Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Endocr Regul ; 53(2): 100-109, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-31517627

ABSTRACT

OBJECTIVES: Despite extensive research efforts, mechanisms participating on development of Alzheimer's disease (AD) are covered only partially. Data from the last decades indicate that various stressors, as etiological factors, may play a role of in the AD. Therefore, we investigated the effect of two acute stressors, immobilization (IMO) and lipopolysaccharide (LPS), on the AD-related neuropathology. METHODS: Adult C57BL/6J mice males were exposed to a single IMO stress or a single intraperitoneal injection of LPS (250 µg/kg body weight). After terminating the experiments, the brains were removed and their cortices isolated. Gene expression of pro-inflammatory cytokines, as well as expression of genes implicated in the AD neuropathology were determined. In addition, mediators related to the activation of the microglia, monocytes, and perivascular macrophages were determined in brain cortices, as well. RESULTS: In comparison with the control animals, we found increased gene expression of proinflammatory mediators in mice brain cortex in both IMO and LPS groups. In stressed animals, we also showed an increased expression of genes related to the AD neuropathology, as well as positive correlations between genes implicated in AD development and associated neuroinflammation. CONCLUSIONS: Our data indicate that acute exposure to a strong IMO stressor, composed of the combined physical and psychological challenges, induces similar inflammatory and other ADrelated neuropathological changes as the immune LPS treatment. Our data also indicate that cytokines are most likely released from the peripheral immune cells, as we detected myeloid cells activity, without any microglia response. We hypothesize that stress induces innate immune response in the brain that consequently potentiate the expression of genes implicated in the AD-related neuropathology.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Inflammation , Lipopolysaccharides , Restraint, Physical/adverse effects , Stress, Psychological/genetics , Acute Disease , Alzheimer Disease/metabolism , Animals , Brain/pathology , Cytokines/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Restraint, Physical/psychology , Stress, Psychological/etiology , Up-Regulation/genetics
2.
J Neuroendocrinol ; : e12595, 2018 Mar 31.
Article in English | MEDLINE | ID: mdl-29604138

ABSTRACT

Corticotropin-releasing factor is well known activator of the hypothalamic-pituitary-adrenocortical axis, that represents crucial system participating on stress response of the organism. Urocortins are members of the corticotropin-releasing factor family of peptides with proposed effects on neuroendocrine and behavioral stress response mechanisms. Urocortin 2, one of three known urocortins, is present in central and peripheral stress response system and its expression can be augmented by glucocorticoids. In the present study we have examined how glucocorticoid withdrawal affects urocortin 2 gene expression after acute immobilization in the adrenal medulla and selected brain areas in rats. We used pharmacological adrenalectomy to block synthesis of corticosterone. Our results show that the immobilization-induced rise in urocortin 2 mRNA levels in rat adrenal medulla was not inhibited by glucocorticoid withdrawal. On the other hand, observed changes in the brain indicate that the effect of stress and pharmacological adrenalectomy on urocortin 2 gene expression is site-specific. While in the paraventricular nucleus and locus coeruleus the immobilization induced rise of urocortin 2 was not inhibited by pharmacological adrenalectomy in the arcuate nucleus and central amygdala it was. Moreover, we have seen a significant depletion of urocortin 2 plasma levels after immobilization. The immobilization induced rise of urocortin 2 gene expression in rat adrenal medulla and brain areas regulating stress response pathways and preservation of its induction after adrenalectomy suggests a role of urocortin 2 in the neuroendocrine stress response of an organism. This article is protected by copyright. All rights reserved.

3.
Cell Mol Neurobiol ; 38(1): 163-169, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28993972

ABSTRACT

The activation of the HPA axis is the endocrine measure of stress responsiveness that is initiated by corticotropin-releasing hormone (CRH). CRH exerts its effects via CRHR1 and CRH-R2 receptors coupled to the cAMP signaling system and this process involves transcription factor cAMP-responsive element-binding protein (CREB).This study investigated the role of CRH and the possible involvement of CREB in gene regulation of CRH receptor, under basal conditions and after stress application in the pituitary. We used wild type (wt +/+) controls and CRH knock-out (CRH-KO -/-) male mice. Using CRH-deficient mice, we were able to investigate the consequences of the lack of the CRH on the expression of CRH receptors and transcriptional regulation mediated by CREB. We estimated the effect of acute (IMO 1×) and repeated (IMO 7×) restraint stressors lasting 30 and 120 min on the expression of mRNA CREB, CRH-R1, and CRH-R2 by qPCR. We found very significant difference in the expression of these peptides under the effect of single and repeated stress in control and CRH-KO mice. Our results indicate that both CRH receptors and CREB might be involved in the regulation of stress response in the pituitary of mice. We propose that regulation of the stress response may be better understood if more were known about the mechanisms of CRH receptor signal transduction and involvement of CREB system.


Subject(s)
Corticotropin-Releasing Hormone/biosynthesis , Pituitary Gland/metabolism , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Stress, Psychological/metabolism , Acute Disease , Animals , Corticotropin-Releasing Hormone/deficiency , Cyclic AMP Response Element-Binding Protein/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stress, Psychological/psychology
4.
Cell Mol Neurobiol ; 38(1): 243-259, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28405903

ABSTRACT

Alzheimer's disease (AD) is a multifactorial disorder; neurofibrillary pathology composed of tau protein is found side by side with amyloid-ß deposits and extensive neuroinflammation. The immune system of the brain is considered as one of the factors that could influence the speed of the progression of AD neuropathology as a potential mediator of the damage induced by AD protein deposits. Alzheimer's disease pathology can be impacted by psychological stress; however, signalling pathways in background are not well known. We have explored possible avenues of how stress could influence the brain's immune system in a rat model of AD. Animals were subjected either to a single or multiple instances of immobilization stress. The analysis of a panel of immunity-related genes was used to evaluate the impact of stress on the immune response in the brain. We have identified 19 stress-responsive genes that are involved in neuroinflammation accompanying tau pathology: Nos2, Ptgs2, IL-8rb, C5, Mmp9, Cx3cr1, CD40lg, Adrb2, IL-6, IL-6r, IL-1r2, Ccl2, Ccl3, Ccl4, Ccl12, TNF-α, IL-1α, IL-1ß, IL-10. Most of them are deregulated under the stress conditions also in control animals; however, the magnitude of the response to either acute or chronic stress differs. This can lead to serious influence, most probably to acceleration of neurodegenerative phenotype in diseased animals. Several of the genes (IL-1ß, Casp1, Cx3cr1 and C5) are deregulated solely in tauopathic animals. The stress-induced changes in the inflammatory picture of the brain highlight the fact that the brain's immune response is highly responsive to environmental stimuli. The pattern of changes is indicative of an attempt to protect the brain in the short term, while being potentially detrimental to the response against a long-term pathological process such as neurofibrillary degeneration.


Subject(s)
Brain/immunology , Immunity, Cellular/physiology , Neurodegenerative Diseases/immunology , Stress, Psychological/immunology , tau Proteins/immunology , Animals , Brain/metabolism , Female , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/psychology , Rats , Rats, Inbred SHR , Rats, Transgenic , Stress, Psychological/metabolism , Stress, Psychological/psychology , tau Proteins/metabolism
5.
Stress ; 20(1): 36-43, 2017 01.
Article in English | MEDLINE | ID: mdl-27873537

ABSTRACT

Brain acetylcholinesterase (AChE) variant AChER expression increases with acute stress, and this persists for an extended period, although the timing, strain and laterality differences, have not been explored previously. Acute stress transiently increases acetylcholine release, which in turn may increase activity of cholinesterases. Also the AChE gene contains a glucocorticoid response element (GRE), and stress-inducible AChE transcription and activity changes are linked to increased glucocorticoid levels. Corticotropin-releasing hormone knockout (CRH-KO) mice have basal glucocorticoid levels similar to wild type (WT) mice, but much lower levels during stress. Hence we hypothesized that CRH is important for the cholinesterase stress responses, including butyrylcholinesterase (BChE). We used immobilization stress, acute (30 or 120 min) and repeated (120 min daily × 7) in 48 male mice (24 WT and 24 CRH-KO) and determined AChER, AChE and BChE mRNA expression and AChE and BChE activities in left and right brain areas (as cholinergic signaling shows laterality). Immobilization decreased BChE mRNA expression (right amygdala, to 0.5, 0.3 and 0.4, × control respectively) and AChER mRNA expression (to 0.5, 0.4 and 0.4, × control respectively). AChE mRNA expression increased (1.3, 1.4 and 1.8-fold, respectively) in the left striatum (Str). The AChE activity increased in left Str (after 30 min, 1.2-fold), decreased in right parietal cortex with repeated stress (to 0.5 × control). BChE activity decreased after 30 min in the right CA3 region (to 0.4 × control) but increased (3.8-fold) after 120 min in the left CA3 region. The pattern of changes in CRH-KO differed from that in WT mice.


Subject(s)
Acetylcholinesterase/metabolism , Brain/metabolism , Butyrylcholinesterase/metabolism , Functional Laterality/physiology , Stress, Physiological/physiology , Stress, Psychological/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Male , Mice , Mice, Knockout , Restraint, Physical
6.
Stress ; 19(4): 395-405, 2016 07.
Article in English | MEDLINE | ID: mdl-27484105

ABSTRACT

Neuroendocrine and behavioral stress responses are orchestrated by corticotropin-releasing hormone (CRH) and norepinephrine (NE) synthesizing neurons. Recent findings indicate that stress may promote development of neurofibrillary pathology in Alzheimer's disease. Therefore, we investigated relationships among stress, tau protein phosphorylation, and brain NE using wild-type (WT) and CRH-knockout (CRH KO) mice. We assessed expression of phosphorylated tau (p-tau) at the PHF-1 epitope and NE concentrations in the locus coeruleus (LC), A1/C1 and A2/C2 catecholaminergic cell groups, hippocampus, amygdala, nucleus basalis magnocellularis, and frontal cortex of unstressed, singly stressed or repeatedly stressed mice. Moreover, gene expression and protein levels of tyrosine hydroxylase (TH) and CRH receptor mRNA were determined in the LC. Plasma corticosterone levels were also measured. Exposure to a single stress increases tau phosphorylation throughout the brain in WT mice when compared to singly stressed CRH KO animals. In contrast, repeatedly stressed CRH KO mice showed exaggerated tau phosphorylation relative to WT controls. We also observed differences in extent of tau phosphorylation between investigated structures, e.g. the LC and hippocampus. Moreover, CRH deficiency leads to different responses to stress in gene expression of TH, NE concentrations, CRH receptor mRNA, and plasma corticosterone levels. Our data indicate that CRH effects on tau phosphorylation are dependent on whether stress is single or repeated, and differs between brain regions. Our findings indicate that CRH attenuates mechanisms responsible for development of stress-induced tau neuropathology, particularly in conditions of chronic stress. However, the involvement of central catecholaminergic neurons in these mechanisms remains unclear and is in need of further investigation.


Subject(s)
Brain/metabolism , Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Stress, Psychological/metabolism , tau Proteins/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Gene Expression , Male , Mice , Mice, Knockout , Phosphorylation , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Restraint, Physical
7.
J Neuroinflammation ; 13: 15, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26792515

ABSTRACT

BACKGROUND: Brain norepinephrine (NE) plays an important role in the modulation of stress response and neuroinflammation. Recent studies indicate that in Alzheimer's disease (AD), the tau neuropathology begins in the locus coeruleus (LC) which is the main source of brain NE. Therefore, we investigated the changes in brain NE system and also the immune status under basal and stress conditions in transgenic rats over-expressing the human truncated tau protein. METHODS: Brainstem catecholaminergic cell groups (LC, A1, and A2) and forebrain subcortical (nucleus basalis of Meynert), hippocampal (cornu ammonis, dentate gyrus), and neocortical areas (frontal and temporal association cortices) were analyzed for NE and interleukin 6 (IL-6) mRNA levels in unstressed rats and also in rats exposed to single or repeated immobilization. Moreover, gene expression of NE-biosynthetic enzyme, tyrosine hydroxylase (TH), and several pro- and anti-inflammatory mediators were determined in the LC. RESULTS: It was found that tauopathy reduced basal NE levels in forebrain areas, while the gene expression of IL-6 was increased in all selected areas at the same time. The differences between wild-type and transgenic rats in brain NE and IL-6 mRNA levels were observed in stressed animals as well. Tauopathy increased also the gene expression of TH in the LC. In addition, the LC exhibited exaggerated expression of pro- and anti-inflammatory mediators (IL-6, TNFα, inducible nitric oxide synthases 2 (iNOS2), and interleukin 10 (IL-10)) in transgenic rats suggesting that tauopathy affects also the immune background in LC. Positive correlation between NE and IL-6 mRNA levels in cornu ammonis in stressed transgenic animals indicated the reduction of anti-inflammatory effect of NE. CONCLUSIONS: Our data thus showed that tauopathy alters the functions of LC further leading to the reduction of NE levels and exaggeration of neuroinflammation in forebrain. These findings support the assumption that tau-related dysfunction of LC activates the vicious circle perpetuating neurodegeneration leading to the development of AD.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , Encephalitis/etiology , Norepinephrine/metabolism , Tauopathies , Analysis of Variance , Animals , Brain/metabolism , Brain/pathology , Cytokines/genetics , Cytokines/metabolism , Gene Expression/genetics , Humans , Male , Microdissection , Nitric Oxide Synthase Type II/metabolism , Norepinephrine/genetics , RNA, Messenger/metabolism , Rats , Rats, Transgenic , Tauopathies/complications , Tauopathies/genetics , Tauopathies/pathology , Tyrosine 3-Monooxygenase/metabolism
8.
Eur J Neurosci ; 42(2): 1872-86, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25994480

ABSTRACT

Brainstem catecholaminergic neurons significantly participate in the regulation of neuroendocrine system activity, particularly during stressful conditions. However, so far the precise quantitative characterisation of basal and stress-induced changes in gene expression and protein levels of catecholaminergic biosynthetic enzymes in these neurons has been missing. Using a quantitative reverse transcription-polymerase chain reaction method, we investigated gene expression of catecholamine biosynthetic enzymes in brainstem noradrenergic and adrenergic cell groups in rats under resting conditions as well as in acutely and repeatedly stressed animals. For the first time, we described quantitative differences in basal levels of catecholamine biosynthetic enzyme mRNA in brainstem catecholaminergic ascending and descending projecting cell groups. Moreover, we found and defined some differences among catecholaminergic cell groups in the time-course of mRNA levels of catecholaminergic enzymes following a single and especially repeated immobilisation stress. The data obtained support the assumption that brainstem catecholaminergic cell groups represent a functionally differentiated system, which is highly (but specifically) activated in rats exposed to stress. Therefore, potential interventions for the treatment of stress-related diseases need to affect the activity of brainstem catecholaminergic neurons not uniformly but with some degree of selectivity.


Subject(s)
Brain Stem/metabolism , Catecholamines/metabolism , Dopamine beta-Hydroxylase/metabolism , Gene Expression Regulation, Enzymologic/physiology , Immobility Response, Tonic/physiology , Phenylethanolamine N-Methyltransferase/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Dopamine beta-Hydroxylase/genetics , Male , Microdissection , Phenylethanolamine N-Methyltransferase/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Tyrosine 3-Monooxygenase/genetics
9.
J Alzheimers Dis ; 43(4): 1157-61, 2015.
Article in English | MEDLINE | ID: mdl-25147110

ABSTRACT

Stress may accelerate onset of neurodegenerative diseases in vulnerable subjects and, vice versa, neurodegeneration affects the responsiveness to stressors. We investigated the neuroendocrine response to immobilization stress in normotensive Wistar-Kyoto rats (WKY), spontaneously hypertensive rats (SHR), and transgenic rats of respective WKY and SHR strains overexpressing human truncated tau protein. Plasma levels of epinephrine, norepinephrine, and corticosterone were determined. An immobilization-induced elevation of epinephrine and norepinephrine was significantly reduced in WKY transgenic rats compared to WKY wild-type rats, while no differences were seen between SHR transgenic and SHR wild-type animals. Our data have shown that sympathoadrenal system response to stress strongly depends on both tau protein-induced neurodegeneration and genetic background of experimental animals.


Subject(s)
Genetic Predisposition to Disease , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Tauopathies/genetics , Tauopathies/physiopathology , Animals , Corticosterone/blood , Disease Models, Animal , Epinephrine/blood , Norepinephrine/blood , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Transgenic , Restraint, Physical , Species Specificity , tau Proteins/genetics , tau Proteins/metabolism
10.
Gen Physiol Biophys ; 33(3): 357-64, 2014.
Article in English | MEDLINE | ID: mdl-24968410

ABSTRACT

The left and right ventricles fulfill different role in heart function. Here we compare chamber specific changes in local catecholamine concentrations; gene expression and the receptor protein amount of all three ß-adrenoceptors (ß-AR) in rat right heart ventricles exposed to acute (1 session) and repeated (7 sessions) immobilization stress (IMMO) vs. previously observed changes in left ventricles. Density of muscarinic receptors as main cardio-inhibitive receptors was also measured. In the right ventricles, noradrenaline and adrenaline were increased. No ß1-AR changes were observed, in spite of the increased sympathetic activity. On the other hand, we have found a decrease of ß2-AR gene expression (reduction to 30%) after 7 IMMO and protein (to 59%) after 1 IMMO. ß3-AR gene expression was increased after 7 IMMO. Muscarinic receptor density was not changed. When comparing correlation in left and right ventricles, there was strong correlation between adrenaline and ß2-AR gene expression, protein and ß3-AR gene expression in the left ventricles while only correlation between adrenaline and ß2-AR mRNA and protein in the right ventricles was found. Our results show that maintenance of cardiac homeostasis under stress conditions are to a great extent achieved by a balance between different receptors and also by a balanced receptor changes in left vs. right ventricles. Taken together, decrease of cardio-stimulating ß2-AR represents a new important mechanism by which ß2-AR contributes to the heart physiology.


Subject(s)
Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Receptors, Adrenergic, beta/metabolism , Receptors, Muscarinic/metabolism , Stress, Physiological , Animals , Binding Sites , Catecholamines/biosynthesis , Epinephrine/biosynthesis , Gene Expression Profiling , Gene Expression Regulation , Male , Norepinephrine/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Restraint, Physical
11.
Stress Health ; 30(4): 301-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23878066

ABSTRACT

Stress is a contributor of many cardiovascular diseases. Positive inotropic and chronotropic effects of catecholamines are regulated via ß-adrenergic receptors (ARs). Many reports exist concerning changes of cardiac ß1 - and ß2 -ARs in stress, but only a few deal with modulation of cardiac ß3 -AR. Our aim was to analyze the expression and binding sites of ß1 -, ß2 - and ß3 -ARs and adenylyl cyclase activity in the left ventricle, and ß3 -AR expression and binding in the left atrium of rats exposed to acute and chronic immobilization stress (IMO). The concentration of noradrenaline in the ventricle decreased, while adrenaline increased, especially after repeated IMO. The mRNA and protein levels, and binding sites of ß3 -subtype significantly rose following chronic IMO, while all parameters for ß2 -AR dropped after single and repeated exposure. Similarly, the mRNA levels and binding sites for ß3 -subtype increased in the left atrium as a consequence of chronic IMO. The rise in ß3 -subtypes and a drop in ß2 -subtypes resulted in inhibition of adenylyl cyclase activity within the left ventricle. Taken together, among other factors, up-regulation of ß3 -AR could represent an adaptation mechanism, which might be related to altered physiological function of the left ventricle and atrium during prolonged emotional stress and might serve cardioprotective function during catecholamine overload.


Subject(s)
Adenylyl Cyclases/metabolism , Heart Ventricles/metabolism , Receptors, Adrenergic, beta/metabolism , Stress, Psychological/metabolism , Animals , Blotting, Western , Epinephrine/metabolism , Heart Atria/metabolism , Male , Norepinephrine/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
12.
Endocrinology ; 154(10): 3729-38, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24025224

ABSTRACT

Bradykinin, acting via the bradykinin B2 receptor (B2R), is a potent stimulator of adrenomedullary catecholamine biosynthesis and release and likely plays an important role in the adrenomedullary stress response. However, the effects of stress on the expression of this receptor in the adrenal medulla are currently unclear. Here, we examined the changes in adrenomedullary B2R gene expression in male rats in response to single (1 time) and repeated (6 times) exposure to 2 hours immobilization stress (IMO). Immediately after 1 or 6 times IMO, B2R mRNA levels were increased by 9-fold and 7-fold, respectively, and returned to unstressed control levels 3 hours later. This large, but transient, increase in mRNA elicited a doubling of protein levels 3 hours after the stress exposure. Next, the role of the hypothalamic-pituitary-adrenocortical axis in the stress-induced upregulation of B2R gene expression was examined. Treatment with endogenous (corticosterone) and synthetic (dexamethasone) glucocorticoids dose-dependently increased B2R mRNA levels in adrenomedullary-derived PC12 cells. Furthermore, cortisol supplementation at levels mimicking stress exposure elevated B2R mRNA levels in the adrenal medulla of hypophysectomized rats. In response to 1 exposure to IMO, the stress-triggered rise in plasma corticosterone and adrenomedullary B2R mRNA levels was attenuated in CRH-knockout mice and absent in pharmacologically adrenalectomized rats, indicating a requirement for glucocorticoids in the upregulation of B2R gene expression with stress. Overall, the increase in B2R gene expression in response to the stress-triggered rise in glucocorticoids likely enhances catecholamine biosynthesis and release and may serve as an adaptive response of the adrenomedullary catecholaminergic system to stress.


Subject(s)
Adrenal Medulla/metabolism , Glucocorticoids/metabolism , Receptor, Bradykinin B2/biosynthesis , Stress, Physiological , Stress, Psychological/metabolism , Up-Regulation , Adrenal Medulla/drug effects , Animals , Corticosterone/blood , Corticosterone/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacology , Hydrocortisone/administration & dosage , Hydrocortisone/metabolism , Hypophysectomy , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice , Mice, Knockout , PC12 Cells , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Restraint, Physical , Stress, Psychological/blood , Stress, Psychological/physiopathology , Up-Regulation/drug effects
13.
Adv Pharmacol ; 68: 359-97, 2013.
Article in English | MEDLINE | ID: mdl-24054153

ABSTRACT

The sympathetic nervous system not only regulates cardiovascular and metabolic responses to stress but also is altered by stress. The sympathoneural and sympathoadrenomedullary systems are modified by different metabolic pathways and have different responses to short- and to long-term stressors. Stress also induces nonneuronal catecholamine enzymes, primarily through corticosteroids. Catecholamine synthetic enzymes are induced by different pathways in response to short- and long-term acting stressors, like cold exposure or immobilization, and differently in the sympathetic ganglia and the adrenal medulla. However, a long-term exposure to one stressor can increase the response to a second, different stressor. Tyrosine hydroxylase gene transcription increases after only 5min of immobilization through phosphorylation of CREB, but this response is short lived. However, repeated stress gives a longer-lived response utilizing transcription factors such as Egr-1 and Fra-2. Glucocorticoids and ACTH also induce sympathoneural enzymes leading to distinct patterns of short-term and long-lived activation of the sympathetic nervous system. Nonneuronal phenylethanolamine N-methyltransferase (PNMT) develops early in the heart and then diminishes. However, intrinsic cardiac adrenergic cells remain and nonneuronal PNMT is present in many cells of the adult organism and increases in response to glucocorticoids. Both stress-induced and administered glucocorticoids induce fetal PNMT and hypertension. Human stressors such as caring for an ill spouse or sleep apnea cause a persistent increase in blood norepinephrine, increased blood pressure, and downregulated catecholamine receptors. Hypertension is associated with a loss of slow-wave sleep, when sympathetic nerve activity is lowest. These findings indicate that stress-induced alteration of the sympathetic nervous system occurs in man as in experimental animals.


Subject(s)
Catecholamines/physiology , Peripheral Nervous System/physiology , Stress, Physiological/physiology , Stress, Psychological/metabolism , Animals , Humans , Stress, Psychological/physiopathology
14.
PLoS One ; 8(3): e60214, 2013.
Article in English | MEDLINE | ID: mdl-23555928

ABSTRACT

The orexigenic neuropeptide melanin-concentrating hormone (MCH), a product of Pmch, is an important mediator of energy homeostasis. Pmch-deficient rodents are lean and smaller, characterized by lower food intake, body-, and fat mass. Pmch is expressed in hypothalamic neurons that ultimately are components in the sympathetic nervous system (SNS) drive to white and interscapular brown adipose tissue (WAT, iBAT, respectively). MCH binds to MCH receptor 1 (MCH1R), which is present on adipocytes. Currently it is unknown if Pmch-ablation changes adipocyte differentiation or sympathetic adipose drive. Using Pmch-deficient and wild-type rats on a standard low-fat diet, we analyzed dorsal subcutaneous and perirenal WAT mass and adipocyte morphology (size and number) throughout development, and indices of sympathetic activation in WAT and iBAT during adulthood. Moreover, using an in vitro approach we investigated the ability of MCH to modulate 3T3-L1 adipocyte differentiation. Pmch-deficiency decreased dorsal subcutaneous and perirenal WAT mass by reducing adipocyte size, but not number. In line with this, in vitro 3T3-L1 adipocyte differentiation was unaffected by MCH. Finally, adult Pmch-deficient rats had lower norepinephrine turnover (an index of sympathetic adipose drive) in WAT and iBAT than wild-type rats. Collectively, our data indicate that MCH/MCH1R-pathway does not modify adipocyte differentiation, whereas Pmch-deficiency in laboratory rats lowers adiposity throughout development and sympathetic adipose drive during adulthood.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation/physiology , Hypothalamic Hormones/deficiency , Melanins/deficiency , Pituitary Hormones/deficiency , 3T3-L1 Cells , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Hypothalamic Hormones/genetics , Melanins/genetics , Mice , Pituitary Hormones/genetics , Rats , Sympathetic Nervous System/cytology , Sympathetic Nervous System/metabolism
15.
Brain Res Bull ; 94: 30-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23395867

ABSTRACT

Although c-Fos plays a key role in intracellular signalling, the disruption of the c-fos gene has only minor consequences on the central nervous system (CNS) function. As muscarinic receptors (MR) play important roles in many CNS functions (attention, arousal, and cognition), the c-fos knock-out might be compensated through MR changes. The aim of this study was to evaluate changes in the M1-M5 MR mRNA in selected CNS areas: frontal, parietal, temporal and occipital cortex, striatum, hippocampus, hypothalamus and cerebellum (FC, PC, TC, OC, stria, hip, hypo, and crbl, respectively). Knocking out the c-fos gene changed the expression of MR in FC (reduced M1R, M4R and M5R expression), TC (increased M4R expression), OC (decreased M2R and M3R expression) and hippocampus (reduced M3R expression). Moreover, gender differences were observed in WT mice: increased expression of all M1-M5R in the FC in males and M1-M4R in the striatum in females. A detailed analysis of MR transcripts showed pre-existing correlations in the amount of MR-mRNA between specific regions. WT mice showed three major types of cortico-cortical correlations: fronto-occipital, temporo-parietal and parieto-occipital. The cortico-subcortical correlations involved associations between the FC, PC, TC and striatum. In KO mice, a substantial rearrangement of the correlation pattern was observed: only a temporo-parietal correlation and correlations between the FC and striatum remained, and a new correlation between the hypothalamus and cerebellum appeared. Thus, in addition to the previously described dopamine receptor restructuring, the restructuring of MR mRNA correlations reveals an additional mechanism for adaptation to the c-fos gene knockout.


Subject(s)
Brain/metabolism , Gene Expression Regulation/genetics , Genes, fos , Receptors, Muscarinic/biosynthesis , Animals , Female , Genes, fos/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger , Real-Time Polymerase Chain Reaction , Receptors, Muscarinic/genetics , Sex Characteristics , Transcription, Genetic , Transcriptome
16.
Cell Mol Neurobiol ; 33(4): 503-11, 2013 May.
Article in English | MEDLINE | ID: mdl-23430272

ABSTRACT

Glucocorticoids act via glucocorticoid receptors (GR), typically localized in the cytosol (cGR). Rapid action is probably mediated via membrane receptors (mGR). In corticotropin-releasing hormone knockouts (CRH-KO), basal plasma glucocorticoid levels do differ from wild type levels (WT), but are approximately ten times lower during exposure to immobilization stress (IMMO) in comparison to WT. We tested the following hypotheses: (1) the mice lung tissue GR basal numbers would not be changed in CRH-KO (because of similar glucocorticoid levels), (2) the number of GR would be changed in WT but not in KO during short (30, 90, and 120 min) IMMO (because of higher increase of glucocorticoid levels in WT). The basal levels of cGR were not changed in CRH-KO (compared to WT), while mGR were significantly lower (62 %) in CRH-KO. In WT, there was the only decrease (to 32 %) in cGR after 120 min when we also found an increase in mGR in WT (to 201 %). In CRH-KO, IMMO caused gradual decrease in cGR (to 52 % after 30 min, to 46 % after 90 min, and to 32 % after 120 min). In CRH-KO, the only increase in mGR appeared already at 30 min of IMMO. These data suggest, on the contrary to our hypotheses, that CRH-KO are more susceptible to GR changes in early phases of stress.


Subject(s)
Cell Membrane/metabolism , Corticotropin-Releasing Hormone/metabolism , Cytosol/metabolism , Glucocorticoids/metabolism , Immobilization , Lung/metabolism , Stress, Physiological , Animals , Binding Sites , Blotting, Western , Dexamethasone/metabolism , Kinetics , Mice , Mice, Knockout , Receptors, Glucocorticoid/metabolism
17.
J Neurochem ; 125(2): 185-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23320836

ABSTRACT

The corticotropin-releasing hormone (CRH) family regulates the endocrine stress response. Here, we examined the effect of immobilization stress (IMO) on gene expression of adrenomedullary CRH family members. Urocortin 2 (Ucn2) has the highest basal gene expression and is increased by > 30-fold in response to single IMO and about 10-fold after six daily repeated IMO. IMO also induced a smaller rise in CRH (six-fold) and CRH receptor type 1 (CRHR1; two-fold with single IMO). The influence of glucocorticoids was examined. Dexamethasone (DEX) or corticosterone greatly increased Ucn2 mRNA levels in PC12 cells in a dose-dependent manner. The DEX elicited rise in Ucn2 was abolished by actinomycin D pre-treatment, indicating a transcriptionally mediated response. DEX also triggered a rise in CRHR1 and lowered CRH mRNA levels. In CRH-knockout mice, where the IMO-induced rise in corticosterone was attenuated, the response of IMO on Ucn2, as well as CRHR2 mRNAs was absent. Overall, the results suggest that the stress-triggered rise in glucocorticoids is involved in the large induction of Ucn2 mRNA levels by IMO, which may allow Ucn2 to act in an autocrine/paracrine fashion to modulate adrenomedullary function, or act as an endocrine hormone.


Subject(s)
Adrenal Medulla/metabolism , Corticotropin-Releasing Hormone/biosynthesis , Glucocorticoids/metabolism , Stress, Psychological/metabolism , Urocortins/biosynthesis , Animals , Gene Expression Regulation , Male , Mice, Knockout , Peptides , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Restraint, Physical
18.
Immunobiology ; 218(5): 780-9, 2013 May.
Article in English | MEDLINE | ID: mdl-22999161

ABSTRACT

OBJECTIVES: Stress is accompanied also by a rise in splenic catecholamines (CAs). However, indications about endogenous CA production in the spleen exist but there are no data about the cellular source of this production and possible modification by stress. Therefore, our aim was to investigate whether splenic T- and B-cells are one of main sources in the spleen expressing tyrosine hydroxylase (TH), enzyme crucial for CA biosynthesis, and phenylethanolamine N-methyltransferase (PNMT) which is necessary for epinephrine production. We also investigated whether stress is able to modify expression of both enzymes and CA levels within these cell fractions as well as tried to explain functional consequences of changes observed. RESULTS: T-cells contain higher levels of TH mRNA than B-cells although protein levels appeared similar. On contrary, the PNMT mRNA and protein were higher in B-cells, which appeared to be the main source of PNMT in the spleen. T-cells increased TH and PNMT expression after acute stress while similar rise was observed in B-cells after repeated stress, most probably as a consequence of higher CA turnover in both cell populations. The rise in TH and PNMT was accompanied by an elevation of Bax/Bcl-2 mRNA ratio, number of apoptotic cells and also by a decline of IFN-γ mRNA in both cell types. Reduction of IL-2 and IL-4 mRNA was also observed in B-cells. CONCLUSION: Stress-induced stimulation of endogenous CA biosynthesis in lymphocytes is dependent on the type of lymphocyte population and duration of stressor and leads to attenuated IFN-γ expression and induction of apoptosis. These changes might contribute to dysregulation of specific immune functions involving T- and B-cells and may decrease the ability to cope with intracellular agents following stress situations.


Subject(s)
B-Lymphocytes/metabolism , Dopamine/biosynthesis , Epinephrine/biosynthesis , Norepinephrine/biosynthesis , Spleen/metabolism , Stress, Physiological , T-Lymphocytes/metabolism , Animals , B-Lymphocytes/cytology , Gene Expression , Immobilization , Male , Organ Specificity , Phenylethanolamine N-Methyltransferase/genetics , Phenylethanolamine N-Methyltransferase/metabolism , Rats , Rats, Sprague-Dawley , Spleen/cytology , T-Lymphocytes/cytology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Stress ; 16(3): 340-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23035889

ABSTRACT

Catecholamines (CATs), the major regulator of lipolysis in adipose tissue, are produced mainly by the sympathoadrenal system. However, recent studies report endogenous CAT production in adipocytes themselves. This study investigated the effects of single and repeated (7-14 times) immobilization (IMO) stress on CAT production in various fat depots of the rat. Single IMO quickly induced a rise of norepinephrine (NE) and epinephrine (EPI) concentration in mesenteric and brown adipose depots. Adaptive response to repeated IMO included robust increases of NE and EPI levels in mesenteric and subcutaneous adipose tissue. These changes likely reflect the activation of sympathetic nervous system in fat depots by IMO. However, this process was also paralleled by an increase in tyrosine hydroxylase gene expression in mesenteric fat, suggesting regulation of endogenous CAT production in adipose tissue cells. Detailed time-course analysis (time course 10, 30, and 120 min) clearly showed that repeated stress led to increased CAT biosynthesis in isolated mesenteric adipocytes resulting in gradual accumulation of intracellular EPI during IMO exposure. Comparable changes were also found in stromal/vascular fractions, with more pronounced effects of single than repeated IMO. The potential physiological importance of these findings is accentuated by parallel increase in expression of vesicular monoamine transporter 1, indicating a need for CAT storage in adipocyte vesicles. Taken together, we show that CAT production occurs in adipose tissue and may be activated by stress directly in adipocytes.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Catecholamines/metabolism , Restraint, Physical/psychology , Stress, Psychological/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/innervation , Animals , Epinephrine/metabolism , Gene Expression Regulation, Enzymologic , Male , Mesentery , Norepinephrine/metabolism , Phenylethanolamine N-Methyltransferase/genetics , Phenylethanolamine N-Methyltransferase/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/etiology , Stromal Cells/metabolism , Subcutaneous Fat/metabolism , Time Factors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation , Vesicular Monoamine Transport Proteins/genetics , Vesicular Monoamine Transport Proteins/metabolism
20.
Gen Physiol Biophys ; 31(3): 247-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23047937

ABSTRACT

The pain-induced activation of the sympatho-adrenal system is modulated by several brain areas, including brainstem catecholamine cell groups. In the present study, we evaluated the effect of bilateral lesions of the A5 or A7 cell groups or bilateral transections of brainstem catecholaminergic pathways on plasma catecholamine levels in Sprague-Dawley rats injected subcutaneously by formalin or saline. Plasma levels of both epinephrine and norepinephrine were slightly elevated after formalin injections within 15-30 min in rats with or without lesions of the A7 catecholamine cell group. However, saline but not formalin elicited a significant increase in plasma epinephrine level in both sham-operated and A5-lesioned groups. It is more likely, that formalin blocks the effect of the handling and the painful injection procedure. In rats with bilateral partial transections of the lower brainstem, formalin was more effective than saline in the elevation of plasma epinephrine and norepinephrine levels at several time-points through the investigation period. Our data indicate the involvement of A5 and A7 norepinephrine neurons and brainstem catecholaminergic pathways in the regulation of the activity of the sympatho-adrenal system during acute painful situations. Their modulatory effect, however, seems to be a very rapid one, short and moderate.


Subject(s)
Brain Stem/metabolism , Epinephrine/blood , Formaldehyde/administration & dosage , Norepinephrine/blood , Pain/chemically induced , Pain/physiopathology , Adrenergic Neurons , Animals , Brain Stem/surgery , Injections, Subcutaneous , Male , Neural Pathways/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...