Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Behav Brain Res ; 437: 114106, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36089100

ABSTRACT

Approximately 60-70 million people suffer from traumatic brain injury (TBI) each year. Animal models continue to be paramount in understanding mechanisms of cellular dysfunction and testing new treatments for TBI. Enhancing the translational potential of novel interventions therefore necessitates testing pre-clinical intervention strategies with clinically relevant cognitive assays. This study used a unilateral parietal lobe controlled cortical impact (CCI) model of TBI and tested rats on a touchscreen-based Paired Associates Learning (PAL) task, which is part of the Cambridge Neuropsychological Test Automated Battery. In humans, the PAL task has been used to assess cognitive deficits in the ability to form stimulus-location associations in a multitude of disease states, including TBI. Although the use of PAL in animal models could be important for understanding the clinical severity of cognitive impairment post-injury and throughout intervention, to date, the extent to which a rat model of TBI produces deficits in PAL task performance has not yet been reported. This study details the behavioral consequences of the CCI injury model with a Trial-by-Trial analysis of PAL performance that enables behavioral strategy use to be inferred. Following behavior, the extent of the injury was quantified with histology and staining for the presence of glial fibrillary acid protein and ionized calcium-binding adapter molecule 1. Rats that received unilateral CCI were impaired on the PAL task and showed more aberrant response-driven behavior. The magnitude of PAL impairment was also correlated with Iba1 staining in the thalamus. These observations suggest that PAL could be useful for pre-clinical assessments of novel interventions for treating TBI.


Subject(s)
Brain Injuries, Traumatic , Cognition Disorders , Animals , Rats , Brain Injuries, Traumatic/complications , Cognition Disorders/pathology , Disease Models, Animal , Neuropsychological Tests , Paired-Associate Learning , Parietal Lobe/pathology
2.
Nutrients ; 14(19)2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36235630

ABSTRACT

Declining health, gut dysbiosis, and cognitive impairments are hallmarks of advanced age. While caloric restriction is known to robustly extend the healthspan and alter gut microbiome composition, it is difficult maintain. Time-restricted feeding or changes in dietary macronutrient composition could be feasible alternatives for enhancing late life cognitive and physical health that are easier to comply with for extended periods of time. To investigate this possibility, 8-month-old rats were placed on time-restricted feeding with a ketogenic or micronutrient- and calorically matched control diet for 13 months. A third group of rats was permitted to eat standard chow ad libitum during this time. At 22 months, all rats were tested on a biconditional association task and fecal samples were collected for microbiome composition analysis. Regardless of dietary composition, time-restricted-fed rats had better cognitive performance than ad libitum-fed rats. This observation could not be accounted for by differences in motivation, procedural or sensorimotor impairments. Additionally, there were significant differences in gut microbiome diversity and composition between all diet conditions. Allobaculum abundance was associated with cognitive task performance, indicating a link between gut health and cognitive outcomes in aged subjects. Overall, time restricted feeding had the largest influence on cognitive performance in aged rats.


Subject(s)
Fasting , Gastrointestinal Microbiome , Animals , Cognition , Micronutrients , Nutrients , Rats
3.
Front Aging Neurosci ; 12: 276, 2020.
Article in English | MEDLINE | ID: mdl-32982717

ABSTRACT

Introduction: Dual-task walking is common in daily life but becomes more difficult with aging. Little is known about the neurobiological mechanisms affecting competing cognitive demands. Translational studies with human and animal models are needed to address this gap. This pilot study investigated the feasibility of implementing a novel cross-species dual-task model in humans and rats and aimed to establish preliminary evidence that the model induces a dual-task cost. Methods: Young and older humans and rats performed an object discrimination task (OD), a baseline task of typical walking (baseline), an alternation turning task on a Figure 8 walking course (Alt), and a dual-task combining object discrimination with the alternation task (AltOD). Primary behavioral assessments including walking speed and correct selections for object discrimination and turning direction. In humans, left prefrontal cortex activity was measured with functional near-infrared spectroscopy (fNIRS). Results: Human subjects generally performed well on all tasks, but the older adults exhibited a trend for a slowing of walking speed immediately before the turning decision for Alt and AltOD compared to baseline. Older adults also had heightened prefrontal activity relative to young adults for the Alt and AltOD tasks. Older rodents required more training than young rodents to learn the alternation task. When tested on AltOD with and without a 15-s delay between trials, older rodents exhibited a substantial performance deficit for the delayed version on the initial day of testing. Old rats, however, did not show a significant slowing in walking speed with increasing task demand, as was evident in the young rats. Discussion: This study demonstrates the feasibility and challenges associated with implementing a cross-species dual-task model. While there was preliminary evidence of dual-task cost in both humans and rats, the magnitude of effects was small and not consistent across species. This is likely due to the relative ease of each task in humans and the walking component in rats not being sufficiently challenging. Future versions of this test should make the cognitive tasks more challenging and the motor task in rats more complex.

4.
Aging (Albany NY) ; 12(9): 7963-7984, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32369441

ABSTRACT

The ability to switch between glycolysis and ketosis promotes survival by enabling metabolism through fat oxidation during periods of fasting. Carbohydrate restriction or stress can also elicit metabolic switching. Keto-adapting from glycolysis is delayed in aged rats, but factors mediating this age-related impairment have not been identified. We measured metabolic switching between glycolysis and ketosis, as well as glycogen dynamics, in young and aged rats undergoing time-restricted feeding (TRF) with a standard diet or a low carbohydrate ketogenic diet (KD). TRF alone reversed markers of insulin-related metabolic deficits and accelerated metabolic switching in aged animals. A KD+TRF, however, provided additive benefits on these variables. Remarkably, the ability to keto-adapt was not related to glycogen levels and KD-fed rats showed an enhanced elevation in glucose following epinephrine administration. This study provides new insights into the mechanisms of keto-adaptation demonstrating the utility of dietary interventions to treat metabolic impairments across the lifespan.


Subject(s)
Adaptation, Physiological/physiology , Aging/metabolism , Glycogen/metabolism , Ketosis/metabolism , Animals , Blood Glucose/metabolism , Diet, Ketogenic , Disease Models, Animal , Glycolysis , Male , Rats , Rats, Inbred BN , Rats, Inbred F344
5.
Front Aging Neurosci ; 10: 391, 2018.
Article in English | MEDLINE | ID: mdl-30559660

ABSTRACT

Age-related cognitive decline has been linked to a diverse set of neurobiological mechanisms, including bidirectional changes in proteins critical for neuron function. Importantly, these alterations are not uniform across the brain. For example, the hippocampus (HPC) and prefrontal cortex (PFC) show distinct patterns of dysfunction in advanced age. Because higher cognitive functions require large-scale interactions across prefrontal cortical and hippocampal networks, selectively targeting an alteration within one region may not broadly restore function to improve cognition. One mechanism for decline that the PFC and HPC share, however, is a reduced ability to utilize glucose for energy metabolism. Although this suggests that therapeutic strategies bypassing the need for neuronal glycolysis may be beneficial for treating cognitive aging, this approach has not been empirically tested. Thus, the current study used a ketogenic diet (KD) as a global metabolic strategy for improving brain function in young and aged rats. After 12 weeks, rats were trained to perform a spatial alternation task through an asymmetrical maze, in which one arm was closed and the other was open. Both young and aged KD-fed rats showed resilience against the anxiogenic open arm, training to alternation criterion performance faster than control animals. Following alternation testing, rats were trained to perform a cognitive dual task that required working memory while simultaneously performing a bi-conditional association task (WM/BAT), which requires PFC-HPC interactions. All KD-fed rats also demonstrated improved performance on WM/BAT. At the completion of behavioral testing, tissue punches were collected from the PFC for biochemical analysis. KD-fed rats had biochemical alterations within PFC that were dissociable from previous results in the HPC. Specifically, MCT1 and MCT4, which transport ketone bodies, were significantly increased in KD-fed rats compared to controls. GLUT1, which transports glucose across the blood brain barrier, was decreased in KD-fed rats. Contrary to previous observations within the HPC, the vesicular glutamate transporter (VGLUT1) did not change with age or diet within the PFC. The vesicular GABA transporter (VGAT), however, was increased within PFC similar to HPC. These data suggest that KDs could be optimal for enhancing large-scale network function that is critical for higher cognition.

6.
Neurobiol Aging ; 70: 217-232, 2018 10.
Article in English | MEDLINE | ID: mdl-30031931

ABSTRACT

The link between age-related cellular changes within brain regions and larger scale neuronal ensemble dynamics critical for cognition has not been fully elucidated. The present study measured neuron activity within medial prefrontal cortex (PFC), perirhinal cortex (PER), and hippocampal subregion CA1 of young and aged rats by labeling expression of the immediate-early gene Arc. The proportion of cells expressing Arc was quantified at baseline and after a behavior that requires these regions. In addition, PER and CA1 projection neurons to PFC were identified with retrograde labeling. Within CA1, no age-related differences in neuronal activity were observed in the entire neuron population or within CA1 pyramidal cells that project to PFC. Although behavior was comparable across age groups, behaviorally driven Arc expression was higher in the deep layers of both PER and PFC and lower in the superficial layers of these regions. Moreover, age-related changes in activity levels were most evident within PER cells that project to PFC. These data suggest that the PER-PFC circuit is particularly vulnerable in advanced age.


Subject(s)
Aging/physiology , Association Learning/physiology , Neurons/physiology , Perirhinal Cortex/physiology , Prefrontal Cortex/physiology , Temporal Lobe/physiology , Animals , Behavior, Animal , CA1 Region, Hippocampal/physiology , Cytoskeletal Proteins/metabolism , Male , Nerve Tissue Proteins/metabolism , Neural Pathways/physiology , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...