Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 378: 112237, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31525404

ABSTRACT

Voluntary exercise increases stress resistance by modulating stress-responsive neurocircuitry, including brainstem serotonergic systems. However, it remains unknown how exercise produces adaptations to serotonergic systems. Recruitment of serotonergic systems during repeated, daily exercise could contribute to the adaptations in serotonergic systems following exercise, but whether repeated voluntary exercise recruits serotonergic systems is unknown. In this study, we investigated the effects of six weeks of voluntary or forced exercise on rat brain serotonergic systems. Specifically, we analyzed c-Fos and FosB/ΔFosB as markers of acute and chronic cellular activation, respectively, in combination with tryptophan hydroxylase, a marker of serotonergic neurons, within subregions of the dorsal raphe nucleus using immunohistochemical staining. Compared to sedentary controls, rats exposed to repeated forced exercise, but not repeated voluntary exercise, displayed decreased c-Fos expression in serotonergic neurons in the rostral dorsal portion of the dorsal raphe nucleus (DRD) and increased c-Fos expression in serotonergic neurons in the caudal DR (DRC), and interfascicular part of the dorsal raphe nucleus (DRI) during the active phase of the diurnal activity rhythm. Similarly, increases in c-Fos expression in serotonergic neurons in the DRC, DRI, and ventral portion of the dorsal raphe nucleus (DRV) were observed in rats exposed to repeated forced exercise, compared to rats exposed to repeated voluntary exercise. Six weeks of forced exercise, relative to the sedentary control condition, also increased FosB/ΔFosB expression in DRD, DRI, and DRV serotonergic neurons. While both voluntary and forced exercise increase stress resistance, these results suggest that repeated forced exercise, but not repeated voluntary exercise, increases activation of DRI serotonergic neurons, an effect that may contribute to the stress resistance effects of forced exercise. These results also suggest that mechanisms of exercise-induced stress resistance may differ depending on the controllability of the exercise.


Subject(s)
Behavior, Animal/physiology , Motor Activity/physiology , Physical Conditioning, Animal/physiology , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/metabolism , Serotonergic Neurons/metabolism , Serotonin/metabolism , Tryptophan Hydroxylase/metabolism , Animals , Immunohistochemistry , Male , Rats , Rats, Inbred F344
2.
Int Rev Neurobiol ; 131: 165-191, 2016.
Article in English | MEDLINE | ID: mdl-27793217

ABSTRACT

The gut microbial ecosystem can mediate the negative health impacts of stress on the host. Stressor-induced disruptions in microbial ecology (dysbiosis) can lead to maladaptive health effects, while certain probiotic organisms and their metabolites can protect against these negative impacts. Prebiotic diets and exercise are feasible and cost-effective strategies that can increase stress-protective bacteria and produce resistance against the detrimental behavioral and neurobiological impacts of stress. The goal of this review is to describe research demonstrating that both prebiotic diets and exercise produce adaptations in gut ecology and the brain that arm the organism against inescapable stress-induced learned helplessness. The results of this research support the novel hypothesis that some of the stress-protective effects of prebiotics and exercise are due to increases in stress-protective gut microbial species and their metabolites. In addition, new evidence also suggests that prebiotic diet or exercise interventions are most effective if given early in life (juvenile-adolescence) when both the gut microbial ecosystem and the brain are plastic. Based on our new understanding of the mechanistic convergence of these interventions, it is feasible to propose that in adults, both interventions delivered in combination may elevate their efficacy to promote a stress-resistant phenotype.


Subject(s)
Bacteria/drug effects , Butyrates/pharmacology , Exercise/physiology , Gastrointestinal Tract , Prebiotics/administration & dosage , Stress, Psychological , Animals , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology , Humans , Stress, Psychological/microbiology , Stress, Psychological/prevention & control , Stress, Psychological/rehabilitation
3.
Int J Tuberc Lung Dis ; 20(5): 582-7, 2016 May.
Article in English | MEDLINE | ID: mdl-27084809

ABSTRACT

SETTING: Tertiary referral center, National Institutes of Health (NIH), USA. OBJECTIVE: To estimate the mortality rate and its correlates among persons with pulmonary non-tuberculous mycobacteria (PNTM) disease. DESIGN: A retrospective review of 106 patients who were treated at the NIH Clinical Center and met American Thoracic Society/Infectious Diseases Society of America criteria for PNTM. Eligible patients were aged ⩾18 years and did not have cystic fibrosis or human immunodeficiency virus (HIV) infection. RESULTS: Of 106 patients followed for a median of 4.9 years, 27 (25%) died during follow-up, for a mortality rate of 4.2 per 100 person-years. The population was predominantly female (88%) and White (88%), with infrequent comorbidities. Fibrocavitary disease (adjusted hazard ratio [aHR] 3.3, 95% confidence interval [CI] 1.3-8.3) and pulmonary hypertension (aHR 2.1, 95%CI 0.9-5.1) were associated with a significantly elevated risk of mortality in survival analysis. CONCLUSIONS: PNTM remains a serious public health concern, with a consistently elevated mortality rate across multiple populations. Significant risk factors for death include fibrocavitary disease and pulmonary hypertension. Further research is needed to more specifically identify clinical and microbiologic factors that jointly influence disease outcome.


Subject(s)
Lung/microbiology , Mycobacterium Infections, Nontuberculous/mortality , Nontuberculous Mycobacteria/isolation & purification , Respiratory Tract Infections/mortality , Female , Humans , Hypertension, Pulmonary/microbiology , Hypertension, Pulmonary/mortality , Kaplan-Meier Estimate , Lung/diagnostic imaging , Male , Middle Aged , Mycobacterium Infections, Nontuberculous/diagnostic imaging , Mycobacterium Infections, Nontuberculous/microbiology , National Institutes of Health (U.S.) , Nontuberculous Mycobacteria/classification , Proportional Hazards Models , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/mortality , Respiratory Tract Infections/diagnostic imaging , Respiratory Tract Infections/microbiology , Retrospective Studies , Risk Factors , Tertiary Care Centers , Time Factors , Tomography, X-Ray Computed , United States/epidemiology
4.
Acta Physiol (Oxf) ; 211(2): 447-65, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24447583

ABSTRACT

AIM: To identify objective factors that can predict future sensitized stress responses, thus allowing for effective intervention prior to developing sensitization and subsequent stress-related disorders, including post-traumatic stress disorder (PTSD). METHODS: Adult male F344 rats implanted with biotelemetry devices were exposed to repeated conditioned fear or control conditions for 22 days followed by exposure to either no, mild or severe acute stress on day 23. Diurnal rhythms of locomotor activity (LA), heart rate (HR) and core body temperature (CBT) were biotelemetrically monitored throughout the study. In a subset of rat not implanted, corticosterone and indices of chronic stress were measured immediately following stress. RESULTS: Rats exposed to repeated fear had fear-evoked increases in behavioural freezing and HR/CBT during exposure to the fear environment and displayed indices of chronic stress. Repeated fear produced flattening of diurnal rhythms in LA, HR and CBT. Repeated fear did not sensitize the corticosterone response to acute stress, but produced sensitized HR/CBT responses following acute stress, relative to the effect of acute stress in the absence of a history of repeated fear. Greater diurnal rhythm disruptions during repeated fear predicted sensitized acute stress-induced physiological responses. Rats exposed to repeated fear also displayed flattened diurnal LA and basal increases in HR. CONCLUSIONS: Exposure to repeated fear produces outcomes consistent with those observed in PTSD. The results suggest that diurnal rhythm disruptions during chronic stressors may help predict sensitized physiological stress responses following traumatic events. Monitoring diurnal disruptions during repeated stress may thus help predict susceptibility to PTSD.


Subject(s)
Circadian Rhythm/physiology , Fear/physiology , Stress Disorders, Post-Traumatic/physiopathology , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Animals , Body Temperature/physiology , Conditioning, Classical , Disease Models, Animal , Heart Rate/physiology , Male , Motor Activity/physiology , Rats , Rats, Inbred F344
5.
J Neurosci ; 32(33): 11187-200, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895704

ABSTRACT

Opioid action was thought to exert reinforcing effects solely via the initial agonism of opioid receptors. Here, we present evidence for an additional novel contributor to opioid reward: the innate immune pattern-recognition receptor, toll-like receptor 4 (TLR4), and its MyD88-dependent signaling. Blockade of TLR4/MD2 by administration of the nonopioid, unnatural isomer of naloxone, (+)-naloxone (rats), or two independent genetic knock-outs of MyD88-TLR4-dependent signaling (mice), suppressed opioid-induced conditioned place preference. (+)-Naloxone also reduced opioid (remifentanil) self-administration (rats), another commonly used behavioral measure of drug reward. Moreover, pharmacological blockade of morphine-TLR4/MD2 activity potently reduced morphine-induced elevations of extracellular dopamine in rat nucleus accumbens, a region critical for opioid reinforcement. Importantly, opioid-TLR4 actions are not a unidirectional influence on opioid pharmacodynamics, since TLR4(-/-) mice had reduced oxycodone-induced p38 and JNK phosphorylation, while displaying potentiated analgesia. Similar to our recent reports of morphine-TLR4/MD2 binding, here we provide a combination of in silico and biophysical data to support (+)-naloxone and remifentanil binding to TLR4/MD2. Collectively, these data indicate that the actions of opioids at classical opioid receptors, together with their newly identified TLR4/MD2 actions, affect the mesolimbic dopamine system that amplifies opioid-induced elevations in extracellular dopamine levels, therefore possibly explaining altered opioid reward behaviors. Thus, the discovery of TLR4/MD2 recognition of opioids as foreign xenobiotic substances adds to the existing hypothesized neuronal reinforcement mechanisms, identifies a new drug target in TLR4/MD2 for the treatment of addictions, and provides further evidence supporting a role for central proinflammatory immune signaling in drug reward.


Subject(s)
Analgesics, Opioid/administration & dosage , Conditioning, Operant/drug effects , Reinforcement, Psychology , Toll-Like Receptor 4/metabolism , Analgesics, Opioid/blood , Analysis of Variance , Animals , Conditioning, Operant/physiology , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microdialysis , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Myeloid Differentiation Factor 88/deficiency , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Pain Threshold/drug effects , Pain Threshold/physiology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Self Administration , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/deficiency
6.
Neuroscience ; 197: 132-44, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21958863

ABSTRACT

Uncontrollable stress can interfere with instrumental learning and induce anxiety in humans and rodents. While evidence supports a role for serotonin (5-HT) and serotonin 2C receptors (5-HT(2C)R) in the behavioral consequences of uncontrollable stress, the specific sites of action are unknown. These experiments sought to delineate the role of 5-HT and 5-HT(2C)R in the dorsal striatum (DS) and the lateral/basolateral amygdala (BLA) in the expression of stress-induced instrumental escape deficits and exaggerated fear, as these structures are critical to instrumental learning and fear behaviors. Using in vivo microdialysis, we first demonstrated that prior uncontrollable, but not controllable, stress sensitizes extracellular 5-HT in the dorsal striatum, a result that parallels prior work in the BLA. Additionally, rats were implanted with bi-lateral cannula in either the DS or the BLA and exposed to uncontrollable tail shock stress. One day later, rats were injected with 5-HT(2C)R antagonist (SB242084) and fear and instrumental learning behaviors were assessed in a shuttle box. Separately, groups of non-stressed rats received an intra-DS or an intra-BLA injection of the 5-HT(2C)R agonist (CP809101) and behavior was observed. Intra-DS injections of the 5-HT(2C)R antagonist prior to fear/escape tests completely blocked the stress-induced interference with instrumental escape learning; a partial block was observed when injections were in the BLA. Antagonist administration in either region did not influence stress-induced fear behavior. In the absence of prior stress, intra-DS administration of the 5-HT(2C)R agonist was sufficient to interfere with escape behavior without enhancing fear, while intra-BLA administration of the 5-HT(2C)R agonist increased fear behavior but had no effect on escape learning. Results reveal a novel role of the 5-HT(2C)R in the DS in the expression of instrumental escape deficits produced by uncontrollable stress and demonstrate that the involvement of 5-HT(2C)R activation in stress-induced behaviors is regionally specific.


Subject(s)
Conditioning, Operant/physiology , Corpus Striatum/metabolism , Helplessness, Learned , Receptor, Serotonin, 5-HT2C/metabolism , Stress, Psychological/metabolism , Amygdala/metabolism , Animals , Behavior, Animal , Male , Microdialysis , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
7.
J Neuroendocrinol ; 22(8): 872-88, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20406350

ABSTRACT

Regular physical exercise is beneficial for both physical and mental health. By contrast, stress is associated with deleterious effects on health and there is growing evidence that regular physical exercise counteracts some of the effects of stress. However, most previous studies have suggested that prior exercise does not alter the acute hypothalamic pituitary adrenal (HPA) axis responses to stress. The present series of studies provides evidence that in rats, 6 weeks (but not 1 or 3 weeks) of voluntary wheel running reduces the HPA axis responses to lower-intensity stressors such as an i.p. saline injection, exposure to a novel environment or exposure to moderate intensity noise, but not to more intense stressors such as predator odour exposure or restraint. Daily exercise does not appear to be necessary for the reduction in HPA axis responses, with intermittent access (24 h out of each 72-h period) to a running wheel for 6 weeks, resulting in similar decrements in adrenocorticotrophic hormone and corticosterone release in response to 85 dBA noise exposure. Data from in situ hybridisation for c-fos mRNA are consistent with the hypothesis that voluntary exercise results in a decrease in HPA axis responsiveness to a low-intensity stressor at a central level, with no changes in primary sensory processing. Together, these data suggest that 6 weeks of daily or intermittent exercise constrains the HPA axis response to mild, but not more intense stressors, and that this regulation may be mediated at a central level beyond the primary sensory input.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Motor Activity/physiology , Pituitary-Adrenal System/physiology , Running , Stress, Physiological , Acoustic Stimulation , Adrenocorticotropic Hormone/blood , Animals , Brain/anatomy & histology , Brain/metabolism , Male , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
8.
Brain Behav Immun ; 22(6): 923-32, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18295445

ABSTRACT

Based upon a prior cross-sectional study, we hypothesized that an aerobic exercise intervention in sedentary older adults would improve a primary T cell-dependent immune response. Participants were a subset of older subjects from a large, ongoing exercise intervention study who were randomly assigned to either an aerobic exercise (Cardio, n=30, 68.9+0.8 years) or flexibility/balance (Flex, n=20, 69.9+1.2 years) intervention. The intervention consisted of either three aerobic sessions for 30-60 min at 55-70% VO(2 max) or two 60 min flexibility/balance sessions weekly for 10 months. Eight months into the intervention, samples were collected before intramuscular administration of KLH (125 microg), followed by sampling at 2, 3, and 6 weeks post-KLH. Serum anti-KLH IgM, IgG1, and IgG2 was measured by ELISA. Physiological and psychosocial measures were also assessed pre- and post-intervention. While there was no difference in the anti-KLH IgG2 response between groups, Cardio displayed significantly (p<0.05) higher anti-KLH IgG1 (at weeks 2, 3, and 6 post) and IgM responses when compared to Flex. Despite cardiovascular intervention-induced improvement in physical fitness (approximately 11% vs. 1% change in VO(2 peak) in Cardio vs. Flex, respectively), we found no relationship between improved fitness and enhanced anti-KLH antibody responses. Optimism, perceived stress, and affect were all associated with enhanced immune response. We have shown for the first time that cardiovascular training in previously sedentary elderly results in significantly higher primary IgG1 and IgM antibody responses, while having no effect on IgG2 production.


Subject(s)
Antibody Formation/immunology , Exercise/physiology , Hemocyanins/immunology , Postural Balance/physiology , Aged , Aged, 80 and over , Enzyme-Linked Immunosorbent Assay , Follow-Up Studies , Hemocyanins/administration & dosage , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Injections, Intramuscular , Monitoring, Physiologic
9.
Neuroscience ; 144(4): 1193-208, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17161541

ABSTRACT

Reduced levels of brain-derived neurotrophic factor (BDNF) in the hippocampus have been implicated in human affective disorders and behavioral stress responses. The current studies examined the role of BDNF in the behavioral consequences of inescapable stress, or learned helplessness. Inescapable stress decreased BDNF mRNA and protein in the hippocampus of sedentary rats. Rats allowed voluntary access to running wheels for either 3 or 6 weeks prior to exposure to stress were protected against stress-induced reductions of hippocampal BDNF protein. The observed prevention of stress-induced deceases in BDNF, however, occurred in a time course inconsistent with the prevention of learned helplessness by wheel running, which is evident following 6 weeks, but not 3 weeks, of wheel running. BDNF suppression in physically active rats was produced by administering a single injection of the selective serotonin reuptake inhibitor fluoxetine (10 mg/kg) just prior to stress. Despite reduced levels of hippocampal BDNF mRNA following stress, physically active rats given the combination of fluoxetine and stress remained resistant against learned helplessness. Sedentary rats given both fluoxetine and stress still demonstrated typical learned helplessness behaviors. Fluoxetine by itself reduced BDNF mRNA in sedentary rats only, but did not affect freezing or escape learning 24 h later. Finally, bilateral injections of BDNF (1 mug) into the dentate gyrus prior to stress prevented stress-induced reductions of hippocampal BDNF but did not prevent learned helplessness in sedentary rats. These data indicate that learned helplessness behaviors are independent of the presence or absence of hippocampal BDNF because blocking inescapable stress-induced BDNF suppression does not always prevent learned helplessness, and learned helplessness does not always occur in the presence of reduced BDNF. Results also suggest that the prevention of stress-induced hippocampal BDNF suppression is not necessary for the protective effect of wheel running against learned helplessness.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Helplessness, Learned , Hippocampus/metabolism , Stress, Psychological/metabolism , Animals , Antidepressive Agents, Second-Generation/pharmacology , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Down-Regulation/drug effects , Down-Regulation/physiology , Fear/drug effects , Fear/physiology , Fluoxetine/pharmacology , Hippocampus/drug effects , Male , Motor Activity/drug effects , Motor Activity/physiology , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Serotonin/metabolism , Stress, Psychological/physiopathology
10.
J Appl Physiol (1985) ; 101(2): 566-75, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16690792

ABSTRACT

The majority of previous work examining stress responses has been done in males. Recently, it has become clear that the impact of stressor exposure is modulated by sex. One stress response that may be affected by sex is the induction of intracellular heat shock protein (HSP) 72, which is a stress- responsive molecular chaperone that refolds denatured proteins and promotes cellular survival. The following study compared HSP72 in males and females and also examined whether the estrous cycle altered HSP72 induction in females. We hypothesized that females compared with males would have a constrained HSP72 response after an acute stressor and that the stress-induced HSP72 response in females would fluctuate with the estrous cycle. Male and female F344 rats were either left in their home cage or exposed to acute tail-shock stress (8-10/group). Immediately following stressor, trunk blood was collected and tissues were flash frozen. Vaginal smear and estrogen enzyme immunoassay were used to categorize the phase of estrous. Results show that female rats had a greater corticosterone response than males, that both males and females exhibit a stress-induced release of progesterone, and that males and females had equal levels of stress-induced circulating norepinephrine. Sexual dimorphism of the HSP72 (ELISA) response existed in pituitary gland, mesenteric lymph nodes, and liver such that female rats had an attenuated HSP72 response compared with males after stress. The adrenal glands, spleen, and heart did not exhibit sexual dimorphism of the HSP72 response. The estrous cycle did not have a significant effect on basal or stress-induced HSP72 in females.


Subject(s)
Gene Expression Regulation/physiology , HSP72 Heat-Shock Proteins/metabolism , Sex Characteristics , Stress, Physiological/physiopathology , Adrenal Cortex Hormones/blood , Adrenal Glands/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Estrogens/blood , Estrous Cycle/physiology , Female , HSP72 Heat-Shock Proteins/genetics , Liver/metabolism , Lymph Nodes/metabolism , Male , Muscle, Skeletal/metabolism , Myocardium/metabolism , Norepinephrine/blood , Pituitary Gland/metabolism , Progesterone/blood , Rats , Spleen/metabolism , Stress, Physiological/genetics
11.
Neuroscience ; 135(4): 1295-307, 2005.
Article in English | MEDLINE | ID: mdl-16165282

ABSTRACT

Proinflammatory cytokines act at receptors in the CNS to alter physiological and behavioral responses. Exposure to stressors increases both peripheral and central proinflammatory cytokines, yet the mechanism(s) of induction remain unknown. Experiments here examined the role of catecholamines in the in vivo induction of proinflammatory cytokines following tailshock stress. Rats were pretreated i.p. with 2.0 mg/kg prazosin (alpha1-adrenoceptor antagonist), 10.0 mg/kg propranolol (beta-adrenoceptor antagonist), or 5.0 mg/kg labetalol (alpha1- and beta-adrenoceptor antagonist) 30 min prior to tailshock exposure and plasma interleukin-1beta (IL-1beta) and IL-6, along with tissue interleukin-1beta from the hypothalamus, hippocampus, and pituitary were measured immediately following stressor termination. Prazosin attenuated stress-induced plasma IL-1beta and IL-6, but had no effect on tissue IL-1beta levels, while propranolol attenuated plasma IL-6 and blocked tissue IL-1beta elevation, and labetalol, which cannot cross the blood-brain barrier, attenuated plasma IL-1beta and IL-6, blocked pituitary IL-1beta, but had no effect on central tissue IL-1beta levels. Furthermore, administration of 50.0 mg/kg N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride, a neurotoxin that lesions neural projections from the locus coeruleus, prevented stress-induced elevation in hippocampal IL-1beta, a region highly innervated by the locus coeruleus, but had no effect on hypothalamic IL-1beta, a region that receives few locus coeruleus projections. Finally, i.p. injection of 5.0 mg/kg isoproterenol (beta-adrenoceptor agonist) was sufficient to induce circulating IL-1 and IL-6, and tissue IL-1beta. These data suggest catecholamines play an important role in the induction of stress-induced proinflammatory cytokines and that beta-adrenoceptors are critical for tissue IL-1beta induction, while both alpha- and beta-adrenoceptors contribute to the induction of plasma cytokines.


Subject(s)
Brain Chemistry , Catecholamines/metabolism , Cytokines/metabolism , Stress, Psychological/physiopathology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Brain Chemistry/drug effects , Catecholamines/analysis , Cytokines/analysis , Electroshock , Immunohistochemistry , Labetalol/pharmacology , Male , Prazosin/pharmacology , Propranolol/pharmacology , Rats , Rats, Inbred F344 , Tyrosine 3-Monooxygenase/metabolism
12.
Am J Physiol Regul Integr Comp Physiol ; 289(6): R1665-74, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16081876

ABSTRACT

Physically active rats have facilitated heat shock protein 72 (Hsp72) responses after stressor exposure in both brain and peripheral tissues compared with sedentary rats. This study verifies that physically active animals do not have elevated Hsp72 levels compared with sedentary animals in the hypothalamus, pituitary, or dorsal vagal complex. We then examined whether 1) physically active rats respond more efficiently than sedentary rats to a bacterial challenge; 2) peripheral immune challenge elicits brain induction of Hsp72; 3) this induction is facilitated by prior freewheel running; and 4) Hsp72 upregulation produced by peripheral immune challenge results in a commensurate decrease in the proinflammatory cytokine IL-1beta. Adult male Fischer 344 rats were housed with either a mobile or locked running wheel. Six weeks later, rats were injected intraperitoneally with saline or Escherichia coli and killed 30 min, 2.5 h, 6 h, and 24 h later. Serum endotoxin and IL-1beta, and peritoneal fluid endotoxin and E. coli colony-forming units (CFUs) were measured. Hsp72 and IL-1beta were measured in hypothalamus, pituitary, and dorsal vagal complex. The results were that physically active rats had a faster reduction in endotoxin and E. coli CFUs and lower levels of circulating endotoxin and cytokines compared with sedentary rats. E. coli challenge elicited significantly greater time-dependent increases of both Hsp72 and IL-1beta in hypothalamus, pituitary, and dorsal vagal complex of physically active animals but not sedentary animals. Contrary to our hypothesis, increases in Hsp72 were positively correlated with IL-1beta. This study extends our findings that physical activity facilitates stress-induced Hsp72 to include immunological stressors such as bacterial challenge and suggests that brain Hsp72 and IL-1beta responses to peripheral immune challenge may contribute to exercise-mediated resistance to long-term sickness.


Subject(s)
Brain/immunology , Encephalitis/immunology , Escherichia coli Infections/immunology , HSP72 Heat-Shock Proteins/immunology , Interleukin-1/immunology , Motor Activity/immunology , Physical Exertion , Adaptation, Physiological/immunology , Animals , Encephalitis/microbiology , Male , Rats , Rats, Inbred F344
13.
Int J Hyperthermia ; 21(5): 457-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16048842

ABSTRACT

Exposure to acute physical and/or psychological stressors induces a cascade of physiological changes collectively termed the stress response. The stress response is demonstrable at the behavioural, neural, endocrine and cellular levels. Stimulation of the stress response functions to improve an organism's chance of survival during acute stressor challenge. The current review focuses on one ubiquitous cellular stress response, up-regulation of heat shock protein 72 (Hsp72). Although a great deal is known about the function of intra-cellular Hsp72 during exposure to acute stressors, little is understood about the potential function of endogenous extra-cellular Hsp72 (eHsp72). The current review will develop the hypothesis that eHsp72 release may be a previously unrecognized feature of the acute stress response and may function as an endogenous 'danger signal' for the immune system. Specifically, it is proposed that exposure to physical or psychological acute stressors stimulate the release of endogenous eHsp72 into the blood via an alpha1-adrenergic receptor-mediated mechanism and that elevated eHsp72 functions to facilitate innate immunity in the presence of bacterial challenge.


Subject(s)
Heat-Shock Proteins/physiology , Heat-Shock Response/physiology , Immunity, Innate/physiology , Animals , Bacteria/immunology , Exocytosis/physiology , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/blood , Heat-Shock Proteins/metabolism , Humans , Immunity, Innate/immunology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Models, Biological , Nitric Oxide/metabolism , Nitric Oxide/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Stress, Physiological/physiopathology , Toll-Like Receptors
14.
J Neuroimmunol ; 165(1-2): 150-60, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15978673

ABSTRACT

Exposure to an intense acute stressor immediately following immunization leads to a reduction in anti-KLH IgM, IgG, and IgG2a, but not IgG1. Stress also depletes splenic norepinephrine (NE) content. Immunization during pharmacological (alpha-methyl-p-tyrosine) or stress-induced splenic NE depletion results in antibody suppression similar to that found in rats immunized prior to stressor exposure. Prevention of splenic NE depletion during stress by tyrosine, but not pharmacological elevation (mirtazapine) of NE, resulted in normal antibody responses. These data support the hypothesis that splenic NE depletion is necessary and sufficient for stress-induced suppression of antibody to a T-cell dependent antigen.


Subject(s)
Immunoglobulin G , Immunoglobulin M , Immunosuppression Therapy , Norepinephrine/antagonists & inhibitors , Norepinephrine/metabolism , Spleen/immunology , Spleen/metabolism , Stress, Physiological/immunology , Stress, Physiological/metabolism , Animals , Catecholamines/biosynthesis , Hemocyanins/administration & dosage , Hemocyanins/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/metabolism , Immunoglobulin M/biosynthesis , Immunoglobulin M/metabolism , Immunosuppression Therapy/methods , Injections, Intraperitoneal , Male , Methyltyrosines/administration & dosage , Mianserin/administration & dosage , Mianserin/analogs & derivatives , Mirtazapine , Rats , Rats, Inbred F344 , Stress, Physiological/physiopathology , Time Factors , Tyrosine/administration & dosage
15.
J Neurosci ; 24(33): 7353-65, 2004 Aug 18.
Article in English | MEDLINE | ID: mdl-15317861

ABSTRACT

The present experiments examined the role of spinal proinflammatory cytokines [interleukin-1beta (IL-1)] and chemokines (fractalkine) in acute analgesia and in the development of analgesic tolerance, thermal hyperalgesia, and tactile allodynia in response to chronic intrathecal morphine. Chronic (5 d), but not acute (1 d), intrathecal morphine was associated with a rapid increase in proinflammatory cytokine protein and/or mRNA in dorsal spinal cord and lumbosacral CSF. To determine whether IL-1 release modulates the effects of morphine, intrathecal morphine was coadministered with intrathecal IL-1 receptor antagonist (IL-1ra). This regimen potentiated acute morphine analgesia and inhibited the development of hyperalgesia, allodynia, and analgesic tolerance. Similarly, intrathecal IL-1ra administered after the establishment of morphine tolerance reversed hyperalgesia and prevented the additional development of tolerance and allodynia. Fractalkine also appears to modulate the effects of intrathecal morphine because coadministration of morphine with intrathecal neutralizing antibody against the fractalkine receptor (CX3CR1) potentiated acute morphine analgesia and attenuated the development of tolerance, hyperalgesia, and allodynia. Fractalkine may be exerting these effects via IL-1 because fractalkine (CX3CL1) induced the release of IL-1 from acutely isolated dorsal spinal cord in vitro. Finally, gene therapy with an adenoviral vector encoding for the release of the anti-inflammatory cytokine IL-10 also potentiated acute morphine analgesia and attenuated the development of tolerance, hyperalgesia, and allodynia. Taken together, these results suggest that IL-1 and fractalkine are endogenous regulators of morphine analgesia and are involved in the increases in pain sensitivity that occur after chronic opiates.


Subject(s)
Analgesics, Opioid/pharmacology , Chemokines, CX3C/physiology , Hyperalgesia/immunology , Interleukin-1/physiology , Membrane Proteins/physiology , Morphine/pharmacology , Analgesics, Opioid/administration & dosage , Animals , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Drug Tolerance , Genetic Therapy , Hot Temperature , Hyperalgesia/therapy , Inflammation/immunology , Injections, Spinal , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/biosynthesis , Interleukin-1/cerebrospinal fluid , Interleukin-10/genetics , Male , Membrane Proteins/pharmacology , Morphine/administration & dosage , Pain/immunology , Pain Management , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytokine/antagonists & inhibitors , Receptors, HIV/antagonists & inhibitors , Sialoglycoproteins/administration & dosage , Sialoglycoproteins/pharmacology , Spinal Cord/drug effects , Spinal Cord/immunology
16.
Neuroscience ; 120(1): 269-81, 2003.
Article in English | MEDLINE | ID: mdl-12849759

ABSTRACT

Modulation of sympathetic drive to the spleen is one potential mechanism whereby physical activity prevents stress-induced splenic immune suppression in rats. The current study tested the hypothesis that voluntary freewheel running reduces peripheral sympathetic drive by modulating stress-induced activity of brain regions synaptically linked to sympathetically innervated peripheral organs, including the adrenals and spleen. To this end, adrenal and splenic catecholamine content and activity of the central sympathetic circuit indexed by c-Fos protein induction, elicited by acute exposure to inescapable tail shock, were measured. Stressor exposure depleted adrenal and splenic norepinephrine content and elicited a robust increase in c-Fos in the brains of sedentary rats. Physical activity status had no effect on adrenal norepinephrine content. Indicative of attenuated sympathetic drive to the spleen, however, 6 weeks of voluntary freewheel running diminished stress-induced splenic norepinephrine depletion, and significantly attenuated stress-induced c-Fos in specific brain regions responsible for sympathetic regulation, including tyrosine hydroxylase-immunoreactive neurons of the locus coeruleus, A5 cell group and rostral ventrolateral medulla. Results suggest that voluntary activity attenuates sympathetic drive to the spleen during stressor exposure by selectively modulating stress-induced activity of the central sympathetic circuit. The attenuation of sympathetic responses observed in this study may be one important mechanism for the protective effect of physical activity against stress-related illness and immunosuppression.


Subject(s)
Catecholamines/metabolism , Genes, fos/physiology , Motor Activity/physiology , Stress, Physiological/metabolism , Sympathetic Nervous System/metabolism , Adrenal Glands/metabolism , Animals , Brain/metabolism , Catecholamines/genetics , Gene Expression Regulation/physiology , Male , Rats , Rats, Sprague-Dawley , Spleen/metabolism
17.
J Appl Physiol (1985) ; 95(5): 1873-82, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12871965

ABSTRACT

Peripheral administration of a variety of inflammatory stimuli, such as endotoxin or cytokines, induces an orchestrated set of brain-mediated events referred to as the sickness response. The mechanism for how immune products signal the brain is not clear, but accumulating evidence supports the existence of neural as well as blood-borne pathways. Although endotoxin or cytokine administration results in sickness responses, few data exist regarding the role of circulating endotoxin or cytokines in the induction of sickness during a real bacterial infection. Thus the present studies examined whether subcutaneously administered Escherichia coli can activate sickness responses and whether circulating endotoxin and/or proinflammatory cytokines are a prerequisite for these responses. Male Sprague-Dawley rats were injected subcutaneously with one of three doses (2.5 x 10(7), 2.5 x 10(8), 2.5 x 10(9) colony-forming units) of replicating E. coli, a ubiquitous bacterial strain, or vehicle. Core body temperature (Tc) and activity were measured for 3 days after the injection. A second set of groups of animals were killed 3, 6, 12, 18, 24, and 48 h after the injection, and blood samples and brains were collected. Injections dose dependently and consistently increased Tc and decreased activity, with increases in Tc beginning 4 h after the injection. In addition, E. coli significantly increased serum interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha and brain IL-1beta levels beginning at the 6-h time point. Corticosterone and endotoxin were first elevated in the circulation at 3 and 18 h after the injection, respectively. Because fever onset preceded brain cytokine induction, we also examined cytokine levels in the serum, brain, and inflammation site 2 and 4 h after injection. Cytokines were elevated at the inflammation site but were not detectable in the serum or brain at 2 and 4 h. We conclude that subcutaneous injection of replicating E. coli induces a consistent and naturalistic infection that includes features of the sickness response as well as increases in circulating, brain, and inflammation site tissue cytokines. In addition, injection of replicating E. coli produces a robust fever and corticosterone response at a time when there are no detectable increases in circulating cytokines or endotoxin. These results suggest that elevated levels of circulating cytokines and endotoxin are not necessary for the activation of the sickness or corticosterone response. Therefore, fever, activity reduction, and corticosterone elevation induced by E. coli infection may have been evoked by a neural, rather than a humoral, pathway from the periphery to the brain.


Subject(s)
Corticosterone/blood , Escherichia coli Infections/immunology , Interleukin-1/immunology , Neuroimmunomodulation/physiology , Tumor Necrosis Factor-alpha/immunology , Animals , Brain/immunology , Brain/microbiology , Escherichia coli Infections/blood , Fever/immunology , Fever/microbiology , Interleukin-1/metabolism , Lipopolysaccharides/blood , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
18.
J Appl Physiol (1985) ; 94(1): 43-52, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12391077

ABSTRACT

Acute stress can compromise acquired, and potentiate innate, immunity. Recent evidence suggests that the impact of stress on measures of immunity can be modulated by the physical activity status of the organism and that extracellular heat shock protein 72 (eHSP72) contributes to the activation of innate immunity produced by stress. Therefore, this study investigated whether physical activity status would impact the immunologically enhancing effects of stressor exposure [inescapable tail-shock stress (IS)] on innate immunity and whether changes in eHSP72 responses could play a role. Adult, male Fischer 344 rats lived with mobile (physically active) or immobile (sedentary) running wheels. After 6 wk, rats were exposed to IS or to no stress. Immediately after IS, all rats were injected subcutaneously with live Escherichia coli. Inflammation was assessed daily, and plasma eHSP72 was measured at various time points. Rats exposed to IS resolved their inflammation faster than nonstressed rats, but the beneficial impact of stress on recovery was greater in physically active rats. All rats had equal increases in circulating eHSP72 after IS. Splenocytes harvested from a separate cohort of nonstressed rats were cultured with eHSP72, and nitric oxide and cytokines were measured. Physically active rats responded to eHSP72 stimulation in vitro with a greater nitric oxide and cytokine response than sedentary rats. Thus physically active rats both recover faster than sedentary rats after bacterial challenge + IS exposure and demonstrate potentiated cellular responses to eHSP72 activation that could be important for bacterial recovery.


Subject(s)
Extracellular Space/metabolism , Heat-Shock Proteins/metabolism , Motor Activity/physiology , Stress, Physiological/immunology , Animals , Body Weight , Cytokines/metabolism , Dermatitis/microbiology , Dermatitis/pathology , Dermatitis/physiopathology , Escherichia coli Infections , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/blood , Immunity/physiology , Male , Nitric Oxide/metabolism , Rats , Rats, Inbred F344
19.
J Appl Physiol (1985) ; 91(5): 2143-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641355

ABSTRACT

Interleukin-6 (IL-6), an important cytokine involved in a number of biological processes, is consistently elevated during periods of stress. The mechanisms responsible for the induction of IL-6 under these conditions remain uncertain. This study examined the effect of alpha-adrenergic blockade on the IL-6 response to acute and chronic high-altitude exposure in women both at rest and during exercise. Sixteen healthy, eumenorrheic women (aged 23.2 +/- 1.4 yr) participated in the study. Subjects received either alpha-adrenergic blockade (prazosin, 3 mg/day) or a placebo in a double-blinded, randomized fashion. Subjects participated in submaximal exercise tests at sea level and on days 1 and 12 at altitude (4,300 m). Resting plasma and 24-h urine samples were collected throughout the duration of the study. At sea level, no differences were found at rest for plasma IL-6 between groups (1.5 +/- 0.2 and 1.2 +/- 0.3 pg/ml for placebo and blocked groups, respectively). On acute ascent to altitude, IL-6 levels increased significantly in both groups compared with sea-level values (57 and 84% for placebo and blocked groups, respectively). After 12 days of acclimatization, IL-6 levels remained elevated for placebo subjects; however, they returned to sea-level values in the blocked group. alpha-Adrenergic blockade significantly lowered the IL-6 response to exercise both at sea level (46%) and at altitude (42%) compared with placebo. A significant correlation (P = 0.004) between resting IL-6 and urinary norepinephrine excretion rates was found over the course of time while at altitude. In conclusion, the results indicate a role for alpha-adrenergic regulation of the IL-6 response to the stress of both short-term moderate-intensity exercise and hypoxia.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Altitude , Exercise/physiology , Interleukin-6/biosynthesis , Adrenergic alpha-Agonists/pharmacology , Adult , Anaerobic Threshold/physiology , Catecholamines/urine , Female , Humans , Male , Menstrual Cycle/physiology , Ovary/metabolism , Oxygen Consumption/physiology , Phenylephrine/pharmacology
20.
J Appl Physiol (1985) ; 91(2): 821-31, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457799

ABSTRACT

The purpose of this study was to determine the effect of arousal in men and women on the moment-to-moment performance of a simple motor task. We examined the control of a precision task in the presence and absence of imposed stressors. Twenty-nine subjects (14 men, 15 women; 18--44 yr) were randomly assigned to either a control group or one of two stressor groups, Mental Math or Electric Shock. Subjects presented with Math and Shock stressors, which lasted 10 min, experienced significant increases in cognitive and physiological arousal compared with baseline and control subjects. Heart rate, systolic blood pressure, and electrodermal activity were elevated 5--80% with presentation of the stressors, whereas diastolic blood pressure and salivary cortisol were unchanged. The greater levels of cognitive and physiological arousal were associated with reductions in steadiness of a pinch grip for the Shock subjects (approximately 130% reduction from baseline) but not for the subjects in the Math group, who experienced heightened arousal but no change in steadiness (10% reduction from baseline). Although women exhibited more of a reduction in steadiness than men, the effect was largely unrelated to the magnitude of the change in arousal.


Subject(s)
Arousal/physiology , Cognition/physiology , Psychomotor Performance/physiology , Stress, Psychological/physiopathology , Adult , Analysis of Variance , Anxiety/physiopathology , Blood Pressure , Electroshock , Female , Hand Strength , Heart Rate , Humans , Male , Mathematics , Mental Status Schedule , Reference Values , Sex Characteristics , Skin/innervation , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...