Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Neurobiol Learn Mem ; 212: 107937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735637

ABSTRACT

Systemic manipulations that enhance dopamine (DA) transmission around the time of fear extinction can strengthen fear extinction and reduce conditioned fear relapse. Prior studies investigating the brain regions where DA augments fear extinction focus on targets of mesolimbic and mesocortical DA systems originating in the ventral tegmental area, given the role of these DA neurons in prediction error. The dorsal striatum (DS), a primary target of the nigrostriatal DA system originating in the substantia nigra (SN), is implicated in behaviors beyond its canonical role in movement, such as reward and punishment, goal-directed action, and stimulus-response associations, but whether DS DA contributes to fear extinction is unknown. We have observed that chemogenetic stimulation of SN DA neurons during fear extinction prevents the return of fear in contexts different from the extinction context, a form of relapse called renewal. This effect of SN DA stimulation is mimicked by a DA D1 receptor (D1R) agonist injected into the DS, thus implicating DS DA in fear extinction. Different DS subregions subserve unique functions of the DS, but it is unclear where in the DS D1R agonist acts during fear extinction to reduce renewal. Furthermore, although fear extinction increases neural activity in DS subregions, whether neural activity in DS subregions is causally involved in fear extinction is unknown. To explore the role of DS subregions in fear extinction, adult, male Long-Evans rats received microinjections of either the D1R agonist SKF38393 or a cocktail consisting of GABAA/GABAB receptor agonists muscimol/baclofen selectively into either dorsomedial (DMS) or dorsolateral (DLS) DS subregions immediately prior to fear extinction, and extinction retention and renewal were subsequently assessed drug-free. While increasing D1R signaling in the DMS during fear extinction did not impact fear extinction retention or renewal, DMS inactivation reduced later renewal. In contrast, DLS inactivation had no effect on fear extinction retention or renewal but increasing D1R signaling in the DLS during extinction reduced fear renewal. These data suggest that DMS and DLS activity during fear extinction can have opposing effects on later fear renewal, with the DMS promoting renewal and the DLS opposing renewal. Mechanisms through which the DS could influence the contextual gating of fear extinction are discussed.


Subject(s)
Corpus Striatum , Extinction, Psychological , Fear , Receptors, Dopamine D1 , Animals , Fear/physiology , Fear/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Rats , Corpus Striatum/drug effects , Corpus Striatum/physiology , Corpus Striatum/metabolism , Receptors, Dopamine D1/physiology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Dopamine Agonists/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Substantia Nigra/drug effects , Substantia Nigra/physiology , Rats, Long-Evans , Dopamine/metabolism , Dopamine/physiology
2.
Stress ; 26(1): 2245492, 2023 11.
Article in English | MEDLINE | ID: mdl-37549016

ABSTRACT

Common stress-related mental health disorders affect women more than men. Physical activity can provide protection against the development of future stress-related mental health disorders (i.e. stress resistance) in both sexes, but whether there are sex differences in exercise-induced stress resistance is unknown. We have previously observed that voluntary wheel running (VWR) protects both female and male rats against the anxiety- and exaggerated fear-like behavioral effects of inescapable stress, but the time-course and magnitude of VWR-induced stress resilience has not been compared between sexes. The goal of the current study was to determine whether there are sex differences in the time-course and magnitude of exercise-induced stress resistance. In adult female and male Sprague Dawley rats, 6 weeks of VWR produced robust protection against stress-induced social avoidance and exaggerated fear. The magnitude of stress protection was similar between the sexes and was independent of reactivity to shock, general locomotor activity, and circulating corticosterone. Interestingly, 3 weeks of VWR prevented both stress-induced social avoidance and exaggerated fear in females but only prevented stress-induced social avoidance in males. Ovariectomy altered wheel-running behavior in females such that it resembled that of males, however; 3 weeks of VWR still protected females against behavioral consequences of stress regardless of the absence of ovaries. These data indicate that female Sprague Dawley rats are more responsive to exercise-induced stress resistance than are males.


The duration of wheel running required to enable stress resistance differs between the sexes in a behavior-dependent manner.Wheel running enables rapid protection against stress-induced social avoidance in both male and female Sprague Dawley rats.Wheel running enables protection against stress-induced exaggerated fear more readily in female Sprague Dawley rats compared to males.Ovarian hormones are not necessary for stress-protection produced by 3 weeks of wheel running in female Sprague Dawley rats.


Subject(s)
Motor Activity , Physical Conditioning, Animal , Rats , Animals , Female , Male , Humans , Rats, Sprague-Dawley , Stress, Psychological , Ovariectomy , Fear
3.
Dev Psychobiol ; 63(1): 74-87, 2021 01.
Article in English | MEDLINE | ID: mdl-32524583

ABSTRACT

Vulnerabilities to fear-related disorders can be enhanced following early life adversity. This study sought to determine whether post-weaning social isolation (PSI), an animal model of early life adversity, alters the development of social fear in an innovative model of conditioned social fear. Male and female Sprague-Dawley rats underwent either social rearing (SR) or PSI for 4 weeks following weaning. Rats were then assigned to groups consisting of either Footshock only, Social conditioned stimulus (CS) only, or Paired footshock with a social CS. Social behavior was assessed the next day. We observed a novel behavioral response in PSI rats, running in circles, that was rarely observed in SR rats; moreover, this behavior was augmented after Paired treatment in PSI rats. Other social behaviors were altered by both PSI and Paired footshock and social CS. The mammalian target of rapamycin (mTOR) pathway was assessed using immunohistochemistry for phosphorylated ribosomal protein S6 (pS6) in subregions of the prefrontal cortex (PFC) and amygdala. Paired treatment produced opposite effects in the PFC and amygdala in males, but no differences were observed in females. Conditioned social fear produced alterations in social behavior and the mTOR pathway that are dependent upon rearing condition and sex.


Subject(s)
Conditioning, Classical , Fear , Animals , Behavior, Animal , Female , Male , Rats , Rats, Sprague-Dawley , Social Behavior , TOR Serine-Threonine Kinases
4.
Eur J Neurosci ; 52(1): 2530-2547, 2020 07.
Article in English | MEDLINE | ID: mdl-31800125

ABSTRACT

Adverse life events can lead to stable changes in brain structure and function and are considered primary sources of risk for post-traumatic stress disorder, depression and other neuropsychiatric disorders. However, most individuals do not develop these conditions following exposure to traumatic experiences, and research efforts have identified a number of experiential factors associated with an individual's ability to withstand, adapt to and facilitate recovery from adversity. While multiple animal models of stress resilience exist, so that the detailed biological mechanisms can be explored, studies have been disproportionately conducted in male subjects even though the prevalence and presentation of stress-linked disorders differ between sexes. This review focuses on (a) the mechanisms by which experiential factors (behavioral control over a stressor, exercise) reduce the impact of adverse events as studied in males; (b) whether other manipulations (ketamine) that buffer against stress-induced sequelae engage the same circuit features; and (c) whether these processes operate similarly in females. We argue that investigation of experiential factors that produce resistance/resilience rather than vulnerability to adversity will generate a unique set of biological mechanisms that potentially underlie sex differences in mood disorders.


Subject(s)
Ketamine , Stress Disorders, Post-Traumatic , Animals , Brain , Female , Humans , Male , Sex Characteristics , Stress, Psychological
5.
Am J Physiol Regul Integr Comp Physiol ; 319(2): R171-R183, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32551825

ABSTRACT

Exercise is often used as a strategy for weight loss maintenance. In preclinical models, we have shown that exercise may be beneficial because it counters the biological drive to regain weight. However, our studies have demonstrated sex differences in the response to exercise in this context. In the present study, we sought to better understand why females and males exhibit different compensatory food eating behaviors in response to regular exercise. Using a forced treadmill exercise paradigm, we measured weight gain, energy expenditure, food intake in real time, and the anorectic effects of leptin. The 4-wk exercise training resulted in reduced weight gain in males and sustained weight gain in females. In male rats, exercise decreased intake, whereas it increased food intake in females. Our results suggest that the anorectic effects of leptin were not responsible for these sex differences in appetite in response to exercise. If these results translate to the human condition, they may reveal important information for the use and application of regular exercise programs.


Subject(s)
Appetite/physiology , Body Weight/physiology , Eating/physiology , Energy Metabolism/physiology , Physical Conditioning, Animal/physiology , Animals , Energy Intake/physiology , Female , Male , Rats
6.
Neurobiol Learn Mem ; 176: 107328, 2020 12.
Article in English | MEDLINE | ID: mdl-33075479

ABSTRACT

Impaired fear extinction, combined with the likelihood of fear relapse after exposure therapy, contributes to the persistence of many trauma-related disorders such as anxiety and post-traumatic stress disorder. Identifying mechanisms to aid fear extinction and reduce relapse could provide novel strategies for augmentation of exposure therapy. Exercise can enhance learning and memory and augment fear extinction of traumatic memories in humans and rodents. One factor that could contribute to enhanced fear extinction following exercise is the mammalian target of rapamycin (mTOR). mTOR is a translation regulator involved in synaptic plasticity and is sensitive to many exercise signals such as monoamines, growth factors, and cellular metabolism. Further, mTOR signaling is increased after chronic exercise in brain regions involved in learning and emotional behavior. Therefore, mTOR is a compelling potential facilitator of the memory-enhancing and overall beneficial effects of exercise on mental health.The goal of the current study is to test the hypothesis that mTOR signaling is necessary for the enhancement of fear extinction produced by acute, voluntary exercise. We observed that intracerebral-ventricular administration of the mTOR inhibitor rapamycin reduced immunoreactivity of phosphorylated S6, a downstream target of mTOR, in brain regions involved in fear extinction and eliminated the enhancement of fear extinction memory produced by acute exercise, without reducing voluntary exercise behavior or altering fear extinction in sedentary rats. These results suggest that mTOR signaling contributes to exercise-augmentation of fear extinction.


Subject(s)
Extinction, Psychological/physiology , Physical Conditioning, Animal , TOR Serine-Threonine Kinases/metabolism , Animals , Brain/drug effects , Brain/metabolism , Extinction, Psychological/drug effects , Fear , Male , Memory , Rats, Long-Evans , Signal Transduction/physiology , Sirolimus/administration & dosage , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology
7.
Proc Natl Acad Sci U S A ; 113(22): E3130-9, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27185913

ABSTRACT

The prevalence of inflammatory diseases is increasing in modern urban societies. Inflammation increases risk of stress-related pathology; consequently, immunoregulatory or antiinflammatory approaches may protect against negative stress-related outcomes. We show that stress disrupts the homeostatic relationship between the microbiota and the host, resulting in exaggerated inflammation. Repeated immunization with a heat-killed preparation of Mycobacterium vaccae, an immunoregulatory environmental microorganism, reduced subordinate, flight, and avoiding behavioral responses to a dominant aggressor in a murine model of chronic psychosocial stress when tested 1-2 wk following the final immunization. Furthermore, immunization with M. vaccae prevented stress-induced spontaneous colitis and, in stressed mice, induced anxiolytic or fear-reducing effects as measured on the elevated plus-maze, despite stress-induced gut microbiota changes characteristic of gut infection and colitis. Immunization with M. vaccae also prevented stress-induced aggravation of colitis in a model of inflammatory bowel disease. Depletion of regulatory T cells negated protective effects of immunization with M. vaccae on stress-induced colitis and anxiety-like or fear behaviors. These data provide a framework for developing microbiome- and immunoregulation-based strategies for prevention of stress-related pathologies.


Subject(s)
Anxiety/complications , Bacterial Vaccines/administration & dosage , Behavior, Animal , Colitis/prevention & control , Mycobacterium/growth & development , Stress, Psychological/complications , Vaccines, Inactivated/administration & dosage , Animals , Anxiety/physiopathology , Colitis/etiology , Colitis/pathology , Immunization , Male , Mice , Mice, Inbred C57BL , Stress, Psychological/physiopathology , T-Lymphocytes, Regulatory/immunology
8.
Neurobiol Learn Mem ; 151: 28-34, 2018 05.
Article in English | MEDLINE | ID: mdl-29614374

ABSTRACT

Extinction-based exposure therapy is the most common behavioral therapy for anxiety and trauma-related disorders, but fear tends to resurface even after successful extinction. Identification of novel strategies to enhance fear extinction and reduce fear relapse is of paramount importance to mental health. Exercise can enhance cognitive function, but it is not yet well understood whether exercise can be an effective augmentation strategy for fear extinction. In the current review, we present the current state of knowledge on the effects of exercise on fear extinction. Effects of exercise duration, explanations for conflicting results, and potential mechanisms, focusing on a hypothesized role for dopamine, are all discussed. We also provide new data suggesting that the timing in which acute exercise occurs relative to fear extinction, is a crucial variable in determining whether exercise can enhance fear extinction. Clinical implications and ideas to guide future research endeavors in this area are provided.


Subject(s)
Brain/physiology , Extinction, Psychological/physiology , Fear/physiology , Implosive Therapy , Physical Conditioning, Animal , Animals , Humans , Models, Neurological , Motor Activity , Recurrence , Secondary Prevention
9.
Learn Mem ; 24(8): 358-368, 2017 08.
Article in English | MEDLINE | ID: mdl-28716955

ABSTRACT

Fear extinction-based exposure therapy is the most common behavioral therapy for anxiety and trauma-related disorders, but fear extinction memories are labile and fear tends to return even after successful extinction. The relapse of fear contributes to the poor long-term efficacy of exposure therapy. A single session of voluntary exercise can enhance the acquisition and consolidation of fear extinction in male rats, but the effects of exercise on relapse of fear after extinction are not well understood. Here, we characterized the effects of 2 h of voluntary exercise during the consolidation phase of contextual or auditory fear extinction learning on long-term fear extinction memory and renewal in adult, male and female, Long-Evans rats. Results indicate that exercise enhances consolidation of fear extinction memory and reduces fear relapse after extinction in a sex-dependent manner. These data suggest that brief bouts of exercise could be used as an augmentation strategy for exposure therapy, even in previously sedentary subjects. Fear memories of discrete cues, rather than of contextual ones, may be most susceptible to exercise-augmented extinction, especially in males. Additionally, exercise seems to have the biggest impact on fear relapse phenomena, even if fear extinction memories themselves are only minimally enhanced.


Subject(s)
Extinction, Psychological , Fear , Memory Consolidation , Running/psychology , Sex Characteristics , Analysis of Variance , Animals , Auditory Perception , Electroshock , Estrus/physiology , Extinction, Psychological/physiology , Fear/physiology , Female , Freezing Reaction, Cataleptic/physiology , Male , Memory Consolidation/physiology , Psychological Tests , Rats, Long-Evans , Running/physiology , Volition
10.
Eur J Neurosci ; 45(3): 342-357, 2017 02.
Article in English | MEDLINE | ID: mdl-27763700

ABSTRACT

Manipulating gut microbes may improve mental health. Prebiotics are indigestible compounds that increase the growth and activity of health-promoting microorganisms, yet few studies have examined how prebiotics affect CNS function. Using an acute inescapable stressor known to produce learned helplessness behaviours such as failure to escape and exaggerated fear, we tested whether early life supplementation of a blend of two prebiotics, galactooligosaccharide (GOS) and polydextrose (PDX), and the glycoprotein lactoferrin (LAC) would attenuate behavioural and biological responses to stress later in life. Juvenile, male F344 rats were fed diets containing either GOS and PDX alone, LAC alone, or GOS, PDX and LAC. All diets altered gut bacteria, while diets containing GOS and PDX increased Lactobacillus spp. After 4 weeks, rats were exposed to inescapable stress, and either immediately killed for blood and tissues, or assessed for learned helplessness 24 h later. Diets did not attenuate stress effects on spleen weight, corticosterone and blood glucose; however, all diets differentially attenuated stress-induced learned helplessness. Notably, in situ hybridization revealed that all diets reduced stress-evoked cfos mRNA in the dorsal raphe nucleus (DRN), a structure important for learned helplessness behaviours. In addition, GOS, PDX and LAC diet attenuated stress-evoked decreases in mRNA for the 5-HT1A autoreceptor in the DRN and increased basal BDNF mRNA within the prefrontal cortex. These data suggest early life diets containing prebiotics and/or LAC promote behavioural stress resistance and uniquely modulate gene expression in corresponding circuits.


Subject(s)
Diet , Helplessness, Learned , Lactoferrin/therapeutic use , Prebiotics , Stress, Psychological/diet therapy , Animals , Brain-Derived Neurotrophic Factor/metabolism , Lactoferrin/pharmacology , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/growth & development , Raphe Nuclei/metabolism , Rats , Rats, Inbred F344 , Receptor, Serotonin, 5-HT1A/metabolism , Stress, Psychological/prevention & control
11.
Eur J Neurosci ; 43(9): 1190-202, 2016 05.
Article in English | MEDLINE | ID: mdl-26833814

ABSTRACT

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.


Subject(s)
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
12.
Stress ; 19(3): 312-24, 2016 05.
Article in English | MEDLINE | ID: mdl-27124542

ABSTRACT

Regular physical activity produces resistance to the negative health consequences of stressor exposure. One way that exercise may confer stress resistance is by reducing the impact of stress on diurnal rhythms and sleep; disruptions of which contribute to stress-related disease including mood disorders. Given the link between diurnal rhythm disruptions and stress-related disorders and that exercise both promotes stress resistance and is a powerful non-photic biological entrainment cue, we tested if wheel running could reduce stress-induced disruptions of sleep/wake behavior and diurnal rhythms. Adult, male F344 rats with or without access to running wheels were instrumented for biotelemetric recording of diurnal rhythms of locomotor activity, heart rate, core body temperature (CBT), and sleep (i.e. REM, NREM, and WAKE) in the presence of a 12 h light/dark cycle. Following 6 weeks of sedentary or exercise conditions, rats were exposed to an acute stressor known to disrupt diurnal rhythms and produce behaviors associated with mood disorders. Prior to stressor exposure, exercise rats had higher CBT, more locomotor activity during the dark cycle, and greater %REM during the light cycle relative to sedentary rats. NREM and REM sleep were consolidated immediately following peak running to a greater extent in exercise, compared to sedentary rats. In response to stressor exposure, exercise rats expressed higher stress-induced hyperthermia than sedentary rats. Stressor exposure disrupted diurnal rhythms in sedentary rats; and wheel running reduced these effects. Improvements in sleep and reduced diurnal rhythm disruptions following stress could contribute to the health promoting and stress protective effects of exercise.


Subject(s)
Circadian Rhythm/physiology , Motor Activity/physiology , Physical Conditioning, Animal/physiology , Sleep, REM/physiology , Stress, Psychological/physiopathology , Animals , Heart Rate/physiology , Male , Rats , Rats, Inbred F344 , Running/physiology
13.
Neurobiol Learn Mem ; 125: 224-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26454156

ABSTRACT

Relapse of previously extinguished fear presents a significant, pervasive obstacle to the successful long-term treatment of anxiety and trauma-related disorders. Thus, identification of a novel means to enhance fear extinction to stand the passage of time and generalize across contexts is of the utmost importance. Acute bouts of exercise can be used as inexpensive, noninvasive treatment strategies to reduce anxiety, and have been shown to enhance memory for extinction when performed in close temporal proximity to the extinction session. However, it is unclear whether acute exercise can be used to prevent relapse of fear, and the neural mechanisms underlying this potential effect are unknown. The current study therefore examined whether acute exercise during extinction of auditory fear can protect against the later relapse of fear. Male F344 rats lacking an extended history of wheel running were conditioned to fear a tone CS and subsequently extinguished within either a freely mobile running wheel, a locked wheel, or a control context lacking a wheel. Rats exposed to fear extinction within a freely mobile wheel ran during fear extinction, and demonstrated reduced fear as well as attenuated corticosterone levels during re-exposure to the extinguished CS during the relapse test in a novel context 1week later. Examination of cfos mRNA patterns elicited by re-exposure to the extinguished CS during the relapse test revealed that acute exercise during extinction decreased activation of brain circuits classically involved in driving fear expression and interestingly, increased activity within neurons of the direct striatal pathway involved in reward signaling. These data suggest that exercise during extinction reduces relapse through a mechanism involving the direct pathway of the striatum. It is suggested that a positive affective state could become associated with the CS during exercise during extinction, thus resulting in a relapse-resistant extinction memory.


Subject(s)
Conditioning, Classical/physiology , Corpus Striatum/physiology , Extinction, Psychological/physiology , Fear/physiology , Motor Activity/physiology , Physical Conditioning, Animal/physiology , Acoustic Stimulation , Animals , Fear/psychology , Male , Neural Pathways/physiology , Neurons/physiology , Rats , Rats, Inbred F344
14.
Brain Behav Immun ; 39: 87-98, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24246250

ABSTRACT

To prime local tissues for dealing with potential infection or injury, exposure to an acute, intense stressor evokes increases in circulating and local tissue inflammatory proteins. Regular physical activity facilitates stress-evoked innate reactivity and modulates the expression of inflammatory proteins in immuno-metabolic tissues such as white adipose tissue (WAT). The impact of regular physical activity on stress-evoked inflammatory protein expression in WAT, however, remains unclear. To investigate this question, lean male F344 rats (150-175g) were allowed voluntary access to a running wheel for 6weeks followed by exposure to an acute stressor (100, 1.5mA-5s inescapable tail shocks). Using ELISAs, corticosterone, heat shock protein 72 (Hsp72), macrophage chemoattractant protein (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1ß, IL-6, and IL-10 concentrations were measured in plasma and subcutaneous, intraperitoneal (epididymal and retroperitoneal WAT depots) and visceral (omental and mesenteric WAT depots) WAT compartments. Acute stress increased plasma concentrations of all proteins except TNF-α and, depending upon the compartment examined, WAT concentrations of MCP-1, IL-1ß, IL-6, and IL-10. Exercise ubiquitously increased IL-1ß within WAT, potentiated stress-evoked Hsp72 in plasma and WAT, and differentially increased stress-evoked MCP-1, IL-6, and IL-10 within WAT. These data suggest: (a) inflammatory proteins in non-obese WAT may serve compartment-specific immune and metabolic roles important to the acute stress response and; (b) voluntary habitual exercise may optimize stress-induced augmentation of innate immune function through increases in stress-evoked Hsp72, MCP-1, IL-6, and IL-10 and decreases in IL-1ß/IL10 and TNF-α/IL10 ratios within white adipose tissue.


Subject(s)
Adipose Tissue, White/metabolism , Cytokines/metabolism , HSP72 Heat-Shock Proteins/metabolism , Motor Activity/physiology , Stress, Psychological/metabolism , Animals , Chemokine CCL2/metabolism , Corticosterone/metabolism , Electric Stimulation , Interleukin-10/metabolism , Interleukin-6/metabolism , Male , Rats , Rats, Inbred F344
15.
Stress ; 17(1): 1-12, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23574145

ABSTRACT

Exposure to an uncontrollable stressor elicits a constellation of physiological and behavioral sequel in laboratory rats that often reflect aspects of anxiety and other emotional disruptions. We review evidence suggesting that plasticity within the serotonergic dorsal raphe nucleus (DRN) is critical to the expression of uncontrollable stressor-induced anxiety. Specifically, after uncontrollable stressor exposure subsequent anxiogenic stimuli evoke greater 5-HT release in DRN terminal regions including the amygdala and striatum; and pharmacological blockade of postsynaptic 5-HT(2C) receptors in these regions prevents expression of stressor-induced anxiety. Importantly, the controllability of stress, the presence of safety signals, and a history of exercise mitigate the expression of stressor-induced anxiety. These stress-protective factors appear to involve distinct neural substrates; with stressor controllability requiring the medial prefrontal cortex, safety signals the insular cortex and exercise affecting the 5-HT system directly. Knowledge of the distinct yet converging mechanisms underlying these stress-protective factors could provide insight into novel strategies for the treatment and prevention of stress-related psychiatric disorders.


Subject(s)
Anxiety/physiopathology , Prefrontal Cortex/physiology , Raphe Nuclei/physiopathology , Serotonin/metabolism , Stress, Psychological/psychology , Animals , Helplessness, Learned , Physical Conditioning, Animal/physiology , Rats , Receptor, Serotonin, 5-HT2C/physiology
16.
Article in English | MEDLINE | ID: mdl-39080242

ABSTRACT

Prior physical activity reduces the risk of future stress-related mental health disorders including depression, anxiety, and post-traumatic stress disorder. Rodents allowed to engage in voluntary wheel running are similarly protected from behavioral consequences of stress. The present review summarizes current knowledge on mechanisms underlying exercise-induced stress resistance. A conceptual framework involving the development (during exercise) and expression (during stress) of stress resistance from exercise is proposed. During the development of stress resistance, adaptations involving multiple exercise signals and molecular mediators occur within neural circuits orchestrating various components of the stress response, which then respond differently to stress during the expression of stress resistance. Recent data indicate that the development and expression of stress resistance from exercise involve multiple independent mechanisms that depend on sex, stressor severity, and behavioral outcome. Recent insight into the role of the prefrontal cortex in exercise-induced stress resistance illustrates these multiple mechanisms. This knowledge has important implications for the design of future experiments aimed at identifying the mechanisms underlying exercise-induced stress resistance.

17.
Eur J Neurosci ; 37(3): 469-78, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23121339

ABSTRACT

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.


Subject(s)
Physical Exertion , Prefrontal Cortex/physiopathology , Stress, Physiological , Stress, Psychological/physiopathology , Animals , Anxiety/physiopathology , Conditioning, Classical , Depressive Disorder/physiopathology , Electroshock , Male , Rats , Rats, Inbred F344
18.
Brain Behav Immun ; 28: 54-62, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23103443

ABSTRACT

Exposure to stressors or trauma in the absence of pathogenic challenge can stimulate a systemic sterile inflammatory response characterized by high concentrations of blood and tissue cytokines, chemokines, and danger associated molecular patterns (DAMPs) such as heat shock protein-72 (Hsp72), and uric acid. The signaling pathways responsible for these responses remain unclear, however, the inflammasome may play a role. In vitro, DAMPs are known to stimulate the inflammasome in the presence of LPS to activate caspase-1 which cleaves immature precursors of interleukin (IL)-1ß and IL-18 into their mature releasable forms. Furthermore, in vivo neutralization of the LPS selectively attenuates the stress-induced increase in the inflammasome-dependent cytokines IL-1ß and IL-18. Thus, the current experiments tested the hypothesis that inflammasome-mediated processes are necessary for a systemic stress-induced inflammatory response to an acute stressor. The data presented (1) establish that male F344 rats exposed to an acute severe stressor (100 tail shocks) have elevated plasma concentrations of inflammatory proteins (IL-1ß, IL-18, IL-6, IL-10, and monocyte chemotactic protein (MCP)-1), and DAMPs (uric acid and Hsp72); (2) demonstrate that inhibiting caspase-1 in vivo, using the caspase-1 inhibitor ac-YVAD-cmk, attenuates stress-induced production of IL-1ß, IL-18, and IL-6 in both the circulation and peripheral tissues; and (3) implicates the DAMPs uric acid and Hsp72 as important signals contributing to inflammasome-dependent inflammatory responses using a stepwise multiple regression. The results increase our mechanistic understanding of systemic sterile inflammatory responses, and provide novel evidence that the inflammasome may be an important pharmacological target for treatment of these conditions.


Subject(s)
Chemokines/physiology , Cytokines/physiology , Inflammasomes/physiology , Stress, Psychological/physiopathology , Animals , Caspase 1/drug effects , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Chemokine CCL2/physiology , HSP72 Heat-Shock Proteins/blood , Inflammasomes/drug effects , Interleukin-10/physiology , Interleukin-18/physiology , Interleukin-1beta/physiology , Interleukin-6/physiology , Male , Rats , Rats, Inbred F344 , Stress, Psychological/immunology , Uric Acid/blood
19.
J Anxiety Disord ; 94: 102680, 2023 03.
Article in English | MEDLINE | ID: mdl-36773486

ABSTRACT

Posttraumatic stress disorder (PTSD) is associated with heightened emotional responding, avoidance of trauma related stimuli, and physical health concerns (e.g., metabolic syndrome, type 2 diabetes, cardiovascular disease). Existing treatments such as exposure-based therapies (e.g., prolonged exposure) aim to reduce anxiety symptoms triggered by trauma reminders, and are hypothesized to work via mechanisms of extinction learning. However, these conventional gold standard psychotherapies do not address physical health concerns frequently presented in PTSD. In addition to widely documented physical and mental health benefits of exercise, emerging preclinical and clinical evidence supports the hypothesis that precisely timed administration of aerobic exercise can enhance the consolidation and subsequent recall of fear extinction learning. These findings suggest that aerobic exercise may be a promising adjunctive strategy for simultaneously improving physical health while enhancing the effects of exposure therapies, which is desirable given the suboptimal efficacy and remission rates. Accordingly, this review 1) encompasses an overview of preclinical and clinical exercise and fear conditioning studies which form the basis for this claim; 2) discusses several plausible mechanisms for enhanced consolidation of fear extinction memories following exercise, and 3) provides suggestions for future research that could advance the understanding of the potential importance of incorporating exercise into the treatment of PTSD.


Subject(s)
Diabetes Mellitus, Type 2 , Implosive Therapy , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/psychology , Fear/psychology , Extinction, Psychological , Laboratories, Clinical , Exercise
20.
Psychopharmacology (Berl) ; 239(11): 3697-3709, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36195731

ABSTRACT

RATIONALE: Exercise participation remains low despite clear benefits. Rats engage in voluntary wheel running (VWR) that follows distinct phases of acquisition, during which VWR escalates, and maintenance, during which VWR remains stable. Understanding mechanisms driving acquisition and maintenance of VWR could lead to novel strategies to promote exercise. The two phases of VWR resemble those that occur during operant conditioning and, therefore, might involve similar neural substrates. The dorsomedial (DMS) dorsal striatum (DS) supports the acquisition of operant conditioning, whereas the dorsolateral striatum (DLS) supports its maintenance. OBJECTIVES: Here we sought to characterize the roles of DS subregions in VWR. Females escalate VWR and operant conditioning faster than males. Thus, we also assessed for sex differences. METHODS: To determine the causal role of DS subregions in VWR, we pharmacologically inactivated the DMS or DLS of adult, male and female, Long-Evans rats during the two phases of VWR. The involvement of DA receptor 1 (D1)-expressing neurons in the DS was investigated by quantifying cfos mRNA within this neuronal population. RESULTS: We observed that, in males, the DMS and DLS are critical for VWR exclusively during acquisition and maintenance, respectively. In females, the DMS is also critical only during acquisition, but the DLS contributes to VWR during both VWR phases. DLS D1 neurons could be an important driver of VWR escalation during acquisition. CONCLUSIONS: The acquisition and maintenance of VWR involve unique neural substrates in the DS that vary by sex. Results reveal targets for sex-specific strategies to promote exercise.


Subject(s)
Corpus Striatum , Motor Activity , Rats , Animals , Female , Male , Rats, Long-Evans , Corpus Striatum/physiology , Neostriatum , RNA, Messenger
SELECTION OF CITATIONS
SEARCH DETAIL