Beta adrenergic blockade decreases the immunomodulatory effects of social disruption stress

M L Hanke, N D Powell, L M Stiner, M T Bailey, J F Sheridan, M L Hanke, N D Powell, L M Stiner, M T Bailey, J F Sheridan

Abstract

During physiological or psychological stress, catecholamines produced by the sympathetic nervous system (SNS) regulate the immune system. Previous studies report that the activation of β-adrenergic receptors (βARs) mediates the actions of catecholamines and increases pro-inflammatory cytokine production in a number of different cell types. The impact of the SNS on the immune modulation of social defeat has not been examined. The following studies were designed to determine whether SNS activation during social disruption stress (SDR) influences anxiety-like behavior as well as the activation, priming, and glucocorticoid resistance of splenocytes after social stress. CD-1 mice were exposed to one, three, or six cycles of SDR and HPLC analysis of the plasma and spleen revealed an increase in catecholamines. After six cycles of SDR the open field test was used to measure behaviors characteristic of anxiety and indicated that the social defeat induced increase in anxiety-like behavior was blocked by pre-treatment with the β-adrenergic antagonist propranolol. Pre-treatment with the β-adrenergic antagonist propranolol did not significantly alter corticosterone levels indicating no difference in activation of the hypothalamic-pituitary-adrenal axis. In addition to anxiety-like behavior the SDR induced splenomegaly and increase in plasma IL-6, TNFα, and MCP-1 were each reversed by pre-treatment with propranolol. Furthermore, flow cytometric analysis of cells from propranolol pretreated mice reduced the SDR-induced increase in the percentage of CD11b(+) splenic macrophages and significantly decreased the expression of TLR2, TLR4, and CD86 on the surface of these cells. In addition, supernatants from 18h LPS-stimulated ex vivo cultures of splenocytes from propranolol-treated SDR mice contained less IL-6. Likewise propranolol pre-treatment abrogated the glucocorticoid insensitivity of CD11b(+) cells ex vivo when compared to splenocytes from SDR vehicle-treated mice. Together, this study demonstrates that the immune activation and priming effects of SDR result, in part, as a consequence of SNS activation.

Copyright © 2012 Elsevier Inc. All rights reserved.

Figures

Fig. 1
Fig. 1
Repeated social defeat significantly increased plasma and splenic catecholamines. Separate groups of mice were subjected to one, three or six cycles of repeated social defeat (SDR; filled bars) or left undisturbed in their home cage (HCC; open bars). Immediately following the first, third or sixth stress cycle plasma and spleens were collected to determine catecholamine levels. Mice subjected to SDR had increased levels of (A) plasma norepinephrine and (B) plasma epinephrine as well as (C) increased splenic norepinephrine regardless of the number of SDR cycles experienced. In a follow up study separate groups of mice were subjected to six cycles of SDR in order to collect plasma and spleens at 20 min, 3, 12 or 24 h post termination of the stressor in order to assess catecholamine levels. (D and E) Plasma norepinephrine and epinephrine were elevated 20 min following termination of the stressor but returned to control levels at later time points. (F) Defeated mice had elevated splenic norepinephrine except at 3 h post termination of the stressor. Bars represent the mean ± SEM (n = 6). Means with different letters (a or b) are significantly different (p < 0.05) from each other. (tw = total tissue weight).
Fig. 2
Fig. 2
β-adrenergic antagonism reduced anxiety-like behavior in socially defeated mice. CD-1 male mice were subjected to six days of repeated social defeat (SDR; filled bars) or left undisturbed in their home cage (HCC; open bars) after a s.c. injection of vehicle or propranolol prior to each cycle of defeat. Mice were assessed in the open field test the day after the last stress cycle. (A) Defeated mice spent less time in the center of the open field, propranolol prevents this behavior. (B) Defeated mice took longer to enter the center of the field than controls, propranolol blocks this behavior. (C) Distance traveled and (D) Time spent moving were not significantly different between any groups. Bars represent the mean ± SEM (n = 9). Means with different letters (a or b) are significantly different (p < 0.05) from each other.
Fig. 3
Fig. 3
β-adrenergic antagonism treatment of socially defeated mice significantly altered spleen weight but not serum corticosterone. Male CD-1 mice were injected s.c. with vehicle or propranolol prior to defeat each day for 6 days. Corticosterone was measured via radioimmune assay from defeated (SDR; filled bars) and home cage control (HCC; open bars) mice in the serum obtained via retro orbital plexus (A) immediately following the last cycle of SDR (i.e., at 1900 EST) and at 18 h after the last cycle of SDR (i.e. at 0900 EST). (C) Spleen and body weight were measured the day after the last cycle of defeat and a relative spleen mass (spleen mass/body mass) was calculated. Bars represent the mean ± SEM (n = 9). Means with different letters (a or b) are significantly different (p < 0.05) from each other.
Fig. 4
Fig. 4
β-adrenergic antagonism treatment of socially defeated mice significantly alters cell activation. Male CD-1 mice were injected s.c. with vehicle or propranolol prior to defeat each day for 6 days. (A) The day after the last cycle of defeat there was a significant increase in CD11b+ cells within the spleen of socially defeat (SDR; filled bars) mice compared to home cage control (HCC; open bars) mice as assessed via flow cytometry. (B, C, and D) The expression of Toll-like receptor (TLRs) 2 and 4 (TLR2 and TLR4) and CD86 on splenic CD11b+ monocytes/macrophages was increased as assessed via flow cytometry using fluorescent anti-TLR and anti-CD86 antibody staining. Bars represent the mean ± SEM (n = 9). Means with different letters (a or b) are significantly different (p < 0.05) from each other.
Fig. 5
Fig. 5
β-adrenergic antagonism treatment prevented stress induced cytokines after social defeat in plasma. Male CD-1 mice were subjected to six cycles of repeated social defeat (SDR; filled bars) or left undisturbed in their home cage (HCC; open bars). Mice were injected s.c. with vehicle or propranolol prior to each cycle of defeat. The day after the last cycle of defeat (A) IL-6, (B) MCP-1, and (C) TNF-α protein levels were determined in plasma. Bars represent the mean ± SEM (n = 9). Means with different letters (a or b) are significantly different (p < 0.05) from each other.
Fig. 6
Fig. 6
β-adrenergic antagonism prevented social defeat induced IL-6 production and attenuated social defeat-induced glucocorticoid insensitivity. Spleen cells were harvested from socially defeated (SDR) or home cage control (HCC) mice treated with vehicle or propranolol, stimulated with LPS, and 18 h later IL-6 was quantified from collected supernatants by ELISA. 48 h later GC insensitivity, defined as high cell viability in the presence of increased concentrations of corticosterone, was determined. SDR mice (closed square) demonstrate GC insensitivity compared to control groups (open square & circle). Cell viability was decreased in socially defeated mice treated with propranolol (closed circle). Lines represent the mean ± SEM (n = 9). *p < 0.05 vs. HCC vehicle treated mice; **p < 0.05 vs. SDR vehicle treated mice.

References

    1. Ader R, editor. Psychoneuroimmunology. Academic Press; San Diego: 2006.
    1. Anisman H, Merali Z. Cytokines, stress, and depressive illness. Brain Behav. Immun. 2002;16:513–524.
    1. Anisman H, Ravindran AV, Griffiths J, Merali Z. Endocrine and cytokine correlates of major depression and dysthymia with typical or atypical features. Mol. Psychiatry. 1999;4:182–188.
    1. Ashwell JD. The many paths to p38 mitogen-activated protein kinase activation in the immune system. Nat. Rev. Immunol. 2006;6:532–540.
    1. Avgustinovich DF, Gorbach OV, Kudryavtseva NN. Comparative analysis of anxiety-like behavior in partition and plus-maze tests after agonistic interactions in mice. Physiol. Behav. 1997;61:37–43.
    1. Avitsur R, Padgett DA, Dhabhar FS, Stark JL, Kramer KA, Engler H, Sheridan JF. Expression of glucocorticoid resistance following social stress requires a second signal. J. Leukoc. Biol. 2003;74:507–513.
    1. Avitsur R, Stark JL, Dhabhar FS, Padgett DA, Sheridan JF. Social disruption-induced glucocorticoid resistance. kinetics and site specificity. J. Neuroimmunol. 2002;124:54–61.
    1. Avitsur R, Stark JL, Sheridan JF. Social stress induces glucocorticoid resistance in subordinate animals. Horm. Behav. 2001;39:247–257.
    1. Bailey MT, Engler H, Powell ND, Padgett DA, Sheridan JF. Repeated social defeat increases the bactericidal activity of splenic macrophages through a Toll-like receptor-dependent pathway. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007;293:R1180–R1190.
    1. Bailey MT, Engler H, Sheridan JF. Stress induces the translocation of cutaneous and gastrointestinal microflora to secondary lymphoid organs of C57BL/6 mice. J. Neuroimmunol. 2006;171:29–37.
    1. Bailey MT, Kierstein S, Sharma S, Spaits M, Kinsey SG, Tliba O, Amrani Y, Sheridan JF, Panettieri RA, Haczku A. Social stress enhances allergen-induced airway inflammation in mice and inhibits corticosteroid responsiveness of cytokine production. J. Immunol. 2009a;182
    1. Bailey MT, Kinsey SG, Padgett DA, Sheridan JF, Leblebicioglu B. Social stress enhances IL-1beta and TNF-alpha production by Porphyromonas gingivalis lipopolysaccharide-stimulated CD11b+ cells. Physiol. Behav. 2009b;98:351–358.
    1. Björkqvist K. Social defeat as a stressor in humans. Physiol. Behav. 2001;73:435–442.
    1. Bosisio D, Polentarutti N, Sironi M, Bernasconi S, Miyake K, Webb GR, Martin MU, Mantovani A, Muzio M. Stimulation of Toll-like receptor 4 expression in human mononuclear phagocytes by interferon-gamma: a molecular basis for priming and synergism with bacterial lipopolysaccharide. Blood. 2002;99:3427–3431.
    1. Capuron L, Gumnick JF, Musselman DL, Lawson DH, Reemsnyder A, Nemeroff CB, Miller AH. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology. 2002;26:643–652.
    1. Chrousos GP. Regulation and dysregulation of the hypothalamic-pituitary-adrenal axis. The corticotropin-releasing hormone perspective. Endocrinol. Metab. Clin. North Am. 1992;21:833–858.
    1. Curry JM, Hanke ML, Piper MG, Bailey MT, Bringardner BD, Sheridan JF, Marsh CB. Social disruption induces lung inflammation. Brain Behav. Immun. 2010;24:394–402.
    1. Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve–an integrative interface between two supersystems: the brain and the immune system. Pharmacol. Rev. 2000;52:595–638.
    1. Engler H, Bailey MT, Engler A, Sheridan JF. Effects of repeated social stress on leukocyte distribution in bone marrow, peripheral blood and spleen. J. Neuroimmunol. 2004;148:106–115.
    1. Engler H, Engler A, Bailey MT, Sheridan JF. Tissue-specific alterations in the glucocorticoid sensitivity of immune cells following repeated social defeat in mice. J. Neuroimmunol. 2005;163:110–119.
    1. Gold SM, Irwin MR. Depression and immunity: inflammation and depressive symptoms in multiple sclerosis. Neurol. Clin. 2006;24:507–519.
    1. Hebert MA, Evenson AR, Lumley LA, MeyerhoV JL. Effects of acute social defeat on activity in the forced swim test: parametric studies in DBA/2 mice using a novel measurement device. Aggress. Behav. 1998;24:257–269.
    1. Jeon D, Kim S, Chetana M, Jo D, Ruley HE, Lin S, Rabah D, Kinet JP, H.S. S. Observational fear learning involves affective pain system and Cav1.2 Ca2+ channels in ACC. Nat. Neurosci. 2010;13
    1. Johnson JD, Campisi J, Sharkey CM, Kennedy SL, Nickerson M, Greenwood BN, Fleshner M. Catecholamines mediate stress-induced increases in peripheral and central inflammatory cytokines. Neuroscience. 2005;135:1295–1307.
    1. Johnson RR, Storts R, Welsh TH, Welsh CJ, Meagher MW. Social stress alters the severity of acute Theiler's virus infection. J. Neuroimmunol. 2004;148:74–85.
    1. Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, Frenette PS. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407–421.
    1. Kelley KW, Bluthé RM, Dantzer R, Zhou JH, Shen WH, Johnson RW, Broussard SR. Cytokine-induced sickness behavior. Brain Behav. Immun. 2003;17:S112–118.
    1. Kinsey SG, Bailey MT, Sheridan JF, Padgett DA. The inflammatory response to social defeat is increased in older mice. Physiol. Behav. 2008;93:628–636.
    1. Kinsey SG, Bailey MT, Sheridan JF, Padgett DA, Avitsur R. Repeated social defeat causes increased anxiety-like behavior and alters splenocyte function in C57BL/6 and CD-1 mice. Brain Behav. Immun. 2007;21:458–466.
    1. Klein RL, Wilson SP, Dzielak DJ, Yang WH, Viveros OH. Opioid peptides and noradrenaline co-exist in large dense-cored vesicles from sympathetic nerve. Neuroscience. 1982;7:2255–2261.
    1. Kohut ML, Martin AE, Senchina DS, Lee W. Glucocorticoids produced during exercise may be necessary for optimal virus-induced IL-2 and cell proliferation whereas both catecholamines and glucocorticoids may be required for adequate immune defense to viral infection. Brain Behav. Immun. 2005;19:423–435.
    1. Kudryavtseva NN, Bondar NP, Avgustinovich DF. Effects of repeated experience of aggression on the aggressive motivation and development of anxiety in male mice. Neurosci. Behav. Physiol. 2004;34:721–730.
    1. Madden KS. Catecholamines, sympathetic innervation, and immunity. Brain Behav. Immun. 2003;17:S5–S10.
    1. Madden KS, Sanders VM, Felten DL. Catecholamine influences and sympathetic neural modulation of immune responsiveness. Annu. Rev. Pharmacol. Toxicol. 1995;35:417–448.
    1. Maes M, Scharpé S, Meltzer HY, Bosmans E, Suy E, Calabrese J, Cosyns P. Relationships between interleukin-6 activity, acute phase proteins, and function of the hypothalamic-pituitary-adrenal axis in severe depression. Psychiatry Res. 1993;49:11–27.
    1. Mays JW, Bailey MT, Hunzeker JT, Powell ND, Papenfuss T, Karlsson EA, Padgett DA, Sheridan JF. Influenza virus-specific immunological memory is enhanced by repeated social defeat. J. Immunol. 2010;184:2014–2025.
    1. Meagher MW, Johnson RR, Young EE, Vichaya EG, Lunt S, Hardin EA, Connor MA, Welsh CJ. Interleukin-6 as a mechanism for the adverse effects of social stress on acute Theiler's virus infection. Brain Behav. Immun. 2007;8
    1. Merlot E, Moze E, Dantzer R, Neveu PJ. Cytokine production by spleen cells after social defeat in mice. Activation of T cells and reduced inhibition by glucocorticoids. Stress. 2004;7:55–61.
    1. Miura S, Kawanaka K, Kai Y, Tamura M, Goto M, Shiuchi T, Minokoshi Y, Ezaki O. An increase in murine skeletal muscle peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) mRNA in response to exercise is mediated by beta-adrenergic receptor activation. Endocrinology. 2007;148:3441–3448.
    1. Powell ND, Bailey MT, Mays JW, Stiner-Jones LM, Hanke ML, Padgett DA, Sheridan J. Repeated social defeat activates dendritic cells and enhances Toll-like receptor dependent cytokine secretion. Brain Behav. Immun. 2009;23:225–231.
    1. Quan N, Avitsur R, Stark JL, He L, Lai W, Dhabhar F, Sheridan JF. Molecular mechanisms of glucocorticoid resistance in splenocytes of socially stressed male mice. J. Neuroimmunol. 2003;137:51–58.
    1. Quan N, Avitsur R, Stark JL, He L, Shah M, Caligiuri M, Padgett DA, Marucha PT, Sheridan JF. Social stress increases the susceptibility to endotoxic shock. J. Neuroimmunol. 2001;115:36–45.
    1. Reichenberg A, Yirmiya R, Schuld A, Kraus T, Haack M, Morag A, Pollmächer T. Cytokine-associated emotional and cognitive disturbances in humans. Arch. Gen. Psychiatry. 2001;58:445–452.
    1. Rosen JB, Schulkin J. From normal fear to pathological anxiety. Psychol. Rev. 1998;105:325–350.
    1. Samama P, Cotecchia S, Costa T, Lefkowitz RJ. A mutation-induced activated state of the beta 2-adrenergic receptor. Extending the ternary complex model. J. Biol. Chem. 1993;268:4325–4636.
    1. Schroeder S, Wichers M, Klingmuller D, Hofer M, Lehmann LE, von ST, Hering R, Putensen C, Hoeft A, Stuber F. The hypothalamicpituitary-adrenal axis of patients with severe sepsis: altered response to corticotropin-releasing hormone. Crit. Care Med. 2001;29:310–316.
    1. Serbina NV, Jia T, Hohl TM, Pamer EG. Monocyte-mediated defense against microbial pathogens. Annu. Rev. Immunol. 2008;26:421–452.
    1. Sheridan JF, Padgett DA, Avitsur R, Marucha PT. Experimental models of stress and wound healing. World J. Surg. 2004;28:327–330.
    1. Stark JL, Avitsur R, Hunzeker J, Padgett DA, Sheridan JF. Interleukin-6 and the development of social disruption-induced glucocorticoid resistance. J. Neuroimmunol. 2002;124:9–15.
    1. Stark JL, Avitsur R, Padgett DA, Campbell KA, Beck FM, Sheridan JF. Social stress induces glucocorticoid resistance in macrophages. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2001;280:R1799–R1805.
    1. Tan KS, Nackley AG, Satterfield K, Maixner W, Diatchenko L, Flood PM. β2 adrenergic receptor activation stimulates pro-inflammatory cytokine production in macrophages via PKA- and NF-kappaB-independent mechanisms. Cell. Signal. 2007;19:251–260.
    1. Violin JD, Lefkowitz RJ. Beta-arrestin-biased ligands at seven-transmembrane receptors. Trends Pharmacol. Sci. 2007;28:416–422.
    1. Wohleb ES, Hanke ML, Corona AW, Powell ND, Stiner LM, Bailey MT, Nelson RJ, Godbout JP, Sheridan JF. β-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J. Neurosci. 2011;31:6277–6288.
    1. Zorrilla E, Luborsky L, McKay JR, Rosenthal R, Houldin A, Tax A, McCorkle R, Seligma n.D.A., Schmidt K. The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav. Immun. 2001;15:199–226.

Source: PubMed

3
Iratkozz fel