y atorvastatina atenúa la respuesta de los astrocitos y la microglia en el proceso inflamatorio posterior a la isquemia cerebral por embolia arterial, reduciendo la degeneración neuronal y restableciendo el equilibrio morfológico y funcional del tejido nervioso.


Introduction: Stroke is the second leading cause of death and the first cause of disability in the world, with more than 85% of the cases having ischemic origin. Objective: To evaluate in an embolism model of stroke the effect of atorvastatin and meloxicam on neurons, astrocytes and microglia. This evaluation was done administering each medication individually and in association. Materials and methods: Wistar rats were subjected to carotid arterial embolism and treatment with meloxicam and atorvastatin at 6, 24, 48 and 72 hours. Using immunohistochemistry, we evaluated the immunoreactivity of COX-2 protein, GFAP and OX-42 in neurons, astrocytes and microglia by densitometric and morphological studies. Data were evaluated by variance analysis and non-parametric multiple comparison. Results: Cerebral ischemia by arterial embolism increased significantly the reactivity of microglia and astrocytes (p<0.001), whereas it was reduced by atorvastatin, meloxicam and their association. Ischemia produced astrocytic shortening, cellular thickening, protoplasmic rupture expansions (clasmatodendrosis) and microglial morphological changes characteristic of various activity stages. In perifocal areas, immunoreactivity of COX-2 was increased and in the ischemic focus it was reduced, while meloxicam and atorvastatin significantly reduced (p<0.001) perifocal immunoreactivity, restoring the marking of cyclooxygenase in the ischemic focus. Conclusion: These results suggest that the meloxicam-atorvastatin association attenuates astrocytic and microglial response in the inflammatory process after cerebral ischemia by arterial embolism, reducing neurodegeneration and restoring the morphological and functional balance of nervous tissue .


Subject(s)
Animals , Female , Rats , Brain Ischemia/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Intracranial Embolism/complications , Nerve Degeneration/prevention & control , Pyrroles/therapeutic use , Thiazines/therapeutic use , Thiazoles/therapeutic use , Atorvastatin , /analysis , Astrocytes/drug effects , Astrocytes/pathology , Biomarkers , Brain Ischemia/etiology , Brain Ischemia/pathology , Carotid Stenosis/complications , Carotid Stenosis/pathology , Cyclooxygenase Inhibitors/administration & dosage , Disease Models, Animal , Drug Evaluation, Preclinical , Glial Fibrillary Acidic Protein/analysis , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Inflammation , Intracranial Embolism/pathology , Microglia/drug effects , Microglia/pathology , Nerve Tissue Proteins/analysis , Pyrroles/administration & dosage , Random Allocation , Rats, Wistar , Thiazines/administration & dosage , Thiazoles/administration & dosage
9.
BMC Neurosci ; 14: 104, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24059981

ABSTRACT

BACKGROUND: Some neurodegenerative diseases, such as Alzheimer and Parkinson, present an olfactory impairment in early stages, and sometimes even before the clinical symptoms begin. In this study, we assess the role of CA1 hippocampus (structure highly affected in Alzheimer disease) subfield in the rats' olfactory behavior, and the neuroprotective effect of 17 beta estradiol (E2) against the oxidative stress produced by the injection of amyloid beta 25-35. RESULTS: 162 Wistar rats were ovariectomized and two weeks after injected with 2 µl of amyloid beta 25-35 (A-ß25-35) in CA1 subfield. Olfactory behavior was evaluated with a social recognition test, odor discrimination, and search tests. Oxidative stress was evaluated with FOX assay and Western Blot against 4-HNE, Fluoro Jade staining was made to quantify degenerated neurons; all these evaluations were performed 24 h, 8 or 15 days after A-ß25-35 injection. Three additional groups treated with 17 beta estradiol (E2) were also evaluated. The injection of A-ß25-35 produced an olfactory impairment 24 h and 8 days after, whereas a partial recovery of the olfactory behavior was observed at 15 days. A complete prevention of the olfactory impairment was observed with the administration of E2 two weeks before the amyloid injection (A-ß25-35 24 h + E2) and one or two weeks after (groups 8 A-ß +E2 and 15 A-ß +E2 days, respectively); a decrease of the oxidative stress and neurodegeneration were also observed. CONCLUSIONS: Our finding shows that CA1 hippocampus subfield plays an important role in the olfactory behavior of the rat. The oxidative stress generated by the administration of A-ß25-35 is enough to produce an olfactory impairment. This can be prevented with the administration of E2 before and after amyloid injection. This suggests a possible therapeutic use of estradiol in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , CA1 Region, Hippocampal/pathology , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Olfaction Disorders/prevention & control , Animals , Blotting, Western , CA1 Region, Hippocampal/drug effects , Female , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Olfaction Disorders/chemically induced , Oxidative Stress/drug effects , Rats , Rats, Wistar
10.
Braz J Med Biol Res ; 46(7): 580-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23903681

ABSTRACT

Sublethal ischemic preconditioning (IPC) is a powerful inducer of ischemic brain tolerance. However, its underlying mechanisms are still not well understood. In this study, we chose four different IPC paradigms, namely 5 min (5 min duration), 5×5 min (5 min duration, 2 episodes, 15-min interval), 5×5×5 min (5 min duration, 3 episodes, 15-min intervals), and 15 min (15 min duration), and demonstrated that three episodes of 5 min IPC activated autophagy to the greatest extent 24 h after IPC, as evidenced by Beclin expression and LC3-I/II conversion. Autophagic activation was mediated by the tuberous sclerosis type 1 (TSC1)-mTor signal pathway as IPC increased TSC1 but decreased mTor phosphorylation. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and hematoxylin and eosin staining confirmed that IPC protected against cerebral ischemic/reperfusion (I/R) injury. Critically, 3-methyladenine, an inhibitor of autophagy, abolished the neuroprotection of IPC and, by contrast, rapamycin, an autophagy inducer, potentiated it. Cleaved caspase-3 expression, neurological scores, and infarct volume in different groups further confirmed the protection of IPC against I/R injury. Taken together, our data indicate that autophagy activation might underlie the protection of IPC against ischemic injury by inhibiting apoptosis.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Brain Ischemia/physiopathology , Ischemic Preconditioning/methods , Nerve Degeneration/prevention & control , Reperfusion Injury/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Brain Ischemia/prevention & control , Caspase 3/metabolism , Cerebrum/injuries , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling , Male , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Time Factors , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/metabolism
11.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;46(7): 580-588, ago. 2013. graf
Article in English | LILACS | ID: lil-682394

ABSTRACT

Sublethal ischemic preconditioning (IPC) is a powerful inducer of ischemic brain tolerance. However, its underlying mechanisms are still not well understood. In this study, we chose four different IPC paradigms, namely 5 min (5 min duration), 5×5 min (5 min duration, 2 episodes, 15-min interval), 5×5×5 min (5 min duration, 3 episodes, 15-min intervals), and 15 min (15 min duration), and demonstrated that three episodes of 5 min IPC activated autophagy to the greatest extent 24 h after IPC, as evidenced by Beclin expression and LC3-I/II conversion. Autophagic activation was mediated by the tuberous sclerosis type 1 (TSC1)-mTor signal pathway as IPC increased TSC1 but decreased mTor phosphorylation. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and hematoxylin and eosin staining confirmed that IPC protected against cerebral ischemic/reperfusion (I/R) injury. Critically, 3-methyladenine, an inhibitor of autophagy, abolished the neuroprotection of IPC and, by contrast, rapamycin, an autophagy inducer, potentiated it. Cleaved caspase-3 expression, neurological scores, and infarct volume in different groups further confirmed the protection of IPC against I/R injury. Taken together, our data indicate that autophagy activation might underlie the protection of IPC against ischemic injury by inhibiting apoptosis.


Subject(s)
Animals , Male , Rats , Apoptosis/physiology , Autophagy/physiology , Brain Ischemia/physiopathology , Ischemic Preconditioning/methods , Nerve Degeneration/prevention & control , Reperfusion Injury/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Brain Ischemia/prevention & control , /metabolism , Cerebrum/injuries , In Situ Nick-End Labeling , Immunosuppressive Agents/pharmacology , Rats, Sprague-Dawley , Sirolimus/pharmacology , Time Factors , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
12.
Neurosci Res ; 74(2): 156-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22967672

ABSTRACT

We have previously shown that the intrastriatal injection of the C-terminal domain of tetanus toxin (Hc-TeTx) protects the nigrostriatal-dopaminergic pathways and improves motor behavior in hemiparkinsonism-rat models caused by MPP(+) (1-methyl-4-phenylpyridinium). Here we have investigated the protective effects of the intramuscular application of the Hc-TeTx on motor asymmetry and neurodegeneration in the striatum of 6-hydroxydopamine (6-OHDA)-treated rats. Adult male rats were intramuscularly injected with the recombinant Hc-TeTx protein (0.1-20µg/kg, daily) 3days before the stereotaxic injection of 6-OHDA into the left striatum. Our results showed that the motor-improvement functions were extended for 4weeks in all Hc-TeTx-treated groups, obtaining the maximum performance with the highest dose of Hc-TeTx (20µg/kg). The improvements found were 97%, 87%, and 70% in the turning behavior, stepping test, and cylinder test, respectively. The striatal levels of dopamine and its metabolites did not vary compared to the control group. Moreover, the peripheral treatment with Hc-TeTx in rats prevents, for 30days, the neurodegeneration in the striatum caused by the toxicity of the 6-OHDA. Our results lead us to believe that the Hc-TeTx could be a potential therapeutic agent in pathologies caused by impairment of dopaminergic innervations such as Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Motor Activity/drug effects , Neuroprotective Agents/therapeutic use , Oxidopamine/toxicity , Parkinsonian Disorders/drug therapy , Peptide Fragments/therapeutic use , Tetanus Toxin/therapeutic use , Animals , Apoptosis/drug effects , Astrocytes/pathology , Catecholamines/metabolism , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dopamine/metabolism , Drug Evaluation, Preclinical , Injections, Intramuscular , Locomotion/drug effects , Locomotion/physiology , Male , Motor Activity/physiology , Muscle, Skeletal , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/chemically induced , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Tetanus Toxin/administration & dosage , Tetanus Toxin/pharmacology
13.
Brain Res ; 1474: 110-7, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22885341

ABSTRACT

Status epilepticus (SE) when occurred during brain development can cause short- and long-term consequences, which are frequently associated with NMDA-mediated glutamatergic excitotoxicity. In the present work, we investigated the putative neuroprotective role of ketamine, an NMDA receptor antagonist, on early life SE-induced acute neuronal death and long-term behavioral abnormalities. Male Wistar rats (16 postnatal days) were induced to SE by LiCl-pilocarpine i.p. administration (3 mEq/kg; 60 mg/kg, respectively). Fifteen or 60min after pilocarpine injection, animals received a ketamine administration (22.5mg/kg i.p.). Neuronal degeneration was assessed 24h after SE induction. Another subset of animals was destined to behavioral tasks in adulthood (75-80 postnatal days). Fluoro-Jade C labeling revealed a marked neuronal death on CA1 hippocampal subfield, habenula, thalamus and amygdala in SE animals. Ketamine post-SE onset treatment prevented neuronal death in all regions assessed. In the elevated plus maze, SE induced an increase in anxiety-like behaviors whereas ketamine administration during seizures was able to prevent this alteration. Ketamine administration in non-SE animals resulted in high anxiety levels. There were no observed differences among groups in the open field task in all parameters analyzed. Our results suggest that ketamine post-SE onset treatment was effective in preventing acute and long-standing alterations caused by SE early in life, which indicates a putative role of glutamatergic system on SE-induced brain damage as well as long-lasting behavioral consequences.


Subject(s)
Anxiety/prevention & control , Ketamine/pharmacology , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Status Epilepticus/drug therapy , Animals , Brain/drug effects , Brain/pathology , Convulsants/toxicity , Male , Nerve Degeneration/etiology , Pilocarpine/toxicity , Rats , Rats, Wistar , Status Epilepticus/pathology , Status Epilepticus/psychology
14.
Nutr Neurosci ; 15(2): 62-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22333997

ABSTRACT

UNLABELLED: One of the main concerns regarding organophosphate pesticides (OP) is their possible toxic effects. Doses that do not produce acute toxicity are capable of altering the structure and biochemistry of different tissues and organs by production of reactive oxygen species (ROS). Curcumin (CUR) is the main substance in Curcuma longa (Zingiberacea) rhizome that has strong antioxidant activity. However, the neuroprotective properties of curcumin against oxidative stress induced by prolonged exposure to parathion (PAR) is not clear. OBJECTIVE: The present work evaluated the protective effect of curcumin against the oxidative damage induced in the rat hippocampus by the OP PAR. METHODS: Forty female Wistar rats were distributed in four groups as follows: exposed to PAR by inhalation (PAR group); pre-treated with CUR and then exposed to PAR by inhalation, (CUR + PAR group); exposed to environmental air and treated with CUR in the food (CUR group); and exposed to environmental air (the control group). At the end of the handling process, the concentration of erythrocyte cholinesterase was monitored, as indicator of PAR intoxication and lipoperoxidation, immunohistochemistry for astrocytes, and activated microglia and apoptosis was determined in the hippocampus. RESULTS: In the present study, we show that the administration of CUR (200 mg/kg body weight) significantly diminished the oxidative damage in the hippocampus of rats exposed to the OP PAR. DISCUSSION: These data suggest that CUR may be an alternative to prevent neurodegenerative damage after pesticide exposure.


Subject(s)
Curcumin/pharmacology , Hippocampus/drug effects , Insecticides/toxicity , Oxidative Stress/drug effects , Parathion/toxicity , Plant Extracts/pharmacology , Animals , Antioxidants/pharmacology , Curcuma/chemistry , Female , Hippocampus/pathology , Nerve Degeneration/prevention & control , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
15.
Brain Res Bull ; 84(3): 252-7, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21256199

ABSTRACT

Sleep deprivation (SD) produces numerous deleterious changes in brain cells, including apoptosis. It has been demonstrated that growth hormone (GH) stimulates cell growth and counteracts apoptosis, although this anti-apoptotic effect has not been tested against SD. To determine the protective effect of GH administration on cell proliferation and survival in the dentate gyrus (DG) of the hippocampus after sleep deprivation; we injected Wistar adult rats with a low dose of recombinant human GH (rhGH 5 ng/kg) per seven days and then we gently sleep deprived the animals for 48 consecutive hours. 5-Bromodeoxiuridine (BrdU) was administered to assess cell proliferation after the GH treatment and NeuN was used as marker of cell fate. Our results indicate that GH produced a three fold increase in the number of BrdU positive cells within the DG [Control = 1044 ± 106.38 cells, rhGH = 2952 ± 99.84 cells, P<0.01]. In contrast, 48 h of SD significantly reduced cell proliferation but this effect was antagonized by the GH administration [SD = 540 ± 18.3 cells, rhGH + SD = 1116 ± 84.48 cells, P<0.004]. Paradoxically, SD and GH administration increased cell survival separately but no significantly compared with control animals. However, cell survival was increased in animals treated with rhGH+SD compared to rats injected with saline solution [P<0.04]. Within the survival cells, the percentage of neurons was higher in SD animals [95%] compared with saline group, while this percentage (NeuN positive cells) was increased in animals treated with rhGH+SD [120%] compared with rhGH [25%] alone. Our findings indicate that GH strongly promotes cell proliferation in the adult brain and also protects the hippocampal neuronal precursors against the deleterious effect of prolonged sleep loss.


Subject(s)
Cell Proliferation/drug effects , Hippocampus/drug effects , Human Growth Hormone/pharmacology , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sleep Deprivation/pathology , Animals , Hippocampus/pathology , Hippocampus/physiopathology , Human Growth Hormone/physiology , Humans , Nerve Degeneration/prevention & control , Neurons/pathology , Rats , Rats, Wistar , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/physiology , Sleep Deprivation/complications
16.
Exp Neurol ; 227(2): 237-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21093436

ABSTRACT

Tumor necrosis factor (TNF)-α, a pro-inflammatory cytokine, has been implicated in both neuronal death and survival in Parkinson's disease (PD). The substantia nigra (SN), a CNS region affected in PD, is particularly susceptible to inflammatory insults and possesses the highest density of microglial cells, but the effects of inflammation and in particular TNF-α on neuronal survival in this region remains controversial. Using adenoviral vectors, the CRE/loxP system and hypomorphic mice, we achieved chronic expression of two levels of TNF-α in the SN of adult mice. Chronic low expression of TNF-α levels reduced the nigrostriatal neurodegeneration mediated by intrastriatal 6-hydroxydopamine administration. Protective effects of low TNF-α level could be mediated by TNF-R1, GDNF, and IGF-1 in the SN and SOD activity in the striatum (ST). On the contrary, chronic expression of high levels of TNF-α induced progressive neuronal loss (63% at 20 days and 75% at 100 days). This effect was accompanied by gliosis and an inflammatory infiltrate composed almost exclusively by monocytes/macrophages. The finding that chronic high TNF-α had a slow and progressive neurodegenerative effect in the SN provides an animal model of PD mediated by the chronic expression of a single cytokine. In addition, it supports the view that cytokines are not detrimental or beneficial by themselves, i.e., their level and time of expression among other factors can determine its final effect on CNS damage or protection. These data support the view that new anti-parkinsonian treatments based on anti-inflammatory therapies should consider these dual effects of cytokines on their design.


Subject(s)
Corpus Striatum/metabolism , Dopamine/physiology , Nerve Degeneration/metabolism , Neuroprotective Agents/metabolism , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Age Factors , Animals , Chronic Disease , Corpus Striatum/pathology , Disease Models, Animal , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/prevention & control , Nerve Net/metabolism , Nerve Net/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Substantia Nigra/pathology , Tumor Necrosis Factor-alpha/physiology
17.
Curr Alzheimer Res ; 7(7): 615-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20964623

ABSTRACT

Interleukin-3 (IL-3) regulates the proliferation, survival and differentiation of haematopoietic cells via interaction with specific cell-surface receptors. IL-3 is expressed in several non-hematopoietic cell types. Studies have demonstrated the presence of IL-3 in the central nervous system, however, its physiological role in these cells is poorly understood. Previously we have been demonstrated that IL-3 prevents neuronal death induced by fibrillary ß amyloid in these cells, by PI 3-kinase and Jak/STAT pathway activation. In this study, we demonstrated that IL-3 significantly reduced Aß-promoted neurite degeneration and toxicity. Thus, this cytokine provides cellular protection against Aß neurotoxicity in primary cortical neuronal cells, by modulating microtubular dynamics and prevention of tau cleavage and hyperphosphorylation. We also demonstrates that IL-3 is expressed in the "in vivo" mouse model of AD, Tg2576, which also expresses human AßPP with the Swedish mutation. In summary, these results suggest that IL-3 could play a neuroprotective role in AD.


Subject(s)
Alzheimer Disease/metabolism , Cytoprotection/physiology , Interleukin-3/physiology , Nerve Degeneration/metabolism , Nerve Degeneration/prevention & control , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Cytoprotection/drug effects , Humans , Interleukin-3/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/pathology , tau Proteins/physiology
18.
Neuroimmunomodulation ; 17(3): 146-9, 2010.
Article in English | MEDLINE | ID: mdl-20134188

ABSTRACT

In recent years, a growing list of publications point to the value of steroid hormones as an interesting option for the treatment of several type of lesions and diseases of the nervous system. Progesterone, well known for its role in pregnancy, has recently been shown to exert neuroprotective and promyelinating effects in both, the peripheral and central nervous system, including the injured spinal cord. Previous work from our laboratory has shown that progesterone actions in experimental models of spinal neurodegeneration or injury may involve the modulation of brain-derived neurotrophic factor, a neurotrophin with important implications in neuronal survival and axonal regeneration. The spinal cord is target for progesterone since neurons and glial cells express the intracellular receptors for this neuroactive steroid. However, the presence in the spinal cord of new membrane receptors and the enzymes involved in progesterone metabolism to its reduced derivatives, which modulate the activity of neurotransmitter receptors, suggest that progesterone actions involve pleiotropic mechanisms. Our recent data uncovering several molecular events may help to understand the protective and promyelinating actions of progesterone and further support the role of this steroid as a promising therapeutic agent for neurotrauma and/or neurodegenerative diseases.


Subject(s)
Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Animals , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Humans , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Progesterone/metabolism , Progesterone/therapeutic use , Spinal Cord/metabolism , Spinal Cord/physiopathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
19.
Neurochem Int ; 56(1): 51-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19782116

ABSTRACT

In this study, the cytoprotective effects of caffeine (CAF) and 8-(3-chlorostyryl)-caffeine (CSC), A(2A) receptor antagonists, were tested against 6-OHDA-induced cytotoxicity, in rat mesencephalic cells. Both drugs significantly increased the number of viable cells, after their exposure to 6-OHDA, as measured by the MTT assay. While nitrite levels in the cells were drastically increased by 6-OHDA, their concentrations were brought toward normality after CAF or CSC, indicating that both drugs block 6-OHDA-induced oxidative stress which leads to free radicals generation. A complete blockade of 6-OHDA-induced lipid peroxidation, considered as a major source of DNA damage, was observed after cells treatment with CAF or CSC. 6-OHDA decreased the number of normal cells while increasing the number of apoptotic cells. In the CAF plus 6-OHDA group, a significant recover in the number of viable cells and a decrease in the number of apoptotic cells were seen, as compared to the group treated with 6-OHDA alone. A similar effect was observed after cells exposure to CSC in the presence of 6-OHDA. Unexpectedly, while a significant lower number of activated microglia was observed after cells exposure to CAF plus 6-OHDA, this was not the case after cells exposure to CSC under the same conditions. While CAF lowered the percentage of reactive astrocytes increased by 6-OHDA, CSC presented no effect. The effects of these drugs were also examined on the releases of myeloperoxidase (MPO), an inflammatory marker, and lactate dehydrogenase (LDH), a marker for cytotoxicity, in human neutrophils, in vitro. CSC and CAF (0.1, 1 and 10 microg/ml) produced inhibitions of the MPO release from PMA-stimulated cells, ranging from 45 to 83%. In addition, CSC and CAF (5, 50 and 100 microg/ml) did not show any cytotoxicity in the range of concentrations used, as determined by the LDH assay. All together, our results showed a strong neuroptrotection afforded by caffeine or CSC, on rat mesencephalic cells exposed to 6-OHDA. Furthermore, CSC and caffeine actions, inhibiting MPO as well as LDH releases, would contribute to their possible benefit in the treatment of neurodegenerative diseases, including DP. These effects are partially due to the ability of these A(2A) antagonists to decrease the cells free radicals production and oxidative stress, that are major components of 6-OHDA-induced cytotoxicity.


Subject(s)
Adenosine A2 Receptor Agonists , Caffeine/pharmacology , Neurons/drug effects , Parkinsonian Disorders/drug therapy , Substantia Nigra/drug effects , Xanthines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caffeine/therapeutic use , Cells, Cultured , Encephalitis/drug therapy , Encephalitis/metabolism , Encephalitis/physiopathology , Female , Free Radicals/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Microglia/drug effects , Microglia/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitrites/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Oxidopamine/pharmacology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Rats , Rats, Wistar , Receptor, Adenosine A2A/metabolism , Substantia Nigra/metabolism , Substantia Nigra/physiopathology , Sympatholytics/pharmacology , Xanthines/therapeutic use
20.
Ann Anat ; 192(1): 52-7, 2010 Feb 20.
Article in English | MEDLINE | ID: mdl-19889522

ABSTRACT

Aging leads to changes in cardiac structure and function. Evidence suggests that the practice of regular exercise may prevent disturbances in the cardiovascular system during aging. We studied the effects of aging on the morphology and morphometry of cardiac neurons in Wistar rats and investigated whether a lifelong moderate exercise program could exert a protective effect toward some deleterious effects of aging. Aging caused a significant decline (28%) in the number of NADH-diaphorase-stained cardiac neurons. Animals submitted to a daily session of 60 min, 5 day/week, at 1.1 km/h of running in treadmill over the entire life span exhibited a reversion of the observed decline in the number of cardiac neurons. However, most interesting was that the introduction of this lifelong exercise protocol dramatically altered the sizes of cardiac neurons. There was a notable increase in the percentage of small neurons in the rats of the exercise group compared to the sedentary animals. This is the first time that a protective effect of lifelong regular aerobic exercise has been demonstrated on the deleterious effects of aging in cardiac neurons.


Subject(s)
Heart/physiology , Nerve Degeneration/prevention & control , Physical Conditioning, Animal/physiology , Aging , Animals , Body Weight , Cell Count , Heart/growth & development , Heart/physiopathology , Heart Ventricles/anatomy & histology , Male , Nerve Degeneration/physiopathology , Neurons/cytology , Neurons/physiology , Organ Size , Physical Exertion , Rats , Rats, Wistar , Sedentary Behavior
SELECTION OF CITATIONS
SEARCH DETAIL