The sympathetic nervous response in inflammation

Georg Pongratz, Rainer H Straub, Georg Pongratz, Rainer H Straub

Abstract

Over the past decades evidence has accumulated clearly demonstrating a pivotal role for the sympathetic nervous system (SNS) and its neurotransmitters in regulating inflammation. The first part of this review provides the reader with an overview showing that the interaction of the SNS with the immune system to control inflammation is strongly context-dependent (for example, depending on the activation state of the immune cell or neuro-transmitter concentration). In the second part we focus on autoimmune arthritis as a well investigated example for sympathetically controlled inflammation to show that the SNS and catecholamines play a differential role depending on the time point of ongoing disease. A model will be developed to explain the proinflammatory effects of the SNS in the early phase and the anti-inflammatory effects of catecholamines in the later phase of autoimmune arthritis. In the final part, a conceptual framework is discussed that shows that a major purpose of increased SNS activity is nourishment of a continuously activated immune system at a systemic level using energy-rich fuels (glucose, amino acids, lipids), while uncoupling from central nervous regulation occurs at sites of inflammation by repulsion of sympathetic fibers and local adrenoceptor regulation. This creates zones of ‘permitted local inflammation’. However, if this ‘inflammatory configuration’ persists and is strong, as in autoimmunity, the effects are detrimental because of the resultant chronic catabolic state, leading to cachexia, high blood pressure, insulin resistance, and increased cardiovascular mortality, and so on. Today, the challenge is to translate this conceptual knowledge into clinical benefit.

Figures

Figure 1
Figure 1
Basic neuronal anti-inflammatory reflex. Local inflammation (the fire) is detected by vagal and sensory nerve fibers, which express receptors for inflammatory mediators, like interleukin (IL)-1β (red dots). An afferent signal is generated and transmitted to the brain (central nervous sytem (CNS)), which in turn leads to activation of the sympathetic nervous system (SNS), which has a complex impact on inflammation. Local release of SNS neurotransmitters, like norepinephrine, at the site of inflammation or in secondary lymphoid organs has a net anti-inflammatory outcome. On the other hand, non-specific immune stimulatory processes on a systemic level are supported, like recruitment of leukocytes, increased blood and lymph flow, but also increasing antigen processing and presentation and provision of energy-rich fuels. Ln, lymph node.
Figure 2
Figure 2
Catecholamine effects depend on the distance from catecholamine source. α- and β-adrenoceptors (ARs) show different binding affinities for catecholamines. Norepinephrine, the main neurotransmitter in the sympathetic nervous system (SNS), binds with higher affinity to α-ARs than β-ARs. Simultaneous expression of these receptors on immune cells (for example, macrophages (MΦ)) provides these cells with a passive means to determine the distance to the next catecholamine source. In close proximity to the catecholamine source (for example, sympathetic nerve terminal or catecholamine-producing tyrosine hydroxylase (TH)-positive cell) the concentration is high enough to activate β-ARs, whereas at a greater distance only α-ARs are activated. In the case of innate immune cells, like macrophages, this directly translates into anti-inflammatory (for example, increases in interleukin (IL)-10 via β-AR) or proinflammatory activity (for example, increases in tumor necrosis factor (TNF) via α-AR). Therefore, the simultaneous expression of α-ARs and β-ARs on immune cells provides a mean to regulate inflammatory processes dependent on the distance to the catecholamine source. We hypothesize that the body uses this system to promote local inflammation by repulsion of sympathetic nerve fibers from inflamed areas (zone of inflammation) and, at the same time, locally confines the inflammatory process by suppression of bystander activation in the zone of anti-inflammation.
Figure 3
Figure 3
Current model of sympathetic nervous system influence in arthritis. In early arthritis (left panel), the sympathetic nervous system (SNS) supports inflammation in the joint through a proinflammatory influence on adaptive immune cells; for example, increased specific antibody production by B cells and increased proinflammatory activity of T cells. The SNS also inhibits innate immune cells via stimulation of β2 adrenoceptors (β2ARs), although the net outcome of SNS influence in the early phase is proinflammatory. Then, during the transition phase, we hypothesize that the influence of the SNS changes from pro- to anti-inflammatory. In the later stages, central regulation of the inflammatory process is less important, since sympathetic nerve fibers are repelled from the inflamed area and secondary lymphoid organs. However, local sympathetic influence becomes increasingly important, indicated by the appearance of catecholamine-producing, tyrosine hydroxylase-positive (TH+) cells, which have a dominant anti-inflammatory effect. Possible mechanisms of action are paracrine and autocrine in manner; for example, inhibiting proinflammatory interleukin (IL)-7 receptor-positive B cells, increasing the activity of IL-10-producing anti-inflammatory B cells, or inhibiting innate immune cells via β2AR-mediated effects. AR, adrenoceptor; cAMP, cyclic adenosine monophosphate; CD, cluster of differentiation; FoxP3, forkhead box P3; IFN, interferon; MHC, major histocompatibility complex; pSTAT5, phosphorylated-signal transducer and activator of transcription 5; TCR, T-cell receptor; Th1, T helper 1 cell.
Figure 4
Figure 4
Morphologic adaptation to persistent inflammation. Centrally controlled increase of sympathetic nervous system (SNS) activity is a basic response to inflammation. The constant increase in SNS activity supports inflammation in several ways; for example, increasing blood flow, lymph flow, antigen presentation, and liberation of energy-rich fuels like lipids and glucose from adipose tissue and liver. However, the specific interaction with immune cells in secondary lymphoid organs and at local sites of inflammation (for example, joints) shows a net anti-inflammatory effect. Therefore, to mount an effective immune response, non-specific support of inflammation on a systemic level is maintained, while the anti-inflammatory influence on a local level is decreased and uncoupled from central regulation through repulsion of sympathetic nerve fibers and the appearance of tyrosine hydroxylase (TH) + catecholamine-producing cells during the inflammatory process. In the end, a systemic proinflammatory configuration is established, which helps to optimally clear the antigen. However, if inflammation persists, like during chronic inflammation, this constant increase in SNS activity and resultant catabolic state is detrimental to the body and results in known disease sequelae of chronic inflammatory conditions, like cachexia, diabetes, hyperlipidemia, high blood pressure, increased cardiovascular risk, and so on.

References

    1. Meltzer SJ, Meltzer C. On a difference in the influence upon inflammation between the section of the sympathetic nerve and the removal of the sympathetic ganglion. J Med Res. 1903;10:135–141.
    1. Hopkin DA, Laplane R. James Reilly and the autonomic nervous system. A prophet unheeded? Ann R Coll Surg Engl. 1978;60:108–116.
    1. Besedovsky H, del Rey A, Sorkin E, Dinarello CA. Immunoregulatory feedback between interleukin-1 and glucocorticoid hormones. Science. 1986;233:652–654. doi: 10.1126/science.3014662.
    1. Nance DM, Sanders VM. Autonomic innervation and regulation of the immune system (1987–2007) Brain Behav Immun. 2007;21:736–745. doi: 10.1016/j.bbi.2007.03.008.
    1. Goehler LE, Relton JK, Dripps D, Kiechle R, Tartaglia N, Maier SF, Watkins LR. Vagal paraganglia bind biotinylated interleukin-1 receptor antagonist: a possible mechanism for immune-to-brain communication. Brain Res Bull. 1997;43:357–364. doi: 10.1016/S0361-9230(97)00020-8.
    1. Zielinski MR, Dunbrasky DL, Taishi P, Souza G, Krueger JM. Vagotomy attenuates brain cytokines and sleep induced by peripherally administered tumor necrosis factor-alpha and lipopolysaccharide in mice. Sleep. 2013;36:227–1238.
    1. Pöyhönen-Alho MK, Manhem K, Katzman P, Kibarskis A, Antikainen RL, Erkkola RU, Tuomilehto JO, Ebeling PE, Kaaja RJ. Central sympatholytic therapy has anti-inflammatory properties in hypertensive postmenopausal women. J Hypertens. 2008;26:2445–2449. doi: 10.1097/HJH.0b013e328311cf37.
    1. Bernstein IM, Damron D, Schonberg AL, Shapiro R. The relationship of plasma volume, sympathetic tone, and proinflammatory cytokines in young healthy nonpregnant women. Reprod Sci. 2009;16:980–985. doi: 10.1177/1933719109338876.
    1. Straub RH, Kalden JR. Stress of different types increases the proinflammatory load in rheumatoid arthritis. Arthritis Res Ther. 2009;11:114. doi: 10.1186/ar2712.
    1. del Rey A, Wolff C, Wildmann J, Randolf A, Hahnel A, Besedovsky HO, Straub RH. Disrupted brain-immune system-joint communication during experimental arthritis. Arthritis Rheum. 2008;58:3090–3099. doi: 10.1002/art.23869.
    1. Felten DL, Felten SY, Bellinger DL, Carlson SL, Ackerman KD, Madden KS, Olschowki JA, Livnat S. Noradrenergic sympathetic neural interactions with the immune system: structure and function. Immunol Rev. 1987;100:225–260. doi: 10.1111/j.1600-065X.1987.tb00534.x.
    1. Carr DJJ, Blalock JE. Neuropeptide hormones and receptors common to the immune and neuroendocrine systems: bidirectional pathway of intersystem communication. In: Ader R, Felten DL, Cohen N, editors. Psychoneuroimmunology. 2. San Diego: Academic Press, Inc; 1991. pp. 573–588.
    1. Thompson M, Bywaters EG. Unilateral rheumatoid arthritis following hemiplegia. Ann Rheum Dis. 1962;21:370–377. doi: 10.1136/ard.21.4.370.
    1. Tarkowski E, Naver H, Wallin BG, Blomstrand C, Tarkowski A. Lateralization of T-lymphocyte responses in patients with stroke. Effect of sympathetic dysfunction? Stroke. 1995;26:57–62. doi: 10.1161/01.STR.26.1.57.
    1. Tarkowski E, Naver H, Wallin BG, Blomstrand C, Grimby G, Tarkowski A. Lateralization of cutaneous inflammatory responses in patients with unilateral paresis after poliomyelitis. J Neuroimmunol. 1996;67:1–6. doi: 10.1016/0165-5728(96)00011-2.
    1. Prass K, Meisel C, Höflich C, Braun J, Halle E, Wolf T, Ruscher K, Victorov IV, Priller J, Dirnagl U, Volk HD, Meisel A. Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation. J Exp Med. 2003;198:725–736. doi: 10.1084/jem.20021098.
    1. Straub RH, Pongratz G, Weidler C, Linde HJ, Kirschning CJ, Glück T, Schölmerich J, Falk W. Ablation of the sympathetic nervous system decreases gram-negative and increases gram-positive bacterial dissemination: key roles for tumor necrosis factor/phagocytes and interleukin-4/lymphocytes. J Infect Dis. 2005;192:560–572. doi: 10.1086/432134.
    1. Burnstock G. Cotransmission in the autonomic nervous system. Handb Clin Neurol. 2013;117:23–35. doi: 10.1016/B978-0-444-53491-0.00003-1.
    1. Sung CP, Arleth AJ, Feuerstein GZ. Neuropeptide Y upregulates the adhesiveness of human endothelial cells for leukocytes. Circ Res. 1991;68:314–318. doi: 10.1161/01.RES.68.1.314.
    1. Claxson A, Morris C, Blake D, Sirén M, Halliwell B, Gustafsson T, Löfkvist B, Bergelin I. The anti-inflammatory effects of D-myo-inositol-1.2.6-trisphosphate (PP56) on animal models of inflammation. Agents Actions. 1990;29:68–70. doi: 10.1007/BF01964724.
    1. Howarth D, Burstal R, Hayes C, Lan L, Lantry G. Autonomic regulation of lymphatic flow in the lower extremity demonstrated on lymphoscintigraphy in patients with reflex sympathetic dystrophy. Clin Nucl Med. 1999;24:383–387. doi: 10.1097/00003072-199906000-00001.
    1. Scheiermann C, Kunisaki Y, Lucas D, Chow A, Jang JE, Zhang D, Hashimoto D, Merad M, Frenette PS. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity. 2012;37:290–301. doi: 10.1016/j.immuni.2012.05.021.
    1. Powell ND, Sloan EK, Bailey MT, Arevalo JM, Miller GE, Chen E, Kobor MS, Reader BF, Sheridan JF, Cole SW. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via beta-adrenergic induction of myelopoiesis. Proc Natl Acad Sci U S A. 2013;110:16574–16579. doi: 10.1073/pnas.1310655110.
    1. Merhi M, Helme RD, Khalil Z. Age-related changes in sympathetic modulation of sensory nerve activity in rat skin. Inflamm Res. 1998;47:239–244. doi: 10.1007/s000110050324.
    1. Dawson LF, Phillips JK, Finch PM, Inglis JJ, Drummond PD. Expression of alpha1-adrenoceptors on peripheral nociceptive neurons. Neuroscience. 2011;175:300–314. doi: 10.1016/j.neuroscience.2010.11.064.
    1. Seeley EJ, Barry SS, Narala S, Matthay MA, Wolters PJ. Noradrenergic neurons regulate monocyte trafficking and mortality during gram-negative peritonitis in mice. J Immunol. 2013;190:4717–4724. doi: 10.4049/jimmunol.1300027.
    1. Drummond PD. The effect of sympathetic activity on thermal hyperalgesia in capsaicin-treated skin during body cooling and warming. Eur J Pain. 2001;5:59–67. doi: 10.1053/eujp.2001.0224.
    1. Birklein F, Kunzel W, Sieweke N. Despite clinical similarities there are significant differences between acute limb trauma and complex regional pain syndrome I (CRPS I) Pain. 2001;93:165–171. doi: 10.1016/S0304-3959(01)00309-8.
    1. Spengler RN, Allen RM, Remick DG, Strieter RM, Kunkel SL. Stimulation of alpha-adrenergic receptor augments the production of macrophage-derived tumor necrosis factor. J Immunol. 1990;145:1430–1434.
    1. Severn A, Rapson NT, Hunter CA, Liew FY. Regulation of tumor necrosis factor production by adrenaline and beta-adrenergic agonists. J Immunol. 1992;148:3441–3445.
    1. Szabó C, Haskó G, Zingarelli B, Németh ZH, Salzman AL, Kvetan V, Pastores SM, Vizi ES. Isoproterenol regulates tumour necrosis factor, interleukin-10, interleukin-6 and nitric oxide production and protects against the development of vascular hyporeactivity in endotoxaemia. Immunology. 1997;90:95–100. doi: 10.1046/j.1365-2567.1997.00137.x.
    1. Xiu F, Stanojcic M, Jeschke MG. Norepinephrine inhibits macrophage migration by decreasing CCR2 expression. PLoS One. 2013;8:e69167. doi: 10.1371/journal.pone.0069167.
    1. Szelenyi J, Kiss JP, Vizi ES. Differential involvement of sympathetic nervous system and immune system in the modulation of TNF-alpha production by alpha2- and beta-adrenoceptors in mice. J Neuroimmunol. 2000;103:34–40. doi: 10.1016/S0165-5728(99)00234-9.
    1. Zhou JR, Zhang LD, Wei HF, Wang X, Ni HL, Yang F, Zhang T, Jiang CL. Neuropeptide Y induces secretion of high-mobility group box 1 protein in mouse macrophage via PKC/ERK dependent pathway. J Neuroimmunol. 2013;260:55–59. doi: 10.1016/j.jneuroim.2013.04.005.
    1. Manni M, Granstein RD, Maestroni G. beta2-Adrenergic agonists bias TLR-2 and NOD2 activated dendritic cells towards inducing an IL-17 immune response. Cytokine. 2011;55:380–386. doi: 10.1016/j.cyto.2011.05.013.
    1. Yanagawa Y, Matsumoto M, Togashi H. Adrenoceptor-mediated enhancement of interleukin-33 production by dendritic cells. Brain Behav Immun. 2011;25:1427–1433. doi: 10.1016/j.bbi.2011.04.012.
    1. Yanagawa Y, Matsumoto M, Togashi H. Enhanced dendritic cell antigen uptake via alpha2 adrenoceptor-mediated PI3K activation following brief exposure to noradrenaline. J Immunol. 2010;185:5762–5768. doi: 10.4049/jimmunol.1001899.
    1. Goyarts E, Matsui M, Mammone T, Bender AM, Wagner JA, Maes D, Granstein RD. Norepinephrine modulates human dendritic cell activation by altering cytokine release. Exp Dermatol. 2008;17:188–196. doi: 10.1111/j.1600-0625.2007.00677.x.
    1. Wirth T, Westendorf AM, Bloemker D, Wildmann J, Engler H, Mollerus S, Wadwa M, Schäfer MK, Schedlowski M, del Rey A. The sympathetic nervous system modulates CD4Foxp3 regulatory T cells via noradrenaline-dependent apoptosis in a murine model of lymphoproliferative disease. Brain Behav Immun. 2014;38:100–110. doi: 10.1016/j.bbi.2014.01.007.
    1. Guereschi MG, Araujo LP, Maricato JT, Takenaka MC, Nascimento VM, Vivanco BC, Reis VO, Keller AC, Brum PC, Basso AS. Beta2-adrenergic receptor signaling in CD4+ Foxp3+ regulatory T cells enhances their suppressive function in a PKA-dependent manner. Eur J Immunol. 2013;43:1001–1012. doi: 10.1002/eji.201243005.
    1. Straub RH, Rauch L, Fassold A, Lowin T, Pongratz G. Neuronally released sympathetic neurotransmitters stimulate splenic interferon-gamma secretion from T cells in early type II collagen-induced arthritis. Arthritis Rheum. 2008;58:3450–3460. doi: 10.1002/art.24030.
    1. Ramer-Quinn DS, Swanson MA, Lee WT, Sanders VM. Cytokine production by naive and primary effector CD4+ T cells exposed to norepinephrine. Brain Behav Immun. 2000;14:239–255. doi: 10.1006/brbi.2000.0603.
    1. Swanson MA, Lee WT, Sanders VM. IFN-gamma production by Th1 cells generated from naive CD4+ T cells exposed to norepinephrine. J Immunol. 2001;166:232–240. doi: 10.4049/jimmunol.166.1.232.
    1. Kawamura N, Tamura H, Obana S, Wenner M, Ishikawa T, Nakata A, Yamamoto H. Differential effects of neuropeptides on cytokine production by mouse helper T cell subsets. Neuroimmunomodulation. 1998;5:9–15. doi: 10.1159/000026321.
    1. Pongratz G, Melzer M, Straub RH. The sympathetic nervous system stimulates anti-inflammatory B cells in collagen-type II-induced arthritis. Ann Rheum Dis. 2012;71:432–439. doi: 10.1136/ard.2011.153056.
    1. Pongratz G, Anthofer JM, Melzer M, Anders S, Grassel S, Straub RH. IL-7 receptor alpha expressing B cells act proinflammatory in collagen-induced arthritis and are inhibited by sympathetic neurotransmitters. Ann Rheum Dis. 2014;73:306–312. doi: 10.1136/annrheumdis-2012-202944.
    1. Pongratz G, McAlees JW, Conrad DH, Erbe RS, Haas KM, Sanders VM. The level of IgE produced by a B cell is regulated by norepinephrine in a p38. J Immunol. 2006;177:2926–2938. doi: 10.4049/jimmunol.177.5.2926.
    1. Podojil JR, Sanders VM. Selective regulation of mature IgG1 transcription by CD86 and beta 2-adrenergic receptor stimulation. J Immunol. 2003;170:5143–5151. doi: 10.4049/jimmunol.170.10.5143.
    1. Prösch S, Wendt CE, Reinke P, Priemer C, Oppert M, Krüger DH, Volk HD, Döcke WD. A novel link between stress and human cytomegalovirus (HCMV) infection: sympathetic hyperactivity stimulates HCMV activation. Virology. 2000;272:357–365. doi: 10.1006/viro.2000.0367.
    1. Swain MG, Blennerhassett PA, Collins SM. Impaired sympathetic nerve function in the inflamed rat intestine. Gastroenterology. 1991;100:675–682.
    1. Harle P, Mobius D, Carr DJ, Scholmerich J, Straub RH. An opposing time-dependent immune-modulating effect of the sympathetic nervous system conferred by altering the cytokine profile in the local lymph nodes and spleen of mice with type II collagen-induced arthritis. Arthritis Rheum. 2005;52:1305–1313. doi: 10.1002/art.20987.
    1. Li W, Knowlton D, Woodward WR, Habecker BA. Regulation of noradrenergic function by inflammatory cytokines and depolarization. J Neurochem. 2003;86:774–783. doi: 10.1046/j.1471-4159.2003.01890.x.
    1. Lorton D, Lubahn C, Bellinger DL. Potential use of drugs that target neural-immune pathways in the treatment of rheumatoid arthritis and other autoimmune diseases. Curr Drug Targets Inflamm Allergy. 2003;2:1–30. doi: 10.2174/1568010033344499.
    1. Donoso V, Gomez CR, Orriantia MA, Pérez V, Torres C, Coddou C, Nelson P, Maisey K, Morales B, Fernandez R, Imarai M, Huidobro-Toro JP, Sierra F, Acuña-Castillo C. The release of sympathetic neurotransmitters is impaired in aged rats after an inflammatory stimulus: a possible link between cytokine production and sympathetic transmission. Mech Ageing Dev. 2008;129:728–734. doi: 10.1016/j.mad.2008.09.018.
    1. Olofsson PS, Rosas-Ballina M, Levine YA, Tracey KJ. Rethinking inflammation: neural circuits in the regulation of immunity. Immunol Rev. 2012;248:188–204. doi: 10.1111/j.1600-065X.2012.01138.x.
    1. Bratton BO, Martelli D, McKinley MJ, Trevaks D, Anderson CR, McAllen RM. Neural regulation of inflammation: no neural connection from the vagus to splenic sympathetic neurons. Exp Physiol. 2012;97:1180–1185. doi: 10.1113/expphysiol.2011.061531.
    1. Martelli D, McKinley MJ, McAllen RM. The cholinergic anti-inflammatory pathway: A critical review. Auton Neurosci. 2013;182:65–69. doi: 10.1016/j.autneu.2013.12.007.
    1. Martelli D, Yao ST, McKinley MJ, McAllen RM. Reflex control of inflammation by sympathetic nerves, not the vagus. J Physiol. 2014;592:1677–1686. doi: 10.1113/jphysiol.2013.268573.
    1. Todorov LD, Mihaylova-Todorova ST, Bjur RA, Westfall DP. Differential cotransmission in sympathetic nerves: role of frequency of stimulation and prejunctional autoreceptors. J Pharmacol Exp Ther. 1999;290:241–246.
    1. Stohl LL, Zang JB, Ding W, Manni M, Zhou XK, Granstein RD. Norepinephrine and adenosine-5’-triphosphate synergize in inducing IL-6 production by human dermal microvascular endothelial cells. Cytokine. 2013;64:605–612. doi: 10.1016/j.cyto.2013.08.005.
    1. Chen E, Miller GE. Stress and inflammation in exacerbations of asthma. Brain Behav Immun. 2007;21:993–999. doi: 10.1016/j.bbi.2007.03.009.
    1. Saunders PR, Miceli P, Vallance BA, Wang L, Pinto S, Tougas G, Kamath M, Jacobson K. Noradrenergic and cholinergic neural pathways mediate stress-induced reactivation of colitis in the rat. Auton Neurosci. 2006;124:56–68. doi: 10.1016/j.autneu.2005.12.002.
    1. Straub RH, Grum F, Strauch U, Capellino S, Bataille F, Bleich A, Falk W, Schölmerich J, Obermeier F. Anti-inflammatory role of sympathetic nerves in chronic intestinal inflammation. Gut. 2008;57:911–921. doi: 10.1136/gut.2007.125401.
    1. Steinle JJ. Sympathetic neurotransmission modulates expression of inflammatory markers in the rat retina. Exp Eye Res. 2007;84:118–125. doi: 10.1016/j.exer.2006.09.006.
    1. Lorton D, Lubahn C, Klein N, Schaller J, Bellinger DL. Dual role for noradrenergic innervation of lymphoid tissue and arthritic joints in adjuvant-induced arthritis. Brain Behav Immun. 1999;13:315–334. doi: 10.1006/brbi.1999.0564.
    1. Arnold J, de Arellano ML B, Rüster C, Vercellino GF, Chiantera V, Schneider A, Mechsner S. Imbalance between sympathetic and sensory innervation in peritoneal endometriosis. Brain Behav Immun. 2012;26:132–141. doi: 10.1016/j.bbi.2011.08.004.
    1. Manni M, Maestroni GJ. Sympathetic nervous modulation of the skin innate and adaptive immune response to peptidoglycan but not lipopolysaccharide: involvement of beta-adrenoceptors and relevance in inflammatory diseases. Brain Behav Immun. 2008;22:80–88. doi: 10.1016/j.bbi.2007.06.016.
    1. Grebe KM, Takeda K, Hickman HD, Bailey AL, Embry AC, Bennink JR, Yewdell JW. Cutting edge: Sympathetic nervous system increases proinflammatory cytokines and exacerbates influenza A virus pathogenesis. J Immunol. 2010;184:540–544. doi: 10.4049/jimmunol.0903395.
    1. Machado MP, Rocha AM, de Oliveira LF, de Cuba MB, de Oliveira LI, Castellano LR, Silva MV, Machado JR, Nascentes GA, Paiva LH, Savino W, Junior VR, Brum PC, Prado VF, Prado MA, Silva EL, Montano N, Ramirez LE, VJ D d S. Autonomic nervous system modulation affects the inflammatory immune response in mice with acute Chagas disease. Exp Physiol. 2012;97:1186–1202. doi: 10.1113/expphysiol.2012.066431.
    1. Schlereth T, Drummond PD, Birklein F. Inflammation in CRPS: role of the sympathetic supply. Auton Neurosci. 2013;182:102–107. doi: 10.1016/j.autneu.2013.12.011.
    1. Levick SP, Murray DB, Janicki JS, Brower GL. Sympathetic nervous system modulation of inflammation and remodeling in the hypertensive heart. Hypertension. 2010;55:270–276. doi: 10.1161/HYPERTENSIONAHA.109.142042.
    1. Strack I, Schulte S, Varnholt H, Schievenbusch S, Töx U, Wendland K, Steffen HM, Drebber U, Dienes HP, Odenthal M. beta-Adrenoceptor blockade in sclerosing cholangitis of Mdr2 knockout mice: antifibrotic effects in a model of nonsinusoidal fibrosis. Lab Invest. 2011;91:252–261. doi: 10.1038/labinvest.2010.162.
    1. Hermann G, Beck FM, Tovar CA, Malarkey WB, Allen C, Sheridan JF. Stress-induced changes attributable to the sympathetic nervous system during experimental influenza viral infection in DBA/2 inbred mouse strain. J Neuroimmunol. 1994;53:173–180. doi: 10.1016/0165-5728(94)90027-2.
    1. Fitzgerald PJ. Beta blockers, norepinephrine, and cancer: an epidemiological viewpoint. Clin Epidemiol. 2012;4:151–156. doi: 10.2147/CLEP.S33695.
    1. Oberbeck R, Schmitz D, Wilsenack K, Schüler M, Pehle B, Schedlowski M, Exton MS. Adrenergic modulation of survival and cellular immune functions during polymicrobial sepsis. Neuroimmunomodulation. 2004;11:214–223. doi: 10.1159/000078439.
    1. Zhou M, Hank SH, Wang P. Increased gut-derived norepinephrine release in sepsis: up-regulation of intestinal tyrosine hydroxylase. Biochim Biophys Acta. 2004;1689:212–218. doi: 10.1016/j.bbadis.2004.03.008.
    1. Kidd BL, Cruwys S, Mapp PI, Blake DR. Role of the sympathetic nervous system in chronic joint pain and inflammation. Ann Rheum Dis. 1992;51:1188–1191. doi: 10.1136/ard.51.11.1188.
    1. Levine JD, Fye K, Heller P, Basbaum AI, Whiting-O’Keefe Q. Clinical response to regional intravenous guanethidine in patients with rheumatoid arthritis. J Rheumatol. 1986;13:1040–1043.
    1. Aloe L, Tuveri MA, Levi-Montalcini R. Studies on carrageenan-induced arthritis in adult rats: presence of nerve growth factor and role of sympathetic innervation. Rheumatol Int. 1992;12:213–216. doi: 10.1007/BF00302155.
    1. Levine JD, Moskowitz MA, Basbaum AI. The contribution of neurogenic inflammation in experimental arthritis. J Immunol. 1985;135:843s–847s.
    1. Levine JD, Dardick SJ, Roizen MF, Helms C, Basbaum AI. Contribution of sensory afferents and sympathetic efferents to joint injury in experimental arthritis. J Neurosci. 1986;6:3423–3429.
    1. Ebbinghaus M, Gajda M, Boettger MK, Schaible HG, Brauer R. The anti-inflammatory effects of sympathectomy in murine antigen-induced arthritis are associated with a reduction of Th1 and Th17 responses. Ann Rheum Dis. 2012;71:253–261. doi: 10.1136/ard.2011.150318.
    1. Harle P, Pongratz G, Albrecht J, Tarner IH, Straub RH. An early sympathetic nervous system influence exacerbates collagen-induced arthritis via CD4 + CD25+ cells. Arthritis Rheum. 2008;58:2347–2355. doi: 10.1002/art.23628.
    1. Mapp PI, Walsh DA, Garrett NE, Kidd BL, Cruwys SC, Polak JM, Blake DR. Effect of three animal models of inflammation on nerve fibres in the synovium. Ann Rheum Dis. 1994;53:240–246. doi: 10.1136/ard.53.4.240.
    1. Miller LE, Justen HP, Scholmerich J, Straub RH. The loss of sympathetic nerve fibers in the synovial tissue of patients with rheumatoid arthritis is accompanied by increased norepinephrine release from synovial macrophages. FASEB J. 2000;14:2097–2107. doi: 10.1096/fj.99-1082com.
    1. Fassold A, Falk W, Anders S, Hirsch T, Mirsky VM, Straub RH. Soluble neuropilin-2, a nerve repellent receptor, is increased in rheumatoid arthritis synovium and aggravates sympathetic fiber repulsion and arthritis. Arthritis Rheum. 2009;60:2892–2901. doi: 10.1002/art.24860.
    1. Capellino S, Weber K, Gelder M, Harle P, Straub RH. First appearance and location of catecholaminergic cells during experimental arthritis and elimination by chemical sympathectomy. Arthritis Rheum. 2012;64:1110–1118. doi: 10.1002/art.33431.
    1. Flierl MA, Rittirsch D, Nadeau BA, Chen AJ, Sarma JV, Zetoune FS, McGuire SR, List RP, Day DE, Hoesel LM, Gao H, Van Rooijen N, Huber-Lang MS, Neubig RR, Ward PA. Phagocyte-derived catecholamines enhance acute inflammatory injury. Nature. 2007;449:721–725. doi: 10.1038/nature06185.
    1. Cosentino M, Fietta AM, Ferrari M, Rasini E, Bombelli R, Carcano E, Saporiti F, Meloni F, Marino F, Lecchini S. Human CD4 + CD25+ regulatory T cells selectively express tyrosine hydroxylase and contain endogenous catecholamines subserving an autocrine/paracrine inhibitory functional loop. Blood. 2007;109:632–642. doi: 10.1182/blood-2006-01-028423.
    1. Capellino S, Cosentino M, Wolff C, Schmidt M, Grifka J, Straub RH. Catecholamine-producing cells in the synovial tissue during arthritis: modulation of sympathetic neurotransmitters as new therapeutic target. Ann Rheum Dis. 2010;69:1853–1860. doi: 10.1136/ard.2009.119701.
    1. Jenei-Lanzl Z, Capellino S, Kees F, Fleck M, Lowin T, Straub RH: Anti-inflammatory effects of cell-based therapy with tyrosine hydroxylase-positive catecholaminergic cells in experimental arthritis.Ann Rheum Dis 2013, doi:10.1136/annrheumdis-2013-203925.
    1. Bopp T, Becker C, Klein M, Klein-Hessling S, Palmetshofer A, Serfling E, Heib V, Becker M, Kubach J, Schmitt S, Stoll S, Schild H, Staege MS, Stassen M, Jonuleit H, Schmitt E. Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. J Exp Med. 2007;204:1303–1310. doi: 10.1084/jem.20062129.
    1. Straub RH, Cutolo M, Buttgereit F, Pongratz G. Energy regulation and neuroendocrine-immune control in chronic inflammatory diseases. J Intern Med. 2010;267:543–560. doi: 10.1111/j.1365-2796.2010.02218.x.
    1. Lo EJ, Green PG, Miao FJ, Relchling DB, Levine JD. Bradykinin-induced neurogenic migration of neutrophils into the rat knee joint. Neuroreport. 1999;10:3821–3824. doi: 10.1097/00001756-199912160-00018.
    1. Sanders VM. The beta2-adrenergic receptor on T and B lymphocytes: do we understand it yet? Brain Behav Immun. 2012;26:195–200. doi: 10.1016/j.bbi.2011.08.001.
    1. Grailer JJ, Haggadone MD, Sarma JV, Zetoune FS, Ward PA. Induction of M2 regulatory macrophages through the beta-adrenergic receptor with protection during endotoxemia and acute lung injury. J Innate Immun. 2014;6:607–618. doi: 10.1159/000358524.
    1. Stanojevic S, Dimitrijevic M, Kustrimovic N, Mitic K, Vujic V, Leposavic G. Adrenal hormone deprivation affects macrophage catecholamine metabolism and beta2-adrenoceptor density, but not propranolol stimulation of tumour necrosis factor-alpha production. Exp Physiol. 2013;98:665–678. doi: 10.1113/expphysiol.2012.070524.
    1. Deo SH, Jenkins NT, Padilla J, Parrish AR, Fadel PJ. Norepinephrine increases NADPH oxidase-derived superoxide in human peripheral blood mononuclear cells via alpha-adrenergic receptors. Am J Physiol Regul Integr Comp Physiol. 2013;305:R1124–R1132. doi: 10.1152/ajpregu.00347.2013.
    1. Buxton IL, Brunton LL. Alpha-adrenergic receptors on rat ventricular myocytes: characteristics and linkage to cAMP metabolism. Am J Physiol. 1986;251:H307–H313.
    1. Vega JL, Keino H, Masli S. Surgical denervation of ocular sympathetic afferents decreases local transforming growth factor-beta and abolishes immune privilege. Am J Pathol. 2009;175:1218–1225. doi: 10.2353/ajpath.2009.090264.
    1. Dekkers JC, Geenen R, Godaert GL, Bijlsma JW, van Doornen LJ. Elevated sympathetic nervous system activity in patients with recently diagnosed rheumatoid arthritis with active disease. Clin Exp Rheumatol. 2004;22:63–70.
    1. Straub RH. Evolutionary medicine and chronic inflammatory state - known and new concepts in pathophysiology. J Mol Med (Berl) 2012;90:523–534. doi: 10.1007/s00109-012-0861-8.
    1. Poole RM, Ballantyne AD. Apremilast: first global approval. Drugs. 2014;74:825–837. doi: 10.1007/s40265-014-0218-4.
    1. Schafer P. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem Pharmacol. 2012;83:1583–1590. doi: 10.1016/j.bcp.2012.01.001.

Source: PubMed

3
Se inscrever