Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
3.
Annu Rev Nurs Res ; 33: 31-73, 2015.
Article in English | MEDLINE | ID: mdl-25946383

ABSTRACT

Traumatic brain injury (TBI) is the leading cause of mortality and morbidity in the younger population worldwide. Survivors of TBI often experience long-term disability in the form of cognitive, sensorimotor, and affective impairments. Despite the high prevalence in, and cost of TBI to, both individuals and society, some of its underlying pathophysiology is not completely understood. Animal models have been developed over the past few decades to closely replicate the different facets of TBI in humans to better understand the underlying pathophysiology and behavioral impairments and assess potential therapies that can promote neuroprotection. However, no effective treatment for TBI has been established to date in the clinical setting, despite promising results generated in preclinical studies in the use of neuroprotective strategies. The failure to translate results from preclinical studies to the clinical setting underscores a compelling need to revisit the current state of knowledge in the use of animal models in TBI.


Subject(s)
Behavior, Animal , Biomedical Research , Brain Injuries/physiopathology , Disease Models, Animal , Animals , Blast Injuries/metabolism , Blast Injuries/physiopathology , Blast Injuries/psychology , Brain Concussion/metabolism , Brain Concussion/physiopathology , Brain Concussion/psychology , Brain Injuries/metabolism , Brain Injuries/psychology , Brain Injury, Chronic/metabolism , Brain Injury, Chronic/physiopathology , Brain Injury, Chronic/psychology , Cats , Cell Death , Glucose/metabolism , Head Injuries, Penetrating/metabolism , Head Injuries, Penetrating/physiopathology , Head Injuries, Penetrating/psychology , Homeostasis , Humans , Lipid Peroxidation , Mice , Rats , Swine
4.
Neuropathol Appl Neurobiol ; 41(4): 544-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24750178

ABSTRACT

AIMS: Use of enriched environment (EE) housing has been shown to promote recovery from cerebral ischaemic injury but the underlying mechanisms of their beneficial effects remains unclear. Here we examined whether the beneficial effects of EE housing on ischaemia-induced neurodegeneration and cognitive impairment are associated with increased insulin-like growth factor-1 (IGF-1) signalling in the hippocampus. METHODS: Forty-two adult male Wistar rats were included in the study and received either ischaemia or sham surgery. Rats in each group were further randomized to either: EE or standard laboratory cage housing (control). Rats were placed in their assigned housing condition immediately after recovery from anaesthesia. Behavioural testing in the cued learning and discrimination learning tasks were conducted 2 weeks after ischaemia. Rats were euthanized after behavioural testing and the hippocampus was analysed for IGF-1 level, IGF-1 receptor (IGF-1R) activation, protein kinase B (Akt) pathway activation, neurone loss and caspase 3 expression. RESULTS: Our data showed that EE housing: (1) mitigated ischaemia-induced neuronal loss; (2) attenuated ischaemia-induced increase in caspase 3 immunoreactivity in the hippocampus; (3) ameliorated ischaemia-induced cognitive impairments; and (4) increased IGF-1R activation and signalling through the Akt pathway after ischaemic injury. CONCLUSION: Ultimately, these findings suggest the possibility that IGF-1 signalling may be one of the underlying mechanisms involved in the beneficial effects of EE in optimizing recovery following cerebral ischaemic injury.


Subject(s)
Environment , Insulin-Like Growth Factor I/metabolism , Ischemic Attack, Transient/metabolism , Animals , Apoptosis , Hippocampus/metabolism , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/psychology , Male , Maze Learning , Phosphorylation , Rats , Rats, Wistar , Recovery of Function , Signal Transduction , Up-Regulation
5.
Behav Brain Res ; 279: 112-22, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-24632471

ABSTRACT

Here we examined the involvement of insulin-like growth factor 1 (IGF-1) on chemotherapy-induced cognitive impairment. Sixty-four ovariectomized female Sprague-Dawley rats were included in the study and given cyclophosphamide, methothrexate, and 5-fluorouracil (CMF) drug combination or saline (control). CMF was given once a week for 4 weeks. In one experiment, behavioral testing using the cued learning and spontaneous object recognition tasks were performed either: at the end of treatment or 4 weeks after treatment. In another experiment, rats from the chemotherapy and saline groups received either: continuous insulin-like growth factor 1 (IGF-1) or vehicle delivered subcutaneously via osmotic pump for 21 days (started the week after completion of therapy). Bromodeoxyuridine injections were given for 3 consecutive days starting at 2 weeks after completion of chemotherapy to assess the survival of proliferating cells. Increased levels of IGF-1 and activation of its receptor as well as increased activation of Akt and Erk1/2, its downstream signaling pathways was seen immediately after completion of chemotherapy but decreased 4 weeks after treatment. Behavioral testing showed CMF-induced cognitive impairment after completion of therapy and persisted for 4 weeks. We also found that giving IGF-1 significantly increased activation of its receptor, and the Akt and Erk1/2 pathways, and most importantly attenuated chemotherapy-induced cognitive impairment. CMF-induced neuronal apoptosis was also seen and the ratio of surviving cells that proliferate was higher compared to the number of apoptotic cells in the CMF rats given IGF-1. These results suggest that IGF-1 is involved in CMF-induced cognitive impairment by modulating cell death and cell proliferation.

6.
J Neuroinflammation ; 11: 13, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24447880

ABSTRACT

Although neuroinflammation has been studied extensively in animal models of cerebral ischemia, their contrasting functions are still not completely understood. A major participant in neuroinflammation is microglia and microglial activation usually regulated by the chemokine CX3CL1 (fractalkine) and its receptor, CX3CR1. Here, we examined the involvement of CX3CR1 on ischemia-induced chronic neuroinflammation and cognitive function using small interfering RNA (siRNA). Forty adult male Wistar rats were included in the study and received either ischemia or sham surgery then were randomized to receive either CX3CR1 siRNA or scrambled RNA as control starting at 7 days after reperfusion. Behavioral testing commenced 28 days after siRNA delivery and all rats were euthanized after behavioral testing. Our data showed that: (i) transient global cerebral ischemia significantly decreased fractalkine/CX3CR1 signaling in the hippocampus; (ii) inhibition of CX3CR1 function exacerbated the ischemia-induced chronic increase in microglial activation and pro-inflammatory cytokine levels; (iii) inhibition of CX3CR1 function worsened ischemia-induced chronic cognitive impairment; (iv) inhibition of CX3CR1 function in sham rats resulted in increased IL-1ß expression and impaired behavioral performance. However, no significant effect of CX3CR1 on ischemia-induced neurodegeneration was seen. The present study provides important insight to understanding the involvement of CX3CR1 in chronic neuroinflammation and cognitive impairment.


Subject(s)
Chemokine CX3CL1/metabolism , Cognition Disorders/etiology , Encephalitis/etiology , Ischemic Attack, Transient/complications , Receptors, Chemokine/metabolism , Signal Transduction/physiology , Animals , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Cognition Disorders/therapy , Discrimination Learning/drug effects , Discrimination Learning/physiology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluoresceins , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Maze Learning/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Rats , Rats, Wistar , Receptors, Chemokine/genetics , Signal Transduction/drug effects
7.
Brain Behav Immun ; 35: 23-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23916895

ABSTRACT

Cognitive impairment is commonly reported as a consequence of chemotherapy and can have considerable impact on everyday life on cancer patients. Thus, it is imperative to have a clear understanding of this phenomenon and the underlying mechanism involved. In the present study we examined the role of neuroinflammation and myelination in chemotherapy-related cognitive impairment. Female Sprague-Dawley rats (12-months old) were used in the study (total n=52, 13rats/group). Rats were randomly assigned to either the chemotherapy or saline control group. The drug combination of cyclophosphamide, methotrexate, and 5-fluorouracil (CMF) was given i.p. once a week for 4weeks. Rats in the control group received normal saline of equal volume. Animals from each group were further randomized to receive either: cyclooxygenase (COX-2) inhibitor, NS-393, to block the inflammatory response or vehicle. NS-398 was given at 10mg/kg i.p. and equal volume of saline (vehicle) was injected i.p. as vehicle. Both NS-398 and vehicle were injected 1h after the first CMF dose and then given daily for 28days then rats were tested in the Y maze. Our data showed that: (1) CMF led to the increase in the levels of inflammatory mediators IL-1ß, TNF-α, and COX-2 while levels of the anti-inflammatory cytokine IL-10 decreased; (2) cognitive impairment and neuroinflammation resulting from CMF persisted 4weeks after the treatment ended; and (3) administration of NS-398 attenuated CMF-induced neuroinflammation and effects on myelin and cognitive impairment. These findings suggest the involvement of neuroinflammation in CMF-induced changes in myelin and myelination, and cognitive impairment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Cognition Disorders/chemically induced , Inflammation/chemically induced , Nerve Fibers, Myelinated/ultrastructure , Animals , Cognition Disorders/immunology , Cognition Disorders/pathology , Corpus Callosum/ultrastructure , Cyclophosphamide/administration & dosage , Cyclophosphamide/toxicity , Cytokines/metabolism , Female , Fluorouracil/administration & dosage , Fluorouracil/toxicity , Inflammation/metabolism , Methotrexate/administration & dosage , Methotrexate/toxicity , Random Allocation , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects
8.
Acta Neuropathol Commun ; 1: 57, 2013 09 06.
Article in English | MEDLINE | ID: mdl-24252176

ABSTRACT

BACKGROUND: Persistent neuroinflammation and disruptions in brain energy metabolism is commonly seen in traumatic brain injury (TBI). Because of the lack of success of most TBI interventions and the documented benefits of environmental enrichment (EE) in enhancing brain plasticity, here we focused our study on use of EE in regulating injury-induced neuroinflammation and disruptions in energy metabolism in the prefrontal cortex and hippocampus. Adult male Wistar rats were used in the study and randomly assigned to receive either: mild TBI (mTBI) using the controlled cortical injury model or sham surgery. Following surgery, rats from each group were further randomized to either: EE housing or standard laboratory housing (CON). After 4 weeks of recovery, cognitive testing was performed using the non-matching-to-sample and delayed non-matching-to-sample tasks. After completion of behavioral testing, levels of the pro-inflammatory cytokines IL-1ß and TNF-α and the anti-inflammatory cytokine IL-10 were measured. In addition, levels of AMPK (adenosine monophosphate-activated protein kinase), phosphorylated AMPK and uMtCK (ubiquitous mitochondrial creatine kinase) were assessed as measures of brain energy homeostasis. RESULTS: Our results showed that EE: (1) decreased the pro-inflammatory cytokines IL-1ß and TNF-α and enhanced levels of the anti-inflammatory cytokine IL-10 after mTBI; (2) mitigated mTBI-induced cognitive impairment; and (3) attenuated mTBI-induced downregulation in pAMPK/AMPK ratio and uMtCK levels. CONCLUSIONS: Our data demonstrated the potential of EE to modulate the persistent: (1) neuroinflammatory response seen following mTBI, and (2) persistent disturbance in brain energy homeostasis. It is possible that through the mechanism of modulating neuroinflammation, EE housing was able to restore the disruption in energy metabolism and enhanced functional recovery after mTBI.


Subject(s)
Brain Injuries/physiopathology , Brain Injuries/therapy , Hippocampus/physiopathology , Housing, Animal , Neuroimmunomodulation/physiology , Prefrontal Cortex/physiopathology , AMP-Activated Protein Kinases/metabolism , Animals , Brain Injuries/complications , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Cognition Disorders/therapy , Creatine Kinase, Mitochondrial Form/metabolism , Disease Models, Animal , Environment , Homeostasis/physiology , Male , Phosphorylation , Random Allocation , Rats, Wistar , Recovery of Function/physiology
9.
J Neuroinflammation ; 9: 244, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23098125

ABSTRACT

BACKGROUND: Increasing evidence shows an association between the use of vitamin D and improvement in age-related cognitive decline. In this study, we investigated the possible mechanisms involved in the neuroprotective effects of vitamin D on age-related brain changes and cognitive function. METHODS: Male F344 rats aged 20 months (old) and 6 months (young) were used and randomly assigned to either vitamin D supplementation or no supplementation (control). A total of n = 39 rats were used in the study. Rats were individually housed and the supplementation group received a subcutaneous injection of vitamin D (1, α25-dihydroxyvitamin D3) 42 I.U./Kg for 21 days. Control animals received equal volume of normal saline. Behavioral testing in water maze and spontaneous object recognition tasks started on day 14. Levels of interleukin (IL)-1ß and IL-10 were quantified to assess inflammatory state. Also, beta amyloid (Aß) clearance and Aß load were measured. RESULTS: Our results show that: (1) aged rats demonstrated significant learning and memory impairment overall compared to younger animals. However, the age-related decline in learning and memory was ameliorated by the supplementation of vitamin D. No vitamin D effect on learning and memory was seen in the young animals; 2) the pro-inflammatory cytokine IL-1ß is significantly increased while the anti-inflammatory cytokine IL-10 is significantly decreased in the aged rats compared to the young animals; but this age-related change in inflammatory state was mitigated by vitamin D supplementation. No effects of vitamin D were seen on the IL-1ß and IL-10 expression in the young rats; (3) vitamin D increased Aß clearance and decreased amyloid burden in the aged rats while no significant difference was seen between the young animal groups. CONCLUSIONS: Our data suggest that vitamin D supplementation modulated age-related increase in pro-inflammatory state and amyloid burden. It is possible that these effects of vitamin D mediated the decrease memory impairment seen in the aged rats making it a useful therapeutic option to alleviate the effects of aging on cognitive function.


Subject(s)
Aging , Amyloid beta-Peptides/metabolism , Cognition Disorders/diet therapy , Cytokines/metabolism , Vitamin D/administration & dosage , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Analysis of Variance , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cytokines/genetics , Disease Models, Animal , Male , Maze Learning/drug effects , Neprilysin/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Recognition, Psychology/drug effects , Vitamin D/blood
10.
BMC Neurosci ; 12: 124, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152030

ABSTRACT

BACKGROUND: In this study, we examined the effects of cyclophosphamide, methothrexate, and 5-Fluorouracil (CMF) drug combination on various aspects of learning and memory. We also examined the effects of CMF on cell proliferation and chromatin remodeling as possible underlying mechanisms to explain chemotherapy-associated cognitive dysfunction. Twenty-four adult female Wistar rats were included in the study and had minimitter implantation for continuous activity monitoring two weeks before the chemotherapy regimen was started. Once baseline activity data were collected, rats were randomly assigned to receive either CMF or saline injections given intraperitoneally. Treatments were given once a week for a total of 4 weeks. Two weeks after the last injection, rats were tested in the water maze for spatial learning and memory ability as well as discrimination learning. Bromodeoxyuridine (BrdU) injection was given at 100 mg/Kg intraperitoneally 4 hours prior to euthanasia to determine hippocampal cell proliferation while histone acetylation and histone deacetylase activity was measured to determine CMF effects on chromatin remodeling. RESULTS: Our data showed learning and memory impairment following CMF administration independent of the drug effects on physical activity. In addition, CMF-treated rats showed decreased hippocampal cell proliferation, associated with increased histone acetylation and decreased histone deacetylase activity. CONCLUSIONS: These results suggest the negative consequences of chemotherapy on brain function and that anti-cancer drugs can adversely affect the self-renewal potential of neural progenitor cells and also chromatin remodeling in the hippocampus. The significance of our findings lie on the possible usefulness of animal models in addressing the clinical phenomenon of 'chemobrain.'


Subject(s)
Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly/drug effects , Hippocampus/drug effects , Histones/antagonists & inhibitors , Neurogenesis/drug effects , Acetylation/drug effects , Animals , Chromatin Assembly and Disassembly/physiology , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Cognition Disorders/pathology , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Histones/metabolism , Neurogenesis/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Rats , Rats, Wistar
11.
J Neurotrauma ; 28(12): 2485-92, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21612313

ABSTRACT

In this study, we examined whether enriched environment (EE) housing has direct neuroprotective effects on oxidative damage following transient global cerebral ischemia. Fifty-two adult male Wistar rats were included in the study and received either ischemia or sham surgery. Once fully awake, rats in each group were randomly assigned to either: EE housing or socially paired housing (CON). Animals remained in their assigned environment for 7 days, and then were killed. Our data showed that glutamate receptor expression was significantly higher in the hippocampus of the ischemia CON group than in the ischemia EE group. Furthermore, the oxidative DNA damage, protein oxidation, and neurodegeneration in the hippocampus of the ischemia CON group were significantly increased compared to the ischemia EE group. These results suggest that EE housing possibly modulated the ischemia-induced glutamate excitotoxicity, which then attenuated the oxidative damage and neurodegeneration in the ischemia EE rats.


Subject(s)
Brain Ischemia/metabolism , DNA Damage/physiology , Environment , Oxidative Stress/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Social Environment , Animals , Brain Ischemia/prevention & control , Hippocampus/metabolism , Male , Random Allocation , Rats , Rats, Wistar
12.
J Neurotrauma ; 26(4): 613-25, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19271963

ABSTRACT

In this study, we examined whether ischemia-induced amyloidogenesis could be modulated by environmental "experience," and whether this modulation is associated with improved cognitive functioning. Rats were subjected to either global ischemia or sham surgery and then were randomly assigned to either enriched environment housing (EE) or socially paired housing (controls). After 14 days of differential environmental housing, the rats were tested in the water maze. Our results show decreased C-terminal fragments of the beta-amyloid precursor protein (betaAPP) and decreased amyloid beta (Abeta) load in the ischemic EE rats compared to the ischemic control animals. In addition, Abeta oligomerization was significantly decreased in the ischemic EE animals compared to the ischemic control rats. Further, significantly increased levels of neprilysin, but not insulin-degrading enzyme, amyloid-degrading enzymes, were seen in the ischemic EE rats compared to the ischemic control animals. Behavioral analyses showed that ischemic EE rats performed significantly better on the memory task compared to the ischemic control group. These results suggest that use of multi-sensory environmental enrichment following cerebral ischemia may reduce the accumulation of Abeta peptide in the more pathologic oligomeric form, and consequently may enhance functional recovery.


Subject(s)
Amyloid beta-Peptides/metabolism , Cognition Disorders/therapy , Environment, Controlled , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/therapy , Plaque, Amyloid/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Models, Animal , Hypoxia-Ischemia, Brain/physiopathology , Male , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/physiopathology , Memory Disorders/therapy , Neprilysin/metabolism , Neuropsychological Tests , Plaque, Amyloid/pathology , Rats , Rats, Wistar , Recovery of Function/physiology , Treatment Outcome
13.
Annu Rev Nurs Res ; 25: 219-56, 2007.
Article in English | MEDLINE | ID: mdl-17958294

ABSTRACT

The nervous system as well as the endocrine system maintain extensive communication with the immune system through the influence of hormones and neurotransmitters and also by way of the hardwiring of sympathetic and parasympathetic nerves to the lymphoid organs. There is now convincing evidence that the communication between these three body systems is bidirectional. This chapter will provide a succinct review of how neuroendocrine and immune functions are affected in factors that impact vulnerability, such as aging, acute infection, and central nervous system injury. Given that the relevant literature on these topics is vast, the presentation in this chapter will serve to highlight primary references that reflect state of the science in these systems of focus.


Subject(s)
Health Status , Neuroimmunomodulation/physiology , Nursing Research/organization & administration , Psychoneuroimmunology , Vulnerable Populations , Aging/ethnology , Aging/genetics , Aging/immunology , Autonomic Nervous System/physiology , Bibliometrics , Central Nervous System/injuries , Central Nervous System Diseases/complications , Central Nervous System Diseases/immunology , Central Nervous System Diseases/physiopathology , Health Services Needs and Demand , Humans , Hypothalamo-Hypophyseal System/physiology , Nurse's Role , Pituitary-Adrenal System/physiology , Research Design , Socioeconomic Factors , Vulnerable Populations/ethnology , Vulnerable Populations/statistics & numerical data
14.
Behav Brain Res ; 171(1): 17-25, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16621046

ABSTRACT

In this study we examined whether astrocytic and basic fibroblast growth factor changes after cerebral ischemia can be influenced by rehabilitation training and if these changes are associated with functional improvement. After receiving either ischemia or sham surgery, male adult Wistar rats were assigned to one of two rehabilitation training group: complex environment housing (EC) or paired housing as controls (CON). Rats were tested in the water maze after 14 days of rehabilitation training. Results showed increased expression of reactive astrocytes (GFAP) in all ischemic animals and in the sham EC rats with a significant overall increased seen in the ischemia EC housed animals. The pattern of basic fibroblast growth factor (FGF-2) expression seen was somewhat similar to that of GFAP. Behavioral data showed that even though all animals learned to perform the water maze task over time, the ischemia CON rats took longer to learn the task while all the ischemia EC animals performed as well as the sham groups. Regression analysis showed that increased GFAP was able to explain some of the variances in the behavioral parameters in the water maze of the ischemia EC rats suggesting that the activation of astrocytes in this group probably mediated enhanced functional recovery. Lastly, it is possible that the favorable effect of astrocyte activation after cerebral ischemia was mediated by FGF-2.


Subject(s)
Astrocytes/pathology , Brain Ischemia/pathology , Fibroblast Growth Factor 2/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/pathology , Maze Learning/physiology , Analysis of Variance , Animals , Astrocytes/metabolism , Brain Ischemia/metabolism , Brain Ischemia/therapy , Disease Models, Animal , Gliosis/pathology , Gliosis/prevention & control , Hippocampus/metabolism , Housing, Animal , Immunohistochemistry , Male , Motor Activity/physiology , Neurons/pathology , Random Allocation , Rats , Rats, Wistar , Recovery of Function/physiology , Regression Analysis , Social Environment , Tissue Distribution
15.
Curr Alzheimer Res ; 3(1): 49-54, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16472203

ABSTRACT

Age is the biggest risk factor for the development of neurodegenerative diseases. Consequently, as the population ages it becomes more critical to find ways to avoid the debilitating cost of neurodegenerative diseases such as Alzheimer's. Some of the non-invasive strategies that can potentially slow down the mental decline associated with aging are exercise and use of multi-sensory environmental stimulation. The beneficial effects of both exercise and multi-sensory environmental stimulation have been well-documented, thus it is possible that these strategies can either provide neuroprotection or increase resistance to the development of age-related cognitive problems.


Subject(s)
Alzheimer Disease/prevention & control , Environment , Nerve Regeneration/physiology , Physical Fitness/physiology , Aging/physiology , Alzheimer Disease/pathology , Animals , Exercise/physiology , Humans
16.
Exp Neurol ; 198(2): 530-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16483572

ABSTRACT

Reports have shown that damage to the adult brain can result in adaptive changes in regions adjacent or surrounding the site of the principal injury and that these changes may be modulated by rehabilitation training. In this study, we examined the influence of complex environment housing as a rehabilitation strategy on ischemia-induced synaptic and dendritic changes in the hippocampus. Thirty-six adult male Wistar rats were included in the study and assigned to either transient global cerebral ischemia or sham group. Following ischemic or sham surgery, rats were randomized to either complex environment housing (EC) or social condition (SC, paired housing) group during the rehabilitation period. Following 14 days of rehabilitation, rats were tested in the water maze. Our results showed that: (1) ischemic injury and EC housing were able to independently influence synaptogenesis and dendritic growth in the hippocampal area adjacent to the site of injury, and (2) EC housing-induced synaptic and dendritic changes were accompanied by enhanced functional recovery after transient global cerebral ischemia. These data suggest that behavioral experience during the rehabilitation period may be able to alter the neuronal circuitry in the surrounding region where primary neuronal damage was seen and that such modification may have contributed to functional improvement.


Subject(s)
Behavior, Animal/physiology , Dendrites/physiology , Hippocampus/pathology , Ischemic Attack, Transient , Learning/physiology , Spatial Behavior/physiology , Synapses/physiology , Analysis of Variance , Animals , Cell Death/physiology , Dendrites/pathology , Dendrites/ultrastructure , Disease Models, Animal , Fluoresceins , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/rehabilitation , Male , Microscopy, Electron, Transmission/methods , Neurons/pathology , Organic Chemicals , Rats , Rats, Wistar , Reaction Time , Synapses/pathology , Synapses/ultrastructure , Time Factors
17.
Behav Brain Res ; 168(2): 261-71, 2006 Apr 03.
Article in English | MEDLINE | ID: mdl-16356557

ABSTRACT

In this study we examined whether expression of microtubule-associated protein 2 (MAP2) after transient global cerebral ischemia can be influenced by behavioral experience and if the changes are associated with functional improvement. Rats received either ischemia or sham surgery then assigned to: complex environment housing (EC) or social housing (SC) as controls for 14 days followed by water maze testing. Upregulation of MAP2 was seen in all ischemic animals with a significant overall increase evident in the EC housed rats. Behaviorally, all animals learned to perform the water maze task over time but the ischemia SC rats had the worst performance overall while all the EC housed animals demonstrated the best performance in general. Regression analysis showed that increase MAP2 expression was able to explain some of the variance in the behavioral parameters in the water maze suggesting that this cytoskeletal protein probably played a role in mediating enhanced functional outcomes.


Subject(s)
Cognition Disorders/etiology , Environment , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Behavior, Animal , Cell Count/methods , Disease Models, Animal , Fluoresceins , Gene Expression Regulation/physiology , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry/methods , Male , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Organic Chemicals , Rats , Rats, Wistar , Reaction Time/physiology , Regression Analysis , Swimming
18.
Brain Res ; 1033(1): 51-7, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15680339

ABSTRACT

Damage to the adult brain can result in adaptive plasticity in regions adjacent to the site of the principal insult and that the plastic changes may be modulated by post-injury rehabilitation training. In this study, we examined the effects of rehabilitation training on synaptic morphology in the dentate gyrus following transient global cerebral ischemia and the metabolic correlates of the ultrastructural changes. Forty adult male Wistar rats were included in the study and assigned to either ischemia or sham group. Following ischemic or sham surgery, rats were randomized to either complex environment housing (EC), exercise (EX), or social condition (SC, paired housing) group. Electron microscopy and unbiased stereological methods were used to evaluate synaptic plasticity and the number and size of mitochondria in synaptic axon terminals. Increased number of granule neurons was seen in all ischemic groups and in the sham EC rats. Changes in the number of synapses per neuron in the outer and inner molecular layers of the dentate gyrus parallel those seen in granule neurons. Similarly, ischemia and behavioral experience in EC independently increased the number of synaptic mitochondria in presynaptic terminals in both the outer and inner molecular layers; however, no significant changes were seen in mitochondrial size. These data suggest a link between behavioral training and synaptic plasticity in the region adjacent to the injury and that the likely metabolic correlate of this synaptic plasticity is increased number of mitochondria at synaptic axon terminals.


Subject(s)
Brain Ischemia/physiopathology , Brain Ischemia/rehabilitation , Dentate Gyrus/pathology , Exercise Therapy/methods , Mitochondria/physiology , Presynaptic Terminals/physiology , Synapses/physiology , Animals , Behavior, Animal , Cell Count/methods , Disease Models, Animal , Male , Microscopy, Electron, Transmission/methods , Mitochondria/ultrastructure , Neurons/physiology , Rats , Rats, Wistar
19.
Biol Res Nurs ; 6(3): 167-79, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15583357

ABSTRACT

Neurogenesis in the mammalian brain continues throughout adulthood. Several factors have been shown to influence neurogenesis, including experience in a complex environment (EC), exercise (EX), and ischemic insult. The authors investigated the effects of behavioral rehabilitation training following transient global cerebral ischemia on the number of new cells in the dentate gyrus that incorporated bromodeoxyuridine (BrdU), a thymidine analog that labels cells undergoing DNA replication. Seventy-two animals were included in the study, and 4-vessel occlusion was used to induce cerebral ischemia while control animals were subjected to anesthesia and sham surgery alone. Within 3 days of surgery, rats were randomly assigned to either EC, EX, or control (paired housing in standard laboratory conditions) groups. All animals were sacrificed 2 weeks after behavioral training. Immunohistochemistry results showed an increased number of BrdU-labeled cells in the subgranular zone of the dentate gyrus in all ischemic groups and in the EC and EX sham groups, although no significant group differences were seen. Examination of cell phenotype showed that almost all BrdU-positive cells colabeled with TuJ1, an immature neuron marker, in all animals whereas only a few BrdU-positive cells colabeled with NeuN, a mature neuron marker. BrdU/NeuN-labeled cells were seen only in the sham and ischemia EC groups. No new cells showed glial fibrillary acidic protein, astrocyte marker, colabeling. These results suggest that the adult brain has an inherent regenerative capacity after insult and that behavioral training following injury does not have an additive effect on neurogenesis. Finally, the enhanced maturation of BrdU-positive cells seen in the EC rats is probably modulated by environmental cues.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/physiopathology , Dentate Gyrus/physiology , Nerve Regeneration/physiology , Neurons/physiology , Stem Cells/physiology , Animals , Behavior, Animal , Brain Ischemia/rehabilitation , Cell Differentiation , Cell Division , Dentate Gyrus/cytology , Environment Design , Male , Rats , Rats, Wistar
20.
Brain Res ; 1018(1): 130-5, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15262214

ABSTRACT

Studies have demonstrated the effects of complex environment (EC) housing on brain plasticity both during postnatal development and in adulthood, but it is not clear how long these plastic changes persist nor what happens when environmental exposure is discontinued. Here we examined layer IV in the visual cortex of adult male rats for the: (1) effects of EC housing on synaptic plasticity, and (2) persistence of the synaptic changes after withdrawal from the complex environment. Fifty-eight adult male Long Evans rats were assigned to either: EC, socially paired housing (SC), or individual housing (IC). These rats remained in their assigned environment for 30 days. After 30 days, all rats in SC and some animals from the EC and IC groups were removed and perfused. The remaining animals in EC were then assigned to either remain in EC (ECEC) or be subsequently housed in IC (ECIC) for another 30 days. Similarly, rats in the IC group either remained in IC (ICIC) or were subsequently housed in EC (ICEC) for another 30 days. Electron microscopy results showed that all rats exposed to EC had significantly more synapses/neuron compared to SC, IC, and ICIC animals. Longer exposure to EC (ECEC) did not result in statistically more synapses per neuron; however, decreased neuron volume was seen. EC-induced synaptic changes persisted for an additional 30 days after withdrawal from EC (ECIC) confirming that EC-induced plastic changes occur in the brain regardless of age and indicating that once changes occur they tend to persist.


Subject(s)
Environment , Neural Pathways/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Synaptic Transmission/physiology , Visual Cortex/physiology , Animals , Cell Count , Cell Differentiation/physiology , Housing, Animal , Male , Microscopy, Electron , Neural Pathways/growth & development , Neural Pathways/ultrastructure , Photic Stimulation , Rats , Rats, Long-Evans , Synapses/ultrastructure , Visual Cortex/growth & development , Visual Cortex/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...