Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
J Appl Physiol (1985) ; 136(3): 592-605, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38299221

Regular exercise has numerous health benefits, but the human population displays significant variability in exercise participation. Rodent models, such as voluntary wheel running (VWR) in rats, can provide insight into the underlying mechanisms of exercise behavior and its regulation. In this study, we focused on the role of estrogen on VWR in female rats. Female rats run more than males, and we aimed to determine to what extent running levels in females were regulated by estrogen signaling. The running behavior of rats (duration, speed, and total distance run) was measured under normal physiological conditions, ovariectomy (OVX), and estrogen replacement in an OVX background. Results show cyclic variations in running linked to the estrous cycle. Ovariectomy markedly reduced running and eliminated the cyclic pattern. Estrogen replacement through estradiol benzoate (EB) injections and osmotic minipumps reinstated running activity to pre-OVX levels and restored the cyclic pattern. Importantly, individual differences and ranking are preserved such that high versus low runners before OVX remain high and low runners after treatment. Further analysis revealed that individual variation in running distance was primarily caused by rats running different speeds, but rats also varied in running duration. However, it is noteworthy that this model also displays features distinct from estrogen-driven running behavior under physiological conditions, notably a delayed onset and a broader duration of running activity. Collectively, this estrogen causality VWR model presents a unique opportunity to investigate sex-specific mechanisms that control voluntary physical activity.NEW & NOTEWORTHY This study investigates estrogen's role in voluntary wheel running (VWR) behavior in female rats. Female rats exhibit greater running than males, with estrogen signaling regulating this activity. The estrous cycle influences running, whereas ovariectomy reduces it, and estrogen replacement restores it, maintaining individual differences under all conditions. Both running speed and duration contribute to VWR variations. These findings emphasize individual estrogen regulation in female exercise and provide an estrogen replacement animal model for investigating neurobiological underpinnings that drive voluntary exercise behavior.


Individuality , Motor Activity , Male , Humans , Rats , Animals , Female , Motor Activity/physiology , Estrogens/pharmacology , Estradiol/pharmacology , Ovariectomy
2.
Front Behav Neurosci ; 17: 1169151, 2023.
Article En | MEDLINE | ID: mdl-37273279

Introduction: Sedentary lifestyles have reached epidemic proportions world-wide. A growing body of literature suggests that exposures to adverse experiences (e.g., psychological traumas) are a significant risk factor for the development of physically inactive lifestyles. However, the biological mechanisms linking prior stress exposure and persistent deficits in physical activity engagement remains poorly understood. Methods: The purpose of this study was twofold. First, to identify acute stress intensity thresholds that elicit long-term wheel running deficits in rats. To that end, young adult male rats were exposed to a single episode of 0, 50, or 100 uncontrollable tail shocks and then given free access to running wheels for 9 weeks. Second, to identify stress-induced changes to central monoamine neurotransmitters and peripheral muscle physiology that may be maladaptive to exercise output. For this study, rats were either exposed to a single episode of uncontrollable tail shocks (stress) or left undisturbed in home cages (unstressed). Eight days later, monoamine-related neurochemicals were quantified by ultra-high performance liquid chromatography (UHPLC) across brain reward, motor, and emotion structures immediately following a bout of graded treadmill exercise controlled for duration and intensity. Additionally, protein markers of oxidative stress, inflammation, and metabolic activity were assessed in the gastrocnemius muscle by Western blot. Results: For experiment 1, stress exposure caused a shock number-dependent two to fourfold decrease in wheel running distance across the entire duration of the study. For experiment 2, stress exposure curbed an exercise-induced increase of dopamine (DA) turnover measures in the prefrontal cortex and hippocampus, and augmented serotonin (5HT) turnover in the hypothalamus and remaining cortical area. However, stress exposure also caused several monoaminergic changes independent of exercise that could underlie impaired motivation for physical activity, including a mild dopamine deficiency in the striatal area. Finally, stress potently increased HSP70 and lowered SOD2 protein concentrations in the gastrocnemius muscle, which may indicate prolonged oxidative stress. Discussion: These data support some of the possible central and peripheral mechanisms by which exposure to adverse experiences may chronically impair physical activity engagement.

3.
Nutr Neurosci ; 26(9): 875-887, 2023 Sep.
Article En | MEDLINE | ID: mdl-36125026

Background: Childhood malnutrition can have devastating consequences on health, behavior, and cognition. Edible insects are sustainable low cost high protein and iron nutritious foods that can prevent malnutrition. However, it is unclear whether insect-based diets may help prevent changes to brain neurochemistry associated with malnutrition.Materials and Methods: Weanling male Sprague-Dawley rats were malnourished by feeding a low protein-iron diet (LPI, 5% protein and ∼2 ppm Fe) for 3 weeks or nourished by feeding a sufficient protein-iron diet (SPI, 15% protein 20 ppm FeSO4) for the duration of the study. Following 3 weeks of LPI diet, three subsets of the malnourished rats were placed on repletion diets supplemented with cricket, palm weevil larvae, or the SPI diet for 2 weeks, while the remaining rats continued the LPI diet for an additional 2 weeks. Monoamine-related neurochemicals (e.g. serotonin (5-HT), dopamine (DA), norepinephrine) and select monoamine metabolites were measured in the hypothalamus, hippocampus, striatum, and prefrontal cortex using Ultra High-Performance Liquid Chromatography.Results: Five weeks of LPI diets disrupted brain monoamines, most notable in the hypothalamus. Two weeks supplementation with cricket and palm weevil larvae diets prevented changes to measures of 5-HT and DA turnover in the hippocampus and hypothalamus. Moreover, these insect diets prevented the malnutrition-induced imbalance of 5-HT and DA metabolites in the hippocampus, striatum, and hypothalamus.Conclusion: Edible insects such as cricket and palm weevil larvae could be sustainable nutrition intervention to prevent behavioral and cognitive impairment associated abnormal brain monoamine activities that results from early life malnutrition.


Edible Insects , Malnutrition , Rats , Animals , Male , Edible Insects/metabolism , Serotonin/metabolism , Rats, Sprague-Dawley , Brain/metabolism , Malnutrition/complications , Malnutrition/metabolism , Dopamine/metabolism , Norepinephrine/metabolism , Iron/metabolism
4.
Front Physiol ; 13: 1021985, 2022.
Article En | MEDLINE | ID: mdl-36582358

Stress-induced abnormalities in gut monoamine levels (e.g., serotonin, dopamine, norepinephrine) have been linked to gastrointestinal (GI) dysfunction, as well as the worsening of symptoms in GI disorders. However, the influence of stress on changes across the entire intestinal monoamine biogeography has not been well-characterized, especially in the days following stress exposure. Therefore, the aim of this study was to comprehensively assess changes to monoamine neurochemical signatures across the entire rat intestinal tract days after exposure to an acute stressor. To the end, adult male F344 rats were subjected to an episode of unpredictable tail shocks (acute stress) or left undisturbed. Forty-eight hours later rats were euthanized either following a 12 h period of fasting or 30 min of food access to evaluate neurochemical profiles during the peri- and early postprandial periods. Monoamine-related neurochemicals were measured via UHPLC in regions of the small intestine (duodenum, jejunum, ileum), large intestine (cecum, proximal colon, distal colon), cecal contents, fecal contents, and liver. The results suggest a relatively wide-spread increase in measures of serotonin activity across intestinal regions can be observed 48 h after exposure to acute stress, however some evidence was found supporting localized differences in serotonin metabolization. Moreover, acute stress exposure reduced catecholamine-related neurochemical concentrations most notably in the ileum, and to a lesser extent in the cecal contents. Next, stress-related fecal serotonin concentrations were consistent with intestinal profiles. However, fecal dopamine was elevated in association with stress, which did not parallel findings in any other intestinal area. Finally, stress exposure and the food access period together only had minor effects on intestinal monoamine profiles. Taken together, these data suggest nuanced differences in monoaminergic profiles exist across intestinal regions the days following exposure to an acute stressor, highlighting the importance of assessments that consider the entire intestinal tract biogeography when investigating stress-related biological outcomes that may be relevant to GI pathophysiology.

5.
Alcohol ; 104: 45-52, 2022 11.
Article En | MEDLINE | ID: mdl-35926812

Excessive ethanol ingestion can reduce skeletal muscle protein synthesis (MPS) through the disruption of signaling along the Akt-mTOR pathway and increase muscle protein degradation (MPD) through the Ubiquitin Proteasome Pathway (UPP) and autophagy. Identification of interventions that curb the disrupting effects of alcohol misuse on MPS and MPD are of central importance for the prevention of chronic health complications that arise from muscle loss. Physical activity is one potential strategy to combat the deleterious effects of alcohol on skeletal muscle. Therefore, the purpose of this study was to investigate the interaction between daily wheel running and binge-patterned ethanol consumption, through episodes of voluntary binge-patterned ethanol drinking, on signaling factors along the Akt-mTOR, Ubiquitin-Proteasome, and autophagy pathways. Adult female C57BL/6J mice received daily access to cages with or without running wheels for 2.5 h/day for five weeks. During the final five days of the study, mice received 2-4 h of daily access to sipper tubes containing water (n = 14 sedentary; n = 15 running) or 20% ethanol (n = 14 sedentary; n = 16 running) 30 min after running wheel access, using the "Drinking in the Dark" (DID) model of binge-patterned ethanol consumption. Immediately after the final episode of DID, gastrocnemius muscle was extracted. Western blotting was performed to measure proteins along Akt-mTOR, Ubiquitin-Proteasome, and autophagy pathways, and PCR was used to assess mRNA expression of atrogenes. Ethanol access increased expression of MAFbx by 82% (p = 0.048), but did not robustly influence Akt-mTOR or UPP signaling. Daily wheel access did not prevent alcohol-induced MAFbx expression; however, ethanol access decreased the phosphorylation of p70S6K by 45% in running mice (p = 0.020). These results suggest that physical activity may be insufficient to prevent alcohol-induced changes to signaling factors along pathways involved in muscle loss. Instead, binge-patterned ethanol ingestion may impair the benefits of physical activity on factors involved in MPS.


Muscle Proteins , Proteasome Endopeptidase Complex , Mice , Female , Animals , Muscle Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Motor Activity/physiology , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism , Ethanol/metabolism , Muscle, Skeletal/metabolism , Eating , Ubiquitins/metabolism
7.
J Nutr ; 151(12): 3617-3627, 2021 12 03.
Article En | MEDLINE | ID: mdl-34522956

BACKGROUND: Adverse life experiences are a major risk factor for anorexia nervosa (AN). Eating-provoked anxiousness associated with AN is postulated to be due to food-related exaggerated serotonin activity in the brain and imbalances of monoamine neurotransmitters. OBJECTIVES: Using a rodent model of stress-induced hypophagia, we investigated if stress exposure augments food-related serotonin turnover and imbalances in measures of brain serotonin and dopamine activity in manners consistent with anxiousness toward food and restricted eating. METHODS: Adult male F344 rats were conditioned to associate an audio cue with daily food over 2 weeks, after which half of the rats were exposed to a single episode of tail shocks (stress) or left undisturbed (nonstressed). All rats were killed 48 h later, during a control period, the food-associated cue, or a period of food access. Serotonin, dopamine, and norepinephrine, as well as metabolite concentrations, were assessed across brain regions comprising reward, emotion, and feeding circuits relevant to AN in acutely stressed and nonstressed rats using HPLC. Statistical significance level was 5%. RESULTS: Stress-induced rat hypophagia paralleled an augmented serotonin turnover in response to the food-associated cue in the hypothalamus and hippocampus, as well as food access in the hypothalamus and cortical areas (all P < 0.05). Stress exposure increased the ratio of serotonin to dopamine metabolites across several brain areas, but the magnitude of this imbalance was further augmented during the food-associated cue and food access in the brainstem, hippocampus, and cortical areas (all P < 0.05). Finally, stress lowered norepinephrine concentrations by 18% in the hypothalamus (P < 0.05). CONCLUSIONS: The observed stress-induced changes to monoamine profiles in rats could have key implications for physiological states that contribute to restricted eating and may hold relevance for the development of AN precipitated by adverse life experiences.


Anorexia , Serotonin , Animals , Anorexia/etiology , Brain , Dopamine , Male , Norepinephrine , Rats , Rats, Inbred F344
8.
Front Behav Neurosci ; 15: 639790, 2021.
Article En | MEDLINE | ID: mdl-33716684

Monoamine neurotransmitter activity in brain reward, limbic, and motor areas play key roles in the motivation to misuse alcohol and can become modified by exercise in a manner that may affect alcohol craving. This study investigated the influence of daily moderate physical activity on monoamine-related neurochemical concentrations across the mouse brain in response to high volume ethanol ingestion. Adult female C57BL/6J mice were housed with or without 2.5 h of daily access to running wheels for 30 days. On the last 5 days, mice participated in the voluntary binge-like ethanol drinking procedure, "Drinking in the dark" (DID). Mice were sampled immediately following the final episode of DID. Monoamine-related neurochemical concentrations were measured across brain regions comprising reward, limbic, and motor circuits using ultra High-Performance Liquid Chromatography (UHPLC). The results suggest that physical activity status did not influence ethanol ingestion during DID. Moreover, daily running wheel access only mildly influenced alcohol-related norepinephrine concentrations in the hypothalamus and prefrontal cortex, as well as serotonin turnover in the hippocampus. However, access to alcohol during DID eliminated wheel running-related decreases of norepinephrine, serotonin, and 5-HIAA content in the hypothalamus, but also to a lesser extent for norepinephrine in the hippocampus and caudal cortical areas. Finally, alcohol access increased serotonin and dopamine-related neurochemical turnover in the striatum and brainstem areas, regardless of physical activity status. Together, these data provide a relatively thorough assessment of monoamine-related neurochemical levels across the brain in response to voluntary binge-patterned ethanol drinking, but also adds to a growing body of research questioning the utility of moderate physical activity as an intervention to curb alcohol abuse.

9.
Alcohol Clin Exp Res ; 45(5): 996-1012, 2021 05.
Article En | MEDLINE | ID: mdl-33704774

BACKGROUND: Altered monoamine (i.e., serotonin, dopamine, and norepinephrine) activity following episodes of alcohol abuse plays key roles not only in the motivation to ingest ethanol, but also physiological dysfunction related to its misuse. Although monoamine activity is essential for physiological processes that require coordinated communication across the gut-brain axis (GBA), relatively little is known about how alcohol misuse may affect monoamine levels across the GBA. Therefore, we evaluated monoamine activity across the mouse gut and brain following episodes of binge-patterned ethanol drinking. METHODS: Monoamine and select metabolite neurochemical concentrations were analyzed by ultra-high-performance liquid chromatography in gut and brain regions of female and male C57BL/6J mice following "Drinking in the Dark" (DID), a binge-patterned ethanol ingestion paradigm. RESULTS: First, we found that alcohol access had an overall small effect on gut monoamine-related neurochemical concentrations, primarily influencing dopamine activity. Second, neurochemical patterns between the small intestine and the striatum were correlated, adding to recent evidence of modulatory activity between these areas. Third, although alcohol access robustly influenced activity in brain areas in the mesolimbic dopamine system, binge exposure also influenced monoaminergic activity in the hypothalamic region. Finally, sex differences were observed in the concentrations of neurochemicals within the gut, which was particularly pronounced in the small intestine. CONCLUSION: Together, these data provide insights into the influence of alcohol abuse and biological sex on monoamine-related neurochemical changes across the GBA, which could have important implications for GBA function and dysfunction.


Binge Drinking/metabolism , Brain-Gut Axis/drug effects , Brain/drug effects , Central Nervous System Depressants/pharmacology , Dopamine/metabolism , Ethanol/pharmacology , Intestine, Small/drug effects , Norepinephrine/metabolism , Serotonin/metabolism , Animals , Brain/metabolism , Cecum/drug effects , Cecum/metabolism , Chromatography, High Pressure Liquid , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Intestine, Small/metabolism , Limbic System/drug effects , Limbic System/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Neostriatum/drug effects , Neostriatum/metabolism , Sex Factors
10.
Neural Plast ; 2020: 5859098, 2020.
Article En | MEDLINE | ID: mdl-32399024

Adenosine acts as a key regulator of striatum activity, in part, through the antagonistic modulation of dopamine activity. Exercise can increase adenosine activity in the brain, which may impair dopaminergic functions in the striatum. Therefore, long-term repeated bouts of exercise may subsequently generate plasticity in striatal adenosine systems in a manner that promotes dopaminergic activity. This study investigated the effects of long-term voluntary wheel running on adenosine 1 (A1R), adenosine 2A (A2AR), dopamine 1 (D1R), and dopamine 2 (D2R) receptor protein expression in adult mouse dorsal and ventral striatum structures using immunohistochemistry. In addition, equilibrative nucleoside transporter 1 (ENT1) protein expression was examined after wheel running, as ENT1 regulates the bidirectional flux of adenosine between intra- and extracellular space. The results suggest that eight weeks of running wheel access spared age-related increases of A1R and A2AR protein concentrations across the dorsal and ventral striatal structures. Wheel running mildly reduced ENT1 protein levels in ventral striatum subregions. Moreover, wheel running mildly increased D2R protein density within striatal subregions in the dorsal medial striatum, nucleus accumbens core, and the nucleus accumbens shell. However, D1R protein expression in the striatum was unchanged by wheel running. These data suggest that exercise promotes adaptations to striatal adenosine systems. Exercise-reduced A1R and A2AR and exercise-increased D2R protein levels may contribute to improved dopaminergic signaling in the striatum. These findings may have implications for cognitive and behavioral processes, as well as motor and psychiatric diseases that involve the striatum.


Corpus Striatum/metabolism , Physical Conditioning, Animal/physiology , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Purinergic P1/metabolism , Animals , Equilibrative Nucleoside Transporter 1/metabolism , Male , Mice, Inbred C57BL , Motor Activity
12.
Eur J Neurosci ; 43(9): 1190-202, 2016 05.
Article En | MEDLINE | ID: mdl-26833814

Brain reward circuits are implicated in stress-related psychiatric disorders. Exercise reduces the incidence of stress-related disorders, but the contribution of exercise reward to stress resistance is unknown. Exercise-induced stress resistance is independent of exercise controllability; both voluntary running (VR) and forced running (FR) protect rats against the anxiety-like and depression-like behavioural consequences of stress. Voluntary exercise is a natural reward, but whether rats find FR rewarding is unknown. Moreover, the contribution of dopamine (DA) and striatal reward circuits to exercise reward is not well characterized. Adult, male rats were assigned to locked wheels, VR, or FR groups. FR rats were forced to run in a pattern resembling the natural wheel running behavior of rats. Both VR and FR increased the reward-related plasticity marker ΔFosB in the dorsal striatum and nucleus accumbens, and increased the activity of DA neurons in the lateral ventral tegmental area, as revealed by immunohistochemistry for tyrosine hydroxylase and pCREB. Both VR and FR rats developed conditioned place preference (CPP) to the side of a CPP chamber paired with exercise. Re-exposure to the exercise-paired side of the CPP chamber elicited conditioned increases in cfos mRNA in direct-pathway (dynorphin-positive) neurons in the dorsal striatum and nucleus accumbens in both VR and FR rats, and in tyrosine hydroxylase-positive neurons in the lateral ventral tegmental area of VR rats only. The results suggest that the rewarding effects of exercise are independent of exercise controllability and provide insight into the DA and striatal circuitries involved in exercise reward and exercise-induced stress resistance.


Physical Conditioning, Animal , Reward , Stress, Psychological/physiopathology , Animals , Corpus Striatum/cytology , Corpus Striatum/metabolism , Corpus Striatum/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Male , Neuronal Plasticity , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Inbred F344 , Running , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
13.
PLoS One ; 10(11): e0141898, 2015.
Article En | MEDLINE | ID: mdl-26555633

Accumulating evidence from both the human and animal literature indicates that exercise reduces the negative consequences of stress. The neurobiological etiology for this stress protection, however, is not completely understood. Our lab reported that voluntary wheel running protects rats from expressing depression-like instrumental learning deficits on the shuttle box escape task after exposure to unpredictable and inescapable tail shocks (uncontrollable stress). Impaired escape behavior is a result of stress-sensitized serotonin (5-HT) neuron activity in the dorsal raphe (DRN) and subsequent excessive release of 5-HT into the dorsal striatum following exposure to a comparatively mild stressor. However, the possible mechanisms by which exercise prevents stress-induced escape deficits are not well characterized. The purpose of this experiment was to test the hypothesis that exercise blunts the stress-evoked release of 5-HT in the dorsal striatum. Changes to dopamine (DA) levels were also examined, since striatal DA signaling is critical for instrumental learning and can be influenced by changes to 5-HT activity. Adult male F344 rats, housed with or without running wheels for 6 weeks, were either exposed to tail shock or remained undisturbed in laboratory cages. Twenty-four hours later, microdialysis was performed in the medial (DMS) and lateral (DLS) dorsal striatum to collect extracellular 5-HT and DA before, during, and following 2 mild foot shocks. We report wheel running prevents foot shock-induced elevation of extracellular 5-HT and potentiates DA concentrations in both the DMS and DLS approximately 24 h following exposure to uncontrollable stress. These data may provide a possible mechanism by which exercise prevents depression-like instrumental learning deficits following exposure to acute stress.


Corpus Striatum/metabolism , Dopamine/metabolism , Physical Conditioning, Animal/physiology , Running/physiology , Serotonin/metabolism , Stress, Psychological/metabolism , Animals , Electroshock , Helplessness, Learned , Male , Microdialysis , Motor Activity/physiology , Neurons/metabolism , Rats , Rats, Inbred F344
14.
Behav Brain Res ; 272: 252-63, 2014 Oct 01.
Article En | MEDLINE | ID: mdl-25017571

Emerging evidence indicates that adenosine is a major regulator of striatum activity, in part, through the antagonistic modulation of dopaminergic function. Exercise can influence adenosine and dopamine activity, which may subsequently promote plasticity in striatum adenosine and dopamine systems. Such changes could alter activity of medium spiny neurons and impact striatum function. The purpose of this study was twofold. The first was to characterize the effect of long-term wheel running on adenosine 1 (A1R), adenosine 2A (A2AR), dopamine 1 (D1R), and dopamine 2 (D2R) receptor mRNA expression in adult rat dorsal and ventral striatum structures using in situ hybridization. The second was to determine if changes to adenosine and dopamine receptor mRNA from running are associated with altered cfos mRNA induction in dynorphin- (direct pathway) and enkephalin- (indirect pathway) expressing neurons of the dorsal striatum following stress exposure. We report that chronic running, as well as acute uncontrollable stress, reduced A1R and A2AR mRNA levels in the dorsal and ventral striatum. Running also modestly elevated D2R mRNA levels in striatum regions. Finally, stress-induced cfos was potentiated in dynorphin and attenuated in enkephalin expressing neurons of running rats. These data suggest striatum adenosine and dopamine systems are targets for neuroplasticity from exercise, which may contribute to changes in direct and indirect pathway activity. These findings may have implications for striatum mediated motor and cognitive processes, as well as exercise facilitated stress-resistance.


Corpus Striatum/physiopathology , Motor Activity/physiology , Proto-Oncogene Proteins c-fos/metabolism , Running/physiology , Stress, Psychological/physiopathology , Animals , Corticosterone/blood , Dynorphins/metabolism , Electroshock , Enkephalins/metabolism , Gene Expression/physiology , Male , Motor Activity/drug effects , Neurons/physiology , RNA, Messenger/metabolism , Random Allocation , Rats, Inbred F344 , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Stress, Psychological/therapy
15.
Eur J Neurosci ; 37(3): 469-78, 2013 Feb.
Article En | MEDLINE | ID: mdl-23121339

Exercise increases resistance against stress-related disorders such as anxiety and depression. Similarly, the perception of control is a powerful predictor of neurochemical and behavioral responses to stress, but whether the experience of choosing to exercise, and exerting control over that exercise, is a critical factor in producing exercise-induced stress resistance is unknown. The current studies investigated whether the protective effects of exercise against the anxiety- and depression-like consequences of stress are dependent on exercise controllability and a brain region implicated in the protective effects of controllable experiences, the medial prefrontal cortex. Adult male Fischer 344 rats remained sedentary, were forced to run on treadmills or motorised running wheels, or had voluntary access to wheels for 6 weeks. Three weeks after exercise onset, rats received sham surgery or excitotoxic lesions of the medial prefrontal cortex. Rats were exposed to home cage or uncontrollable tail shock treatment three weeks later. Shock-elicited fear conditioning and shuttle box escape testing occurred the next day. Both forced and voluntary wheel running, but not treadmill training, prevented the exaggerated fear conditioning and interference with escape learning produced by uncontrollable stress. Lesions of the medial prefrontal cortex failed to eliminate the protective effects of forced or voluntary wheel running. These data suggest that exercise controllability and the medial prefrontal cortex are not critical factors in conferring the protective effects of exercise against the affective consequences of stressor exposure, and imply that exercise perceived as forced may still benefit affect and mental health.


Physical Exertion , Prefrontal Cortex/physiopathology , Stress, Physiological , Stress, Psychological/physiopathology , Animals , Anxiety/physiopathology , Conditioning, Classical , Depressive Disorder/physiopathology , Electroshock , Male , Rats , Rats, Inbred F344
16.
PLoS One ; 7(9): e46118, 2012.
Article En | MEDLINE | ID: mdl-23049953

Physical activity reduces the incidence and severity of psychiatric disorders such as anxiety and depression. Similarly, voluntary wheel running produces anxiolytic- and antidepressant-like effects in rodent models. The specific neurobiological mechanisms underlying the beneficial properties of exercise, however, remain unclear. One relevant pharmacological target in the treatment of psychiatric disorders is the 5-HT(2C) receptor (5-HT(2C)R). Consistent with data demonstrating the anxiogenic consequences of 5-HT(2C)R activation in humans and rodents, we have previously reported that site-specific administration of the selective 5-HT(2C)R agonist CP-809101 in the lateral/basolateral amygdala (BLA) increases shock-elicited fear while administration of CP-809101 in the dorsal striatum (DS) interferes with shuttle box escape learning. These findings suggest that activation of 5-HT(2C)R in discrete brain regions contributes to specific anxiety- and depression-like behaviors and may indicate potential brain sites involved in the anxiolytic and antidepressant effects of exercise. The current studies tested the hypothesis that voluntary wheel running reduces the behavioral consequences of 5-HT(2C)R activation in the BLA and DS, specifically enhanced shock-elicited fear and interference with shuttle box escape learning. After 6 weeks of voluntary wheel running or sedentary conditions, the selective 5-HT(2C)R agonist CP-809101 was microinjected into either the BLA or the DS of adult Fischer 344 rats, and shock-elicited fear and shuttle box escape learning was assessed. Additionally, in-situ hybridization was used to determine if 6 weeks of voluntary exercise changed levels of 5-HT(2C)R mRNA. We found that voluntary wheel running reduced the behavioral effects of CP-809101 and reduced levels of 5-HT(2C)R mRNA in both the BLA and the DS. The current data indicate that expression of 5-HT(2C)R mRNA in discrete brain sites is sensitive to physical activity status of the organism, and implicates the 5-HT(2C)R as a target for the beneficial effects of physical activity on mental health.


Amygdala/metabolism , Anxiety/metabolism , Brain/metabolism , Brain/physiology , Depression/metabolism , Physical Conditioning, Animal/physiology , Receptor, Serotonin, 5-HT2C/metabolism , Amygdala/physiology , Animals , In Situ Hybridization , Male , Random Allocation , Rats , Receptor, Serotonin, 5-HT2C/genetics
17.
Hippocampus ; 22(9): 1860-7, 2012 Sep.
Article En | MEDLINE | ID: mdl-22467337

Running increases the formation of new neurons in the adult rodent hippocampus. However, the function of new neurons generated from running is currently unknown. One hypothesis is that new neurons from running contribute to enhanced cognitive function by increasing plasticity in the adult hippocampus. An alternative hypothesis is that new neurons generated from running incorporate into experience-specific hippocampal networks that only become active during running. The purpose of this experiment was to determine if new neurons generated from running are selectively activated by running, or can become recruited into granule cell activity occurring during performance on other behavioral tasks that engage the hippocampus. Therefore, the activation of new 5-6 week neurons was detected using BrdU, NeuN, and Zif268 triple-label immunohistochemistry in cohorts of female running and sedentary adult C57BL/6J mice following participation in one of three different tasks: the Morris water maze, novel environment exploration, or wheel running. Results showed that running and sedentary mice displayed a nearly equivalent proportion of new neurons that expressed Zif268 following each task. Since running approximately doubled the number of new neurons, the results demonstrated that running mice had a greater number of new neurons recruited into the Zif268 induction in the granule cell layer following each task than sedentary mice. The results suggest that new neurons incorporated into hippocampal circuitry from running are not just activated by wheel running itself, but rather become broadly recruited into granule cell layer activity during distinct behavioral experiences.


Hippocampus/physiology , Neurogenesis/physiology , Running/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Survival , DNA-Binding Proteins , Early Growth Response Protein 1/metabolism , Exploratory Behavior/physiology , Female , Hippocampus/cytology , Immunohistochemistry , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Models, Neurological , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nuclear Proteins/metabolism
18.
Behav Brain Res ; 226(1): 1-7, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-21896289

Exercise improves performance on a number of hippocampus involved cognitive tasks including contextual fear conditioning, but whether exercise enhances contextual fear when the retention interval is longer than 1 day is not known. Also unknown is whether exercise improves trace conditioning, a task that requires the hippocampus to bridge the time interval between stimuli. Hence, 4-month-old male C57BL/6J mice were housed with or without running wheels. To assess whether hippocampal neurogenesis was associated with behavioral outcomes, during the initial 10 days, mice received Bromodeoxyuridine to label dividing cells. After 30 days, one group of mice was trained in a contextual fear conditioning task. Freezing to context was assessed 1, 7, or 21 days post-training. A separate group was trained on a trace procedure, in which a tone and footshock were separated by a 15, 30, or 45s interval. Freezing to the tone was measured 24h later in a novel environment, and freezing to the training context was measured 48h later. Running enhanced freezing to context when the retention interval was 1, but not 7 or 21 days. Running had no effect on trace conditioning even though runners displayed enhanced freezing to the training context 48h later. Wheel running increased survival of new neurons in the hippocampus. Collectively, findings indicate that wheel running enhances cognitive performance on some tasks but not others and that enhanced neurogenesis is not always associated with improved performance on hippocampus tasks, one example of which is trace conditioning.


Conditioning, Psychological/physiology , Fear/physiology , Hippocampus/physiology , Neurogenesis/physiology , Physical Conditioning, Animal/physiology , Animals , Freezing Reaction, Cataleptic/physiology , Male , Mice , Neurons/physiology
19.
Alcohol ; 45(7): 631-9, 2011 Nov.
Article En | MEDLINE | ID: mdl-21803530

Individual differences in dopamine D2 receptor (D2R) expression in the brain are thought to influence motivation and reinforcement for ethanol and other rewards. D2R exists in two isoforms, D2 long (D2LR) and D2 short (D2SR), produced by alternative splicing of the same gene. The relative contributions of D2LR versus D2SR to ethanol and sugar water drinking are not known. Genetic engineering was used to produce a line of knockout (KO) mice that lack D2LR and consequently have increased expression of D2SR. KO and wild-type (WT) mice of both sexes were tested for intake of 20% ethanol, 10% sugar water and plain tap water using established drinking-in-the-dark procedures. Mice were also tested for effects of the D2 antagonist eticlopride on intake of ethanol to determine whether KO responses were caused by lack of D2LR or overrepresentation of D2SR. Locomotor activity on running wheels and in cages without wheels was also measured for comparison. D2L KO mice drank significantly more ethanol than WT in both sexes. KO mice drank more sugar water than WT in females but not in males. Eticlopride dose dependently decreased ethanol intake in all groups except male KO. KO mice were less physically active than WT in cages with or without running wheels. Results suggest that overrepresentation of D2SR contributes to increased intake of ethanol in the KO mice. Decreasing wheel running and general levels of physical activity in the KO mice rules out the possibility that higher intake results from higher motor activity. Results extend the literature implicating altered expression of D2R in risk for addiction by delineating the contribution of individual D2R isoforms. These findings suggest that D2LR and D2SR play differential roles in consumption of alcohol and sugar rewards.


Carbohydrates/administration & dosage , Ethanol/administration & dosage , Receptors, Dopamine D2/deficiency , Alcoholism/genetics , Alternative Splicing , Animals , Dopamine Antagonists/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/physiology , Salicylamides/pharmacology , Solutions , Water
20.
Behav Brain Res ; 213(2): 246-52, 2010 Dec 01.
Article En | MEDLINE | ID: mdl-20472002

Voluntary wheel running activates dentate gyrus granule neurons and increases adult hippocampal neurogenesis. Average daily running distance typically increases over a period of 3 weeks in rodents. Whether neurogenesis and cell activation are greater at the peak of running as compared to the initial escalation period is not known. Therefore, adult C57BL/6J male mice received 5 days of BrdU injections, at the same age, to label dividing cells during the onset of wheel access or after 21 days during peak levels of running or in sedentary conditions. Mice were sampled either 24h or 25 days after the last BrdU injection to measure cell proliferation and survival, respectively. Immunohistochemistry was performed on brain sections to identify the numbers of proliferating BrdU-labeled cells, and new neurons (BrdU/NeuN co-labeled) in the dentate gyrus. Ki67 was used as an additional mitotic marker. The induction of c-Fos was used to identify neurons activated from running. Mice ran approximately half as far during the first 5 days as compared to after 21 days. Running increased Ki67 cells at the onset but after 21 days levels were similar to sedentary. Numbers of BrdU cells were similar in all groups 24h after the final injection. However, after 25 days, running approximately doubled the survival of new neurons born either at the onset or peak of running. These changes co-varied with c-Fos expression. We conclude that sustained running maintains a stable rate of neurogenesis above sedentary via activity-dependent increases in differentiation and survival, not proliferation, of progenitor cells in the C57BL/6J model.


Hippocampus/physiology , Mice, Inbred C57BL/physiology , Motor Activity/physiology , Neurogenesis/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Cell Proliferation , Cell Survival/physiology , Male , Mice , Time Factors
...