Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jan 22.
Published in final edited form as: Cell Immunol. 2008 Mar 4;252(1-2):27–56. doi: 10.1016/j.cellimm.2007.09.005

Sympathetic Modulation of Immunity: Relevance to Disease

Denise L Bellinger 1, Brooke A Millar 1, Sam Perez 1, Jeff Carter 1, Carlo Wood 1, Srinivasan ThyagaRajan 1, Christine Molinaro, Cheri Lubahn 2, Dianne Lorton 2
PMCID: PMC3551630  NIHMSID: NIHMS65672  PMID: 18308299

Abstract

Optimal host defense against pathogens requires cross-talk between the nervous and immune systems. This paper reviews sympathetic-immune interaction, one major communication pathway, and its importance for health and disease. Sympathetic innervation of primary and secondary immune organs is described, as well as evidence for neurotransmission with cells of the immune system as targets. Most research thus far as focused on neural-immune modulation in secondary lymphoid organs, and have revealed complex sympathetic modulation resulting in both potentiation and inhibition of immune functions. SNS-immune interaction may enhance immune readiness during disease- or injury-induced ‘fight’ responses. Research also indicate that dysregulation of the SNS can significantly affect the progression of immune-mediated diseases. However, a better understanding of neural-immune interactions is needed to develop strategies for treatment of immune-mediated diseases that are designed to return homeostasis and restore normal functioning neural-immune networks.

Keywords: Noradrenergic, autonomic, sympathetic innervation, lymphoid organs, immune modulation

1. Introduction

Autonomic (mainly sympathetic) efferent nerves innervate primary (bone marrow and thymus) and secondary (spleen and lymph nodes) lymphoid organs, providing a conduit for the brain to alter immune reactivity. The origin, pattern of distribution and targets of sympathetic nerves in primary and secondary lymphoid organs across life span are reviewed here. Sympathetic nerves release norepinephrine (NE), as their primary neurotransmitter, into the lymphoid microenvironment to affect the functioning of cells of the immune system. Thus, noradrenergic (NA) influences on immunity, while still not entirely understood, have been the most extensively investigated. Neural regulation of immune function by peptide neurotransmitters that co-localize with NE, such as neuropeptide Y (NPY), adenosine triphosphate (ATP), opioid peptides, corticotropin-releasing hormone (CRH) and vasoactive intestinal peptide (VIP), and their affect NA-immune modulation is much less understood. Studies reporting the expression and location of the neurotransmitter-specific receptors on immune cells and ligand-receptor mediated intracellular signaling to alter immune responses are also described here. Finally, we discuss functional and clinical significance of aging-induced changes in sympathetic-immune interactions and the consequences of sympathetic dysregulation in the development and progression of immune-mediated diseases such as rheumatoid arthritis, infections, cancer, and after major injury.

2. Sympathetic Neurotransmission in Bone Marrow

Lymphohematopoietic stem cells in the bone marrow replenish the immune cells in the adult immune system throughout life. Regulation of hemato- and lympho-poiesis via the brain-immune signaling occurs through nerves that innervate bone marrow cells, as well as neuroendocrine hormones that circulate in the blood. Efferent sympathetic nerves enter the nutrient foramina of long bones, course along blood vessels in the Haversian and Volkmann’s canals to distribute to bone marrow [17]. NA sympathetic nerves provide the densest innervation of rat bone marrow and contain NE, NPY, and VIP. Sympathetic nerves immunoreactive for tyrosine hydroxylase (TH), the rate-limiting enzyme for NE synthesis, are more abundant than those containing NPY and VIP, however, the peptide-containing nerve fibers display a similar pattern of distribution [2].

NA nerves closely associate with hematopoietic and stromal cells in the bone marrow [2,49]. Sympathetic nerve terminals directly appose periarterial adventitial cells, a particular type of stromal cell, that is an important source of growth factors and adhesion molecules [8]. Periarterial adventitial cells interconnect via gap junctions with sinus adventitial reticular cells and intersinusoid reticular cells, which are also apposed by these efferent nerves. Collectively, these findings provide anatomical evidence for sympathetic regulation of hematopoiesis and lymphopoiesis in bone. The findings that temporal development of rat bone marrow innervation correlates with the onset of hemopoietic activity [10] provides additional support for sympathetic nervous system (SNS) regulation of bone marrow function. The SNS also innervates the vasculature to regulate vasomotor activities and the release of mature blood cells from the bone marrow.

In mice, NE concentration ranges from 1–3 ng per g in bone marrow tissue [11,12], 1 to 2 orders of magnitude lower than generally found in rat secondary lymphoid organs [13,14]. NE concentration and its levels and metabolites exhibit diurnal variations in murine bone marrow, with levels peaking at night [11]. Further, NE content, but not epinephrine (EPI), positively correlates with the proportion of cells in the G2/M and S phases of cell cycle [11]. Sympathetic nerves in the bone marrow respond to generalized systemic stressors that increase NE turnover. Exposing mice to cold temperatures increases NE turnover rate by 36%. More impressive is that a primary immune challenge, peritoneal Pseudomonas aeruginosa infection, increases NE turnover rate in the bone marrow by 131% [15].

Functional assays and radioligand binding studies indicate that marrow cells express functional α-adrenergic receptor (AR) [16]. 3H-labeled prazosin, an α1-AR antagonist, binds to both bone marrow cell membranes and intact bone marrow cells with high and low affinity. Lymphoid/stem cell fractions express the high-affinity binding site, but the cell subset that expresses the lower affinity site is not clear. β-AR expression has not been demonstrated on bone marrow cells using radioligand binding, northern blot or reverse transcription-polymerase chain reaction (RT-PCR) techniques. However, both in vivo and in vitro functional studies demonstrating β-AR signal transduction support their expression on bone marrow cells. In mice exposed to NE, EPI, or isoproterenol (β-AR agonist) [17,18], intracellular adenosine 3’, 5’-monophosphate (cAMP) content (second messenger for noradrenergic signaling via β-AR) biphasically increases in bone marrow cells 1 and 15 min later. Similarly, intracellular cAMP concentration rises in bone marrow cells in mice sublethally irradiated [19]. In both cases, the nonselective β-AR antagonist, propranolol blocked the rise in intracellular cAMP content, suggesting a specific β-AR-mediated effect.

Embryonic and fetal development of hematopoietic system occurs in multiple organs including, yolk sac, liver, thymus, spleen, lymph nodes and red bone marrow. After birth, hematopoiesis is largely confined to red bone marrow, with clonal expansion occurring in lymphoid tissue. Stem cells of T lymphocytes migrate from the bone marrow to the thymus to become immunocompetent. After maturation in the bone marrow or thymus, immune cells circulate using the blood vascular and lymphatic systems as conduits to patrol tissues. Specific growth factors, cytokines and cell-to-cell contact regulate hemato- and lymphopoiesis. Sympathetic nerves that supply bone marrow may directly or indirectly affect stem cell development and differentiation by affect the release of signaling molecules that guide these processes. This role for sympathetic nerves is supported by anatomical and pharmacological studies.

Both α-AR and β-AR antagonists provide protection against radiation-induced bone marrow cell death [20, 21]. The protective effects of these antagonists are time dependent with respect to radiation treatment. β-AR antagonists are protective when administered prior to irradiation, while the α-AR antagonist provide more effective protection after radiation exposure. In irradiated mice treated with the β-AR agonist, isoproterenol, proliferation of bone marrow cell increases 16 h later [18]. These early studies suggest that catecholamines stimulate proliferation of bone marrow cells rendering them more vulnerable to the toxic effects of irradiation. Findings using a 3-dimensional culture system support these early studies. Adding isoproterenol to murine bone marrow cultures increases cellular proliferation and granulopoiesis dose-dependently [3], while the addition of either a nonselective β-AR (propranolol) or a selective β2-AR (butoxamine) antagonist to cultured human bone marrow cells reduces cellular proliferation by the number of cells entering the S phase, and slows granulocyte-macrophage colony (GM-CFU) formation [22]. These data also support a temporal expression of α- and β-AR subpopulations on bone marrow cells, whereby β-AR expression predominate during early bone marrow cell activation, and α-AR expression increases during later stages.

β-AR activation affects both proliferation and differentiation of bone marrow cells. Albuterol, a selective β2-AR agonist, added to bone marrow cells cultured from Friend leukemia virus-infected mice inhibited the formation of erythroid colony forming units (CFU-E) [19], an effect that was reversed by addition of the selective β2-AR antagonist, butoxamine. Restraint stress induced an increase in both the proportion and number of specific subpopulations of bone marrow cells, an effect partially reversed by pretreatment with RU-486, a steroid receptor antagonist, and profoundly augmented by chemical sympathectomy with 6-hydroxydopamine (6-OHDA) or in vivo exposure to propranolol [23]. These data taken together suggest that restraint stress induces tissue-specific changes in the immune-cell distribution, that both stress pathways are important for this effect on bone marrow, and that corticosteroids (enhancing) and catecholamines (suppressive) may act in opposition in regulating immune cell accumulation in bone marrow.

Studies support the modulation of hematopoiesis by functional α1-AR on bone marrow cells [reviewed in 24]. Reconstituting NE-depleted mice with syngeneic bone marrow cells increases GM-CFU formation and accelerates myelopoiesis. Similar effects occur in reconstituted, sympathetically-intact mice exposed to prazosin, an α1-AR antagonist, but not to propranolol (β-AR antagonist) [25]. Prazosin treatment in these mice reduces the differentiation of transferred precursor cells into thymocytes and splenic T and B cells [26]. Bone marrow cells treated with NE or methoxamine (α1-AR agonist) reduces the number of GM-CFU, an effect blocked by prazosin [16,26]. Treatment with an α2-AR agonist has a similar, but smaller effect [16]. Collectively, these data suggest that treatment of bone marrow cells with α-AR agonists suppress myelopoiesis and augment lymphopoiesis. Whether NE released from NA nerves exerts these effects directly on stem cells or indirectly by influences bone marrow stromal cells, or both is not known.

The SNS also regulates the migration of immune cells from the bone marrow into the periphery. Within 24 h after transecting the femoral nerve, or after chemical sympathectomy with 6-OHDA, bone marrow cellularity decreases [9]. This effect is due to the egress of mature and progenitor hematopoetic cells into the peripheral blood [9]. In another study, NE mobilized fat from the bone marrow [27]. In contrast, Benestad et al. [28] report no effects of neonatal chemical sympathectomy, surgical denervation of the hind limb, or electrical stimulation of the hind limb on blood flow, bone marrow cellularity, or efflux of tibial marrow cells into the circulation. While these data are controversial they provide evidence that the SNS influences the mobility of bone marrow precursor cells into the peripheral circulation.

3. Sympathetic Neurotransmission in the Thymus

The thymus is the primary site for differentiation and maturation of T lymphocytes, whereby they can selectively recognize and respond appropriately to foreign antigen – a process call T cell “education”. Developing and differentiating thymocytes do not respond to foreign substances, because foreign antigens cannot cross the blood-thymic barrier to enter the thymic microenvironment. Intrathymic T cell precursors undergo a complex series of programmed developmental changes resulting in the production of mature, self-major histocompatibility complex (MHC) restricted, single-positive (SP) T lymphocytes (express either CD4 or CD8 on their cell surface). The earliest thymocytes lack detectable CD4 and CD8 (double negative or DN cells). The majority of DN thymocytes rearrange the T cell receptor (TCR) genes, then transiently express both CD4 and CD8 (double positive or DP thymocytes) before differentiating into mature SP thymocytes. During the selection process most of the immature thymocytes undergo apoptosis, but thymocytes that successfully mature, enter the blood and home to secondary lymphatic tissues. Intrathymic T cell development is controlled by numerous factors, including cytokines, neuroendocrine hormones and locally released neurotransmitters.

The thymus of young adult animals is richly innervated by the SNS [2938]. Thymic NA nerves originate primarily from the superior cervical and stellate ganglia [30,39,40]. Dense plexuses of NA nerves enter the thymus with large blood vessels, coursing into the capsule and interlobular septa. From these vascular nerve plexuses, smaller vascular plexuses diverge into the cortex. NA nerves exit these plexuses to enter parenchymal regions of both the outer and deep cortex where they reside adjacent to stromal cells and thymocytes. NA nerves predominate in the cortex, with a much lower density present in the medulla. A slightly higher density of NA nerves reside near the corticomedullary junction where the more mature thymocytes localize. This is consistent with autoradiographic data demonstrating increased β-AR density in the inner thymic cortex [41]. There is also a greater density of yellow autofluorescent cells, a macrophage-like cell population, in the inner cortex that closely associates with NA nerves. In the deep cortex and medulla, NA nerves are adjacent to the thymic epithelial cells (TEC) [42]. NA nerves coursing in the interlobular septum continue along the venous sinuses, a prominent feature of the thymic medulla [4,5,31,35,38,43,44,45], with the occasional nerve fiber extending from the septa and sinuses into the medullary parenchyma. In the capsule and interlobular septa, NA nerves course adjacent to mast cells [57,46], CRH-immunoreactive cells [7,47], and ED3+ macrophages [7]. NA nerves also course in close proximity to ED1+ corticomedullary macrophages [46].

The tonic release of NE from NA nerves in the thymus and spleen is tightly regulated by numerous receptors types including prejunctional α-AR, muscarinic and nicotinic cholinergic receptors, P1-purinergic and prostaglandin E2 (PGE2) receptors [36,48,49]. NE release from NA nerves feeds back to inhibit its own release via interaction with prejunctional α-AR. Treatment with the α1-AR antagonist prazosin or an α2C-subtype selective AR antagonist [50] augments stimulation-evoked release of NE in vitro [36,48,49]. Stimulation of prejunctional muscarinic receptor also inhibits the release of NE; but activation of prejunctional nicotinic receptors increases the stimulation-evoked release of NE from sympathetic terminals [51,52].

β2-AR are expressed on thymic cell membranes as demonstrated using radioligand binding studies and Northern blot analysis [41]. Compared to mature T cells, unfractionated thymocytes express very low levels of high-affinity β-AR [53]. Mature corticosterone-resistant thymocytes express β-AR at levels equivalent to those seen in peripheral T cell populations [54,55]. These data suggest that β-AR expression is limited on immature thymocytes, but increases as thymocytes mature and differentiate. Autoradiographical data which primarily localizes β2-AR expression to the medullary region of the rat thymus is consistent with a maturation-dependent regulation of β-AR expression in thymocytes [41]. However, the limited numbers of β2-AR expressed on the unfractionated thymocytes may be offset by the efficiency of receptor-coupling to adenylate cyclase, which is greater in the unfractionated thymocytes compared with spleen or lymph nodes [53]. This suggests that thymocytes that do express β-AR are exquisitely sensitive to β-AR agonist signaling.

Changes in biological functions can induce changes in the number of AR expressed on cells in the thymus. For example, β2-AR receptor surface expression on thymocytes changes over the course of an immune response [56]. Thymocytes express fewer β2-AR 3 days, but higher β2-AR number 7 and 15 days after immunization with the protein antigen, bovine serum albumin, compared with naïve controls. The mechanism for immunization induced changes in β2-AR gene expression in the thymus is not known. Changes in hormone levels can also alter the number of β2-AR expressed on rat thymocytes. In the estrous phase of female reproductive cycle, and during pregnancy, β2-AR expression increases, while castration decreases receptor density [41,56] suggesting that steroid sex hormones modulate β2-AR expression on thymocytes. Additionally, in vitro exposure of rat TEC to isoproterenol, a nonselective β-AR agonist, reduces basal and serum-stimulated proliferation, without affecting interleukin (IL)-1 production [34], suggesting the expression of β2-AR on this thymic cell population as well.

Based on Northern blot analysis and RT-PCR for mRNA, radioligand binding, and cAMP induction, TEC, which are essential for positive and negative selection in the thymus, also express α1- and α2-AR [34]. The addition of an α-AR agonist to unstimulated or lipopolysaccharide (LPS)-stimulated cultures of rat TEC dose-dependently increases IL-6 production, but has no effect on IL-1 production. This effect is blocked by an α-AR antagonist [57].

The regulation of thymic function by the autonomic nervous system has been explored by numerous laboratories. Early studies in the 1970s and 1980s allude to β-AR-induced enhancement of thymocyte differentiation and suppression of thymocyte proliferation in the presence of intact NA innervation [5863]. Isoproterenol exposure in vivo reduces thymic weight and thymocyte number in mice [64]. Similarly, catecholamines or isoproterenol decrease concanavalin A (Con A)- or LPS-induced proliferation of murine thymocytes, an effect not observed with α-AR agonist treatment [65]. Although there are no reports directly relating β-AR stimulation and the induction of apoptosis, studies have demonstrated that elevating thymocyte intracellular cAMP induces apoptosis [6668]. Thymocyte proliferation increases with treatment with high concentrations of either EPI or isoproterenol [69] or low concentrations of NE. It has been proposed that signaling differences exist between the signaling pathways induced by β- or α-AR, assuming that NE stimulates the α-AR in this study. Thus, second messenger signaling data support that stimulation of β2-AR on cells within the thymus elevates the level of intracellular cAMP, with the possibility that α-AR stimulation by NE induces a rise in intracellular Ca2+.

Thymic weight and cellularity decrease, apoptosis increases, and the number of T cells proliferating in the periphery are reduced following chemical sympathectomy [35], suggesting that catecholamines exert a positive effect on cell viability and function in the thymus. Chemical sympathectomy in mice also increases thymocyte apoptosis and reduces spontaneous proliferation in vitro [70,71]. NA nerve ablation is necessary to induce this response as pretreatment with desipramine blocks the 6-OHDA-induced increase in apoptosis [71]. However, this effect could be a direct effect of 6-OHDA on thymocytes as incubation of thymocytes in vitro with 6-OHDA (10−5 M) also induces apoptosis, an effect prevented by addition of desipramine to the cultures. These findings suggest that thymocytes may possess catecholamine uptake mechanisms. Therefore, chemical sympathectomy-induced changes in the thymus must be interpreted with caution, because the thymus is not depleted of NE as completely as other tissues, such as the spleen [72]. Furthermore, the possibility that 6-OHDA-induced elevation in corticosterone drives the sympathectomy effects in the thymus has not been adequately addressed. The latter, however, seems unlikely as Delrue-Perollet et al. [70] has shown that corticosterone levels are not altered in sympathectomized animals at the time of sacrifice (3 days after the first injection of 6-OHDA). Together, these data suggest that NA innervation in the thymus can influence thymocyte development.

Sympathetic modulation of thymocyte development and maturation has been evaluated recently using a variety of pharmacological approaches. β-AR blockade with propranolol for 4 or 16 days increases thymocyte proliferation and apoptosis, and causes disturbances in kinetics of thymocyte differentiation, expanding the most mature SP (CD4+, CD8+) thymocyte pool, and increasing the relative number of CD8+CD25+ cells [73,74] in adult male Dark Agouti rats. In contrast, β-AR blockade has no effect on the relative proportion of CD4+ and CD8+ peripheral blood lymphocytes (PBL). Prolonged exposure to the β-AR antagonist augments these responses. Treatment with anabolic doses of clenbuterol, the β2-AR agonist, reduces thymocyte average nuclear area and increases the apoptotic index [75], supporting an immunosuppressive role for β2-AR agonists. Collectively, these studies support the feasibility of using pharmacological agents that target AR as a means of modulating T cell development (and hence T-dependent immune responses). They also provide specific insight into the role of β-AR in T cell maturation.

NE differentially modulates thymocyte maturation in young and aged mice [76], an effect that may be related to increased sympathetic innervation of the aged, involuted thymus of both in rats and mice [31,77]. To study age-associated changes in β-AR modulation of thymocyte differentiation, Madden and Felten [76] have implanted 2- and 18-month-old BALB/c mice with pellets subcutaneously containing the non-selective β-AR antagonist, nadolol. β-AR blockade alters thymocyte CD4/CD8 co-expression in old, but not in young mice. The frequency of the immature DN CD48 population increases, and the intermediate DP CD4+8+ population decreases in nadolol-treated old mice. There is a corresponding increase in the frequency of mature SP CD48+, but not SP CD4+8 cells. This increase in SP CD48+ cells is most likely not mediated by increased positive selection as CD3high expression in the DP CD4+8+ population is not altered by nadolol. In all probability the increase is due to an increase in the numbers of SP CD48+ cells exported from the thymus to the periphery as the percentage of CD8+44low naive cells in peripheral blood increases in nadolol-treated mice. These results indicate age-associated differences in sympathetic modulation of thymocyte maturation. Pharmacological manipulation of NA innervation may provide a novel means of increasing naive T cell populations and improving T cell diversity, thereby, enhancing T cell responses to novel antigens with age.

Chronic treatment (0.20 mg/kg body weight/day for 15 consecutive days, s.c.) with urapidil, an α1-AR antagonist, reduces body weight gain and thymic weight in immature peripubertal rats. Reduced thymic weight in treated rats results from decreased thymocyte numbers and thymic cortical volume compared to age-matched saline-injected controls [78]. The percentage of SP CD4+8 thymocytes is lower, while that of SP CD48+ is higher suggesting dysregulation in the final steps of the positive selection. In contrast, thymic weight, cortical size and cellularity increase after treatment with an α1-AR blocker in adult rats. There is also a decelerated transition from the DN to DP stage of thymocyte development as the percentage of immature DN CD48 cells increases, whereas with percentage of immature CD4+8+ DP thymocytes decreases. The treatment also evokes changes in the relative numbers of SP cells in adult rats, but contrary to immature animals, the maturation of SP CD4+8 is favored over SP CD48+ thymocytes. These findings demonstrate an age-dependent effect of chronic α1-AR blockade on thymic structure and thymocyte differentiation. Further, targeting α1-AR pharmacologically modulates intrathymic T cell maturation under conditions where immunocompetence is impaired, such as in HIV infection and aging.

Xylazine, an α2-AR agonist, both inhibits and promotes thymocyte proliferation depending on its concentration. Treatment with xylazine, an α2-AR agonist, in vivo or applied to unfractionated thymocytes in culture stimulates proliferation triggered by Con A [79]. In contrast, higher concentrations of xylazine, both in vivo and in vitro, are inhibitory [80]. There is also a decrease in IL-2 production (in vivo and in vitro) and down-regulation of IL-2 receptor alpha (IL-2Rα) expression (in vitro). Exogenous IL-2 completely restores the inhibitory effect of xylazine in vivo. However, exogenous IL-12 has a minimal influence on the high-dose xylazine-inhibited thymocyte proliferation in vitro. This stimulatory effect of xylazine on proliferation of thymocytes is mediated through α2-AR, since it is blocked by yohimbine, an α2-AR antagonist. It seems that the pathways involved in inhibition of thymocyte proliferation by high-dose xylazine are more complex because the xylazine-suppressed thymocyte proliferation is potentiated by yohimbine. Furthermore they indicated that these effects are not mediated via α2-AR. Suppressed thymocyte proliferation induced by high-dose xylazine may be mediated by increased cell death, since concentrations of xylazine between 100 and 500 µM induce apoptosis of rat thymocytes in vitro. Xylazine, at concentrations higher than 50 µM, also induce apoptosis of a thymocyte hybridoma (BWRT8), and increase TCR cross-linking-induced apoptosis. Xylazine-induced apoptosis of the BWRT8 hybridoma is not prevented by yohimbine (a selective α-adrenergic antagonist) or by antibodies to Fas and Fas-L. However, cell death is completely blocked by a caspase inhibitor, z-Val-Ala-Asp (OMe)-CH2F. Cyclosporine, a calcineurin blocker, partly inhibits the xylazine-induced apoptosis of activated BWRT8 cells. These data taken together suggest that the β-AR and α-AR are expressed on cells in the thymus and that β-AR stimulation of these cells may exert a suppressive effect, while α-AR stimulation may have a positive effect on thymocyte differentiation.

Findings that NA nerves run in close contact with TEC and TEC express β-AR [42,34,36] suggest that the SNS could also have indirect effects on thymocyte maturation. EPI from the adrenal gland or NE released from perivascular nerves may target the outer thymic cortex through the TEC that form the blood-thymus barrier. TEC create a microenvironment that influences the maturation and differentiation of thymocytes. The demonstration of their capacity to respond to catecholamines suggests that adrenergic stimulation may interfere with the regulation of immune functions. In particular, catecholamines influence the synthesis of IL-6, which is known to affect T cell proliferation and differentiation [81]. Co-stimulation of cultured rat TEC with EPI, NE, or isoprenaline has an additive (tumor necrosis factor (TNF)-α) or synergistic (LPS) effect on IL-6 release mediated by β-AR that is linked to the elevation of intracellular cAMP levels [57]. A close association between mast cells and NA nerves exists in the thymus, which suggests a possible role for NE regulating mast cell function to affect thymocyte development in the thymus. These nervous system-mast cell contacts are seen in all lymphoid organs, with the exception of the spleen [82], and NE, via stimulation of α- and β2-AR, is known to stimulate and inhibit, respectively, the release of histamine from mast cells [83,84]. Regulation of mast cell histamine release is another possible mechanism through which NE released form postganglionic nerve terminals may exert an immunomodulatory role on immune function.

4. Sympathetic Innervation of Secondary Lymphoid Organs

4.1. The spleen

The spleen receives a rich supply of sympathetic nerves primarily from the superior mesenteric and celiac ganglionic plexuses [8587] and to a lesser extent, the sympathetic trunk [86,88]. In fact approximately 98% of the nerve fibers in the splenic nerve are sympathetic [89]. In the rat spleen, NA innervation primarily develops postnatally [9091]. The splenic nerve enters the spleens as perivascular plexuses coursing along the splenic artery, in the splenic capsule and trabeculae in the mature animal [7,29,43,9294]. NA nerves in vascular and trabecular plexuses continue into the white pulp primarily associated with the central arterioles and their branches. NA varicosities labeled using immunocytochemistry for TH radiate from these plexuses into the periarterial lymphatic sheaths (PALS) [43] ending among OX19+ (pan-T cell marker) T lymphocytes and interdigitating cells [4,5,43,90,95100]. NA nerves that enter the marginal and the parafollicular zones distribute among IgM+ B cells and ED3+ macrophages [4,5,43,90,95100]. Few NA nerves course in the parenchyma of the red pulp; most NA nerves present in the red pulp course as plexuses along the trabeculae and venous sinuses [97,100]. NE-containing nerves that distribute to the spleen also express acetylcholinesterase [101], since surgical or chemical ablation, but not bilateral sub-diaphragmatic vagotomy, prevent histochemical staining of acetylcholinesterase+ nerve profiles [85,86,101]. NPY and enkephalin also colocalize with NE in splenic NA nerves [7,102,103].

An ultrastructural study of spleen sections immunocytochemically stained for TH has revealed TH+ nerve terminals in direct contact with lymphocytes in the PALS and marginal zone [99,104,105]. These NA terminals have long, smooth zones of contact with lymphocyte and macrophage plasma membranes, separated by as little as 6 nm. These terminals are not present in spleens from chemical sympathectomized or surgical removal of the superior mesenteric-celiac ganglionic complex with 6-OHDA or after ganglionectomy.

The spleen is hyperinnervated, particularly the marginal zone of the white pulp in transgenic mice that overexpress nerve growth factor (NGF) in the skin and other epithelial structures compared to wild type mice [106]. This finding is consistent with the neurotrophic role of NGF in sympathetic neurons. Spleen cells from NGF transgenic mice display lower splenic mitogen-induced proliferative responses, suggesting that increased NGF expression and/or NGF-induced increase in sympathetic innervation have consequences for immune responses. In contrast, with advancing age splenic NA nerves are lost in Fischer 344 (F344), Lewis and Brown-Norway rats concomitantly with a decline in splenic NGF concentrations [107]. Additionally, changes in NGF concentrations in the red and white pulp of the spleen are associated with the redistribution of NA nerves in experimental arthritis [108].

4.2. The lymph nodes

The distribution of sympathetic nerves has been described in the lymph nodes from rats [7,100,109113], several strains of mice [5,110,111,113,114], cats, pigs and humans [110]; however, the origin of the NA nerves in lymph nodes is still unclear. It is presumed that their innervation is regionally supplied as lymph nodes are regional structures draining the areas with which they are associated. For example, in the rat when the superior cervical ganglia are removed, sympathetic nerves are lost in the cervical nodes [109]. NA nerves supplying the cervical, mesenteric and popliteal lymph nodes course along the vasculature that provides their blood supply [5,7,100,109114]. Sympathetic nerves enter lymph nodes along with blood vessels at the hilus, and continue into the subcapsular plexus or perivascular plexuses in the medullary cords. NA nerves entering the medulla also course adjacent to blood and lymphatic vasculature, where they enter into paracortical and cortical regions. There are also a large number of NA nerves that distribute to medullary and paracortical areas independently of the blood vessels. They supply the T cell-rich regions, such as the paracortical and cortical zones, but not the B cell-containing areas such as lymph nodules and germinal centers. Electron microscopic studies reveal NA nerves establishing contacts with the vascular smooth muscle, reticular cells, plasma cells and lymphocytes in the lymph nodes [115,116]. These nerve fibers contain at least 3 types of synaptic vesicles, each presumed to contain a different neurotransmitter. Similar to the spleen of NGF transgenic mice, peripheral lymph nodes that drain the skin where NGF is overexpressed are more densely innervated compared with control mice and with mesenteric lymph nodes, lymph nodes that do not drain the skin) [106]. The NGF concentration in peripheral lymph nodes from NGF transgenic mice is 13-fold higher than control mice. These findings demonstrate the potential for plasticity of this nerve population under the influence of neurotrophic support by NGF.

4.3. Mucosal-associated lymphoid tissues

The wet epithelial surfaces of the respiratory, urogenital, and gastrointestinal tracts are associated with the majority of lymphoid tissue as these sites are continuously exposed to an external environment of antigens from pathogens, food, and particles in the air. The lamina propria of the intestines receives an extensive plexus of nerves arising from the enteric nervous system, the sympathetic and parasympathetic systems, and the dorsal root ganglia. The relationship between nerves and immune compartments in mucosal-associated lymphoid tissues is not well studied compared with other lymphoid organs, despite the importance of these sites in host defense. Depending on the specific neurotransmitter they contain nerves have a precise location in the lamina propria. Nerves that contain NE extend throughout the lamina propria and form a dense plexus around the epithelial crypts, and ramify throughout the villi in the ileum [7]. TH+ nerves extend beneath the basement membrane of the epithelium and somatostatin-positive nerves course in pericryptal networks, but are infrequently found in the villi. In this region, capillaries supplying the villus coalesce to form postcapillary venules, suggesting a role for these nerves in trafficking of activated effector blast cells, and unactivated effector cells from the blood stream into the mucosa [117119].

Double-label immunocytochemistry demonstrates the spatial relationships between NA nerves and subsets of immunocytes in the rat ileum [7]. These NA nerves (TH+) reside in close proximity to OX19+ T cells and with CD8+ T cells scattered throughout the lamina propria. TH+ nerves course adjacent to the basement membrane of the epithelium in close proximity to CD8+ intraepithelial lymphocytes and closely appose ED3+ macrophages in the lamina propria. Similarly, enteric nerves contact plasma cells and B immunoblasts in the mucosa and submucosa [120].

Aggregated lymphoid tissues located in the gut also receive a rich nerve supply, including the tonsils, Peyer’s patches, and appendix. NA nerves distribute among immunocytes in the palatine tonsils and paratonsillar glands in man [121,122], and the appendix [123], sacculus rotundus, and Peyer’s patches in the rabbit [124]. Similar to the spleen and lymph nodes, these nerves course along the vasculature, ramify into T cell compartments, are scarce in lymph nodules (B cell compartments), and closely appose lymphocytes, enterochromaffin cells, plasma cells and other accessory cells in these compartments.

In bronchus-associated lymphoid tissue, NA nerves also closely appose cells of the immune system [125]. These nerves are associated with smooth muscle layers, and travel along the vasculature, under the epithelium and among immune cells in the bronchus-associated lymphoid tissue (BALT) parenchyma. TH+ nerves preferentially supply blood vessels, enter the central zone of the BALT, and ramify within the parenchyma. Peptidergic and NA nerves form contacts with mast cells, ED1+ macrophages, and other lymphoid cells with varying frequency.

5. AR expression on Cells of the Immune System

5.1. T Lymphocytes

Human and murine T lymphocytes express β2-AR [54,55,126140]. T helper (Th)1 and Th2 cell clones generated from naïve cells differentially express β2-AR [141]. In contrast, normal T cells do not possess functional high-affinity β1-AR or β3-AR. Collectively, these reports indicate a range of 500–2500 binding sites on CD8+ T lymphocytes, and 200–750 binding sites on CD4+ T lymphocytes. Resting Th1 effector cell clones express approximately 250 binding sites per cell with a Kd of 100 pM; however, Th2 clone do not appear to express β2-AR [140,142].

Studies using activated splenic or lymph node T cells have reported either increased [55,134,143] or decreased [55,144] β2-AR expression 24 h after activation with either Con A or phorbol 12-myristate-13-acetate (PMA)/calcium ionophore. Contact sensitization increases expression of β-AR in murine draining lymph node cells [145]; however, immunization with a Th cell-dependent antigen significantly decreases β-AR expression in unfractionated spleen cells [54]. Variability in the absolute numbers of β-AR per cell reported may result from different experimental conditions, such as the T cell isolation techniques, source of T cells, types of radiolabel used to tag pharmacological ligands, using ligands with different affinities, or specific activity of the radioligand. β2-AR number increases over time with Th1 effector cell activation via stimulation of the CD3 complex associated with the TCR, while β2-AR remain undetectable in activated Th2 effector cells [139]. Collectively, these studies indicate the expression of β2-AR on CD4+ Th1 cells and CD8+ T cells and differential expression of β2-AR on murine Th1 and Th2 cells.

Treatment of CD4+ T cells with NE, EPI, isoproterenol (non-selective β-AR agonist) or terbutaline (selective β2-AR agonist) increases intracellular cAMP concentrations [127,128,131,135137,146148] and adenylate cyclase activity via β-AR mechanisms [146,149]. Elevating cAMP inhibits either mitogen- or anti-CD3 antibody-induced T cell proliferation [150156], providing an explanation of β-AR agonist-induced inhibition of T cell proliferation described above. Further, Th1 clones, but not Th2 clones, increase cAMP concentrations after treatment with terbutaline [140]. These data suggest that stimulating β2-AR on T cells increase the activity of protein kinase A (PKA), which activates adenylate cyclase to increase cAMP intracellularly.

5.2. B Lymphocytes

Based on radioligand binding and RT-PCR studies, B cells express a higher density of β-AR than CD4+ Th cells [54,126,129,131,133,157159]. These receptors are of the β2-AR subtype [54,129,131,160] and their stimulation also elevates intracellular concentrations of cAMP [127,131,135137,148,160]. B7-2 expression is enhanced following β2-AR stimulation, an effect inhibited by a PKA inhibitor, suggesting the increased cAMP is PKA-dependent [161]. The enhancement or inhibition of B cell proliferation through the β2-AR is dependent on the type of mitogen used. Receptor stimulation and/or elevation of cAMP inhibit LPS-induced B cell proliferation [151,155] and production of anti-immunoglobulin (Ig) antibodies [162165] through mechanisms that inhibit phosphatidyl inositol hydrolysis and phosphorylation of the phospholipase C-γ1 [162,164,166]. However, activation of β2-AR and increases in cAMP enhances ionomycin-induced B cell proliferation [163,165,167].

5.3. Natural Killer (NK) Cells

Radioligand binding [133,168,169] and functional studies [169,170174] indicated that β2-AR and α1- and α2-AR are expressed on NK cells. Stimulation of the β2-AR on NK cells is also linked to increased intracellular cAMP.

5.4. Macrophages

Radioligand binding studies indicate α2- and β2-AR are expressed on normal macrophages [175181]. The density of β2-AR range from approximately 1,000 or 23,000 binding sites per cell with a Kd of 50 or 800 pM in rat peritoneal [176] or guinea pig alveolar macrophages [180181], respectively. β2-AR stimulation elevates cAMP in a concentration-dependent manner in macrophages from a variety of locations and different species [176,177,181187]. Macrophage β-AR stimulation also inhibits activation of Ca2+-dependent K+ channels [177], while stimulation of α1-AR on macrophages activates Ca2+-dependent K+ channels by mobilizing intracellular Ca2+ stores, presumably via the activation of phospholipase C and a G-protein-dependent mechanism [188]. Activation of Ca2+-dependent K+ channels is important for modulating macrophage phagocytic activity and may explain the α-AR-mediated enhancement of macrophage phagocytic activity [189]. The functional effects of stimulation of AR on macrophages are dependent on the subtype of receptor. β2-AR stimulation suppresses macrophage activity, whereas α2-AR stimulation enhances it [reviewed in 190].

6. Functional Significance of Sympathetic Innervation of Secondary Lymphoid Organs

Immunocompetent cells in secondary lymphoid organs provide for host defense against pathogens. Foreign substances in the blood are filtered through the spleen. Blood flows through the white pulp and continues into the red pulp where macrophages remove foreign substances, and signal other cells of the immune system to help with removal. Like the spleen, lymph nodes filter regionally draining lymph, with the purpose of detecting and removing bacteria and foreign materials. Diffuse lymphatic tissue under wet epithelial surfaces contains dispersed and aggregated lymphocytes, macrophages and other immune cells. In these locations they survey and detect foreign materials and pathogens that enter from the external environment. Thus, microorganisms or other foreign substances, upon detection, can initiate an immune response in the respiratory, urogenital or gastrointestinal tracts, or in the blood or lymph. Activation of the innate immune responses mobilizes cellular and immune mediators to sites of injury or infection. After activation of an acquired immune response, clonally expanded pools of lymphocytes enter the blood stream from the spleen and/or lymph nodes. Subsequently, they leave the blood and enter sites of infection or injury. Sympathetic signaling within lymphoid compartments modulate immune responses to antigens by affecting clonal expansion, cytokine production and/or responsiveness of target cells by altering receptor expression, shifting the balance between cellular and humoral responses, enhancing or inhibiting the inflammatory response and mobilizing immunocytes.

In vitro, adrenergic agonists can modulate all aspects of immune responses (initiative, proliferative and effector phases). Stimulation of either β- or β2-AR inhibits mitogen- or anti-CD3 antibody-induced T cell proliferation [146,149,191196], and particularly inhibits naïve T cells differentiation into Th1 cells [139141,197,198] by affecting early events involved in the initiation of proliferation [199]. β-AR-mediated inhibition of T cell proliferation may be explained, at least in part, by its ability to inhibit IL-2R expression and/or IL-2 production by activated T cells [139,140,146,191,200206].

β-AR stimulation also affects the production of interferon-γ (IFN-γ), which may contribute to this effect under certain conditions. Pretreating with terbutaline and other pharmacological agents that increase cAMP before antigen challenge, but not at the time of or after activation [139], decreases IFN-γ production from Th1 clones [140,197,198]. In contrast, under Th1-promoting culture conditions, β-AR stimulation drives Th1 effector cell development from naïve T cells. These effector Th1 cells produce greater IFN-γ after restimulation [141]. Under Th2-promoting culture conditions, naïve T cells exposed to NE or terbutaline develop into Th2 cells, but adrenergic agents have no affect on IL-4, IL-5 or IL-10 production [139141]. Other studies support a dichotomy in β2-AR effects on Th1 and Th2 cells, such that their activation modulates Th1 cell differentiation and cytokine production, but does not alter Th2 effector cells [131,197,203,198,207209]. Interestingly, increasing intracellular cAMP levels in Th2 cells can modulate IL-4 production [210213]. Therefore, cAMP-mediated modulation of cytokine production by Th2 effector cells may depend on the manner in which the Th2 cells are activated or the level of cAMP attained intracellularly [190].

In vivo studies indicate that β-AR stimulation modulates Th1 effector cell-driven responses, including delayed-type hypersensitivity. Chemical sympathectomy before sensitization with 2,4,6-trinitrochlorobenzene (TNCB), reduces ear swelling in mice with second challenge compared with controls [145]. Splenic T cell from NE-depleted mice proliferate less and produce less IL-2 and IFN-γ in response to Con A in vitro, but lymph node cells proliferate less, despite greater IL-2 and IFN-γ production [214]. Giving the in vitro data, these findings suggest that NE stimulation of β2-AR may play a role in generating Th1 cell-driven cell-mediated immunity.

CD8+ T cells also express β-AR, and depletion of NE prior to TNCB sensitization reduces hapten-specific cytotoxic T lymphocytes (CTL) generation compared with controls [145]. In vitro studies demonstrate that β2-AR stimulation can enhance [215] or suppress [216218] CTL activity. In the latter study, β2-AR stimulation was important for optimal CD8+ T cell lytic activity. CTL clonal studies indicate that rises in intracellular cAMP can inhibit the TCR-dependent release of granules [219], which may explain β2-AR-mediated inhibition of cytotoxic T cell activity.

β2-AR stimulation has been reported to enhance [220] or inhibit LPS-induced B cell proliferation [195,220] and anti-IgG antibody production [166,220]. Similarly, in vitro Th cell-dependent antibody production increases [220,221] or decreases [13,222225] following β-AR stimulation. These studies suggest that stimulation of β2-AR expressed on B cells can either enhance or suppress humoral immunity. These confusing data may be explained in part, by differences in the signaling pathway induced in B cells by LPS or crosslinking surface Ig compared with the signaling pathway engaged by Th cells, and that these pathways are modulated differently by cAMP [reviewed in 190].

Th cell-dependent antibody production can be modulated by the SNS in vivo, which indicates that NE is likely to be required for optimal antibody production, and may directly affect either B cells and/or Th cells that participate in the response. In vivo studies employing chemical sympathectomy in adult rodents prior to antigen challenge with a T-cell dependent antigen reduces [54,91,145,226228] or enhances [13,229,230] antibody production. Chemical sympathectomy in C57Bl/6 mice, a strain with a predominant Th1 antibody profile, enhances Th1-associated serum IgG2a and Th2-associated IgG1 production after immunization with keyhole limpet hemocyanin (KLH). However, in BALB/c mice, a strain with a preference toward a Th2 profile, chemical sympathectomy only slightly elevates serum IgG1 levels [231]. Direct toxic effects of 6-OHDA, bolus release of NE, and elevated corticosterone levels do not mediate the effects of chemical sympathectomy [232]. Taken together, these data indicate that the effect of NA ablation on specific components of the immune response, including the effector (antibody) stage, is dependent on Th dominance. These studies were the first to use an in vivo model system to address the effect of NE-depletion on Th1- and Th2-dependent antibody production, and they suggest that intact NA innervation inhibits the immune response to KLH. Consistent with these findings, antibody production increases in surgically denervated submaxillary lymph nodes relative to sham-denervated lymph [233]. However, these findings conflict with other in vivo and in vitro studies examining sympathetic modulation of cell-mediated immune responses described above.

In another study [160], severe combined immunodeficient (SCID) mice were used to further assess the role of the SNS in primary antibody response. When mice are chemically sympathectomized before reconstitution with murine β2-ARneg KLH-specific Th2 clones and β2-ARpos splenic naïve trinitrophenyl (TNP)-specific B cells IgM and IgG1 antibody production is suppressed during a primary antibody response. However, during a secondary response to antigen only IgG1 production is suppressed [160]. The histology of the spleen is similar in NE-intact and NE-depleted mice before antigen exposure, but the expansion of the follicle and formation of the germinal center are suppressed in NE-depleted mice after antigen exposure. Similar results have been obtained using SCID mice reconstituted with murine clones of β2-ARpos-KLH-specific Th1 cells and β2-ARpos splenic naïve TNP-specific B cells [160]. Collectively, these findings suggest that β2-AR stimulation of B cells is necessary to maintain an optimal primary and secondary Th2 cell-dependent antibody response in vivo.

The number of IgM-secreting cells increases following NE treatment of murine spleen cells cultured with a Th cell-dependent antigen, an effect blocked by addition of a β-AR antagonist [234235]. This finding suggests that the NE-induced enhancement in IgM production is mediated via β-AR stimulation. The timing of β2-AR stimulation is important for this effect, since addition of NE or terbutaline at the time of antigen challenge or shortly after, but not 24 h later, enhances the IgM response. The ability of β2-AR antagonists to block β-AR-mediated enhancement of IgM production is limited to the first 6 h after the addition of the agonist, indicating that the effects of β2-AR stimulation take place during early stages of the IgM response. The target cell(s) through which β-AR agonists interact(s) to produce this effect on antibody response are unknown since whole spleen cell cultures were used in these studies. Another study [236] suggests that β2-AR agonists affect antibody production by increasing the frequency of B cells that differentiate into IgM-secreting cells, without affecting burst size. Since in this study naïve TNP-specific B cells are stimulated with TNP-KLH in the presence of KLH-specific Th2 clone cells that lack detectable levels of β2-AR either at rest or when activated [139,140], B cells may be affected directly by β2-AR agonists in a B cell-Th2 cell clone culture system. Again, the timing of β2-AR stimulation relative to antigen presentation is important for influencing the antibody response. When Th cells are exposed to terbutaline before activation by antigen-presenting B cells, the Th1 or Th2 cell-dependent antibody response is either inhibited or unchanged, respectively [140]. When B cells are exposed to antigen and terbutaline (i.e., during B cell antigen processing) before interacting with Th2 cells, IgG1 and IgE, but not IgM or IgG2, production are enhanced [161]. Increased B cell responsiveness to IL-4 and B7-2 expression and signaling mediate the β2-AR enhancing effect on IgG1 and IgE production [161]. Further, addition of terbutaline 12 h after Th1 cell-B cell or Th2 cell-B cell interaction increases the IgG2a or IgG1 response, respectively, whereas no significant effects are found when terbutaline is added at other times [140].

Contradictory effects of sympathectomy on immune reactivity may be explained by differences in the Th cytokine responses, differences in the microenvironment of the lymphoid organs draining the site of antigen challenge and/or the nature of lymphocyte activation (i.e., the type of antigen-presenting cells that engages the T cell). Differences in the response to sympathectomy in the spleen compared with the inguinal lymph nodes of nonimmunized mice have also been demonstrated, such that mitogen-induced B cell proliferation is increased in lymph nodes but is reduced in the spleen [214]. Sympathectomy reduces Con A-stimulated spleen cell proliferation, which parallels a decrease in IL-2 and IFN-γ production; however, IL-2 production remains unchanged and IFN-γ production increases in lymph nodes. Denervation-induced changes in lymphocyte migration in inguinal lymph nodes, which is not apparent in the spleen, may explain these differences [237]. The immunological stimulus may also affect the outcome following sympathectomy; as sympathectomy reduces spleen cell Con A-stimulated T cell proliferation, but enhances antigen-specific proliferation in mice [238].

Few studies have revealed an effect of α-AR stimulation on B lymphocyte function [13,143,239244]. α1- or α2-AR stimulation increases or decreases in vitro T-dependent IgM production, respectively [13,143]. Stimulation of α-AR during the effector phase of the antibody response may limit the primary antibody response. In mice treated with amphetamine to stimulate NE release in vivo for 9 days after antigen exposure [241], α-AR stimulation suppresses in vitro proliferation of hen eggwhite lysozyme-primed lymph node cells. It is not possible to rule out direct effects of α-AR agonists on macrophages in this study, since macrophages express α-AR and α-AR-mediated changes in macrophage function subsequently influence the ability of T cells to produce cytokines and proliferate.

Macrophages exposed to antigen rapidly increase consumption of oxygen and generate superoxide anions (O2) and hydrogen peroxide. Production of H2O2 and other reactive oxygen species by macrophages is suppressed by adding a α1-AR agonist to cultured pulmonary macrophages [180,245]. However, since this effect is not reversed by adding an α-AR antagonist, the agonist is most likely functioning as a nonspecific antioxidant [180]. β2-AR stimulation suppresses superoxide anion and H2O2 production in human peritoneal macrophages [246]. Similarly, both α1-AR and β2-AR activation suppresses bovine pulmonary alveolar macrophage production of reactive oxygen [247]. Alveolar macrophages exposed to isoprenaline (β-AR agonist), or the β1-AR agonist dobutamine (which has α-AR effects) also reduces PMA-induced production of H2O2, but the β2-AR agonist, salbutamol, has no effect. Further, neither β1- or β2-AR agonist has an effect on phagocytic activity [180]. Isoprenaline also has no effect on zymosan-induced activation or superoxide production by human alveolar macrophages [248], but does suppress the phagocytosis [249]. These data indicate that stimulation of β-AR may induce different effects on reactive oxygen species and hydrogen peroxide, depending on the population of macrophages under study and the stimulus used to activate macrophages.

Macrophage microbicidal and tumoricidal activities not only are mediated through the production of reactive oxygen species, but also by adhesion and phagocytosis and secretion of regulatory proteins like TNF [250]. Macrophage adhesion is augmented following β-AR agonist treatment [251]; however, the β2-AR agonist treatment suppresses phagocytic activity of murine peritoneal macrophages [176,252]. Similarly, α-AR agonist treatment inhibits phagocytosis [176,253] by enhancing Ca2+-dependent K+ channels activation in macrophages [177]. Treatment with low levels of NE enhances macrophage phagocytosis and tumoricidal activity, suggesting α-AR involvement [254]. Similarly, peritoneal macrophage phagocytic activity was enhanced following treatment with metaraminol and phenylephrine, α-AR agonists, as well as NE [255]. Macrophage α1-AR stimulation also induces the production of complement cascade components [256]. α- and β-AR stimulation appear to have opposing effects on TNF production. NE, EPI and isoproterenol reduce the LPS-induced TNF production in rat spleen macrophages cultures [178]. In contrast, α2-AR stimulation enhances LPS-stimulated TNF production in these cells [175,257]. In vivo studies using chemical sympathectomy in mice indicate that the SNS stimulates the production of TNF by macrophage after LPS challenge [258,259]. Collectively, these findings suggest that α2-AR stimulation augments TNF production and phagocytic activity, whereas β2-AR stimulation inhibits these functions.

Functional AR expressed on NK cells influence their lytic activity. Several studies report that under in vitro conditions, prolonged exposure to NE, EPI, β- or β2-agonists reduce NK cell lytic activity [170172,260]. In contrast, pre-treating NK cells with EPI before adding the target cells enhances lytic activity [260]. There also are reports of a transient enhancement of lytic activity after β2-AR stimulation, which returns to control values within 1 h [173,174]. β2-AR- mediated suppression of NK cell adherence to cultured endothelial cells [261,262] may explain in vivo data showing that administration of NE, EPI or a β-AR agonist increases NK cell circulation [133,174,263]. A similar rise in circulating NK cell numbers occurs with natural stimuli that increase circulating catecholamines, such as acute stress or exhaustive exercise, [261,173,264,265]; an effect that is blocked by pretreatment with β2-AR antagonists [264], but not with β1-AR antagonists [173].

Rats treated with a β-AR agonist in vivo have reduced NK cell lytic activity ex vivo and compromised resistances to growth and metastasis of a NK-sensitive tumor in vivo [266]. Chemical sympathectomy increases the number of metastases, but has no effect on NK cell activity in mice challenged with lung tumor cells [267]. Additionally, CRH-induced sympathetic activation reduces NK cell lytic activity, an effect mediated via β2-AR stimulation [268,269].

Lymphocyte trafficking, which is important for recognition of foreign materials, and upon detection, the rapid mobilization of appropriate leukocytes to sites of antigen entry, is also regulated by the SNS. A rapid and transient leukocytosis is induced by physical and mental stressors, increasing NK cells, lymphocytes, and monocytes in the blood. These effects can be mimicked by EPI or isoproterenol treatment and blocked by β-AR antagonist administration [174,270]. The SNS via increased circulating catecholamines promote the homing of lymphocytes to secondary lymphoid organs [237,271,272], an important function since antigen presentation and clonal expansion occurs at these sites. For example, In vitro incubation with isoproterenol increases the numbers of injected murine lymphocytes that home to the spleen and lymph nodes [272]. Consistently, chemical sympathectomy alters the migration of adoptively transferred lymphocytes to Peyer’s patches, mesenteric and inguinal lymph nodes. Catecholamines also regulate the egress of lymphocytes from the spleen. Infusion of catecholamines into the spleen increases leukocyte output by either β- or α-AR mediated mechanisms in the absence of changes in blood flow [273]. Besedovsky and colleagues [274] have demonstrated that sympathetic innervation of the spleen alters flow resistance to affect leukocyte output from the spleen. Collectively these studies support a role for the SNS in regulating the cell entry, retention, and release from lymphoid organs. Understanding the mechanisms through which the SNS regulates lymphocyte trafficking may provide a means of enhancing or preventing cell mobilization during local immune and inflammatory responses.

7. Relevance of Sympathetic-Immune Interactions for Disease

7.1. SNS and infections

The adaptive response that develops after host invasion is governed by the predominant Th cell population, Th1 versus Th2, and will determine the outcome of infectious diseases [275277]. Catecholamines suppress cellular and enhance humoral immunity by inhibiting type 1 and augmenting type 2 cytokine production. This Th1/Th2 balance is regulated in healthy rodents, through the increase in SNS activity following an immune challenge [reviewed in 278]. In individuals whose immune systems are compromised and have heightened SNS activity, the SNS may profoundly affect the susceptibility of the organism to infection and/or influence disease course. Additionally, early life experiences and/or previous stressful life events that developmentally program the SNS may alter an individual’s resiliency against infectious disease. There is abundant literature linking psychosocial factors with autoimmunity, psychosomatic illnesses and infectious diseases. Tuberculosis susceptibility increases and disease prognosis is worse in individuals who experienced stressful life events [discussed in 279]. Since cellular immunity is essential for host defense against mycobacterial infections, particularly IL-12 and its ability to induce IFN-γ secretion [280], SNS inhibition of Th1 cell-dependent immune may explain, at least in part these observations [281,282]. Psychosocial stress and/or elevated inflammatory mediators induce abnormalities in sympathetic activity with H. pylori infection [283], the bacteria associated with chronic gastritis that leads to peptic ulcer development. These events skew the immune response towards a Th2 profile and promote the onset and/or progression of the H. pylori infection [discussed in 284,285,286].

In HIV infection, sympathetic innervation of lymphoid tissue is particularly relevant as the lymphoid organs are the primary site of HIV pathogenesis. HIV1 disease progression correlates with a Th2 shift, reduced IL-12 and enhanced IL-10 production [discussed in 287], which promotes the HIV infection and accelerates HIV-1 replication by up to 11-fold in acutely infected human PMBC [288]. This effect on viral replication is mediated via the β-AR-cAMP-PKA signaling cascade. A synthetic, immunosuppressive, retroviral envelope peptide uses the same mechanism, i.e. induction of intracellular cAMP, to shift the cytokine balance toward a Th2 profile [287,289,290]. Infection with the LP-BM5 mixture, a cocktail of murine retro viruses used in a mouse model for acquired immunodeficiency syndrome (MAIDS), depletes splenic NE concentration as the viruses destroy the sympathetic innervation within two weeks [291,292]. Treatment with an NE reuptake blocker, desipramine does not prevent LP-BM5-induced NE depletion, suggesting that splenic nerve loss is not caused by excess release and reuptake of NE. This phenomenon is consistent with the autonomic and peripheral neuropathies associated with acquired immunodeficiency syndrome (AIDS).

7.2. Sympathetic activation and endotoxic shock: inflammatory and anti-inflammatory effects

The suppression of Th1 cytokine production and cell-mediated defense mechanisms may increase susceptibility to infection. During established sepsis, however, a state of immune hyperactivity occurs that is characterized by the excessive production of multiple inflammatory cytokines and is associated with high mortality that can follow a hyporeactive immune system. While the production of the proinflammatory cytokines TNF-α and IL-1 is critically important to fight gram-negative bacterial infections, when these cytokines are produced in excessive amounts, like in sepsis, they trigger a cascade of events that lead to the production of various inflammatory mediators that can cause septic shock and multiple organ dysfunction syndrome. In endotoxic shock, the excess TNF-α produced enhances nitric oxide (NO) production that promotes hypotension, peripheral vasodilatation, and vascular hyporeactivity to vasoconstrictor agents. Surprisingly, treatment with anti-TNF-α agents, IL-1 receptor antagonists, inhibitors of NO or other agents that target inflammatory mediators have been disappointing and have not improved survival [293,294].

Sustained SNS activation during sepsis [295297], including increased gut-derived NE release [298] may have either beneficial or detrimental consequences. Ablation of the SNS prior to gram-negative bacteria challenge, such as Listeria monocytogenes, Pseudomonas aeruginosa or Escherichia coli, reduces bacterial dissemination [299302]. This SNS effect is mediated via increased secretion of peritoneal TNF, phagocytic activity, and influx of monocytes into the peritoneal cavity [302]. Sympathetic activation by experimental stress elicits a similar increase in gram-negative bacterial tissue burden [303]. In contrast, sympathectomized mice given an injection of gram-positive Staphylococcus aureus have increased bacterial burden, an effect mediated by reduced HPA activity, lower IL-4 production by peritoneal cells, and decreased influx of lymphocytes into the peritoneal cavity [302]. To this end, the SNS may differentially affect the influx of inflammatory cells into bacterially infected sites by regulating adherence of bacteria to the gastrointestinal epithelium [304], chemokine production [305], and/or expression of cellular adhesion molecules [306]. The SNS inhibits CC-chemokine ligand 3 or CCL3 production by subsets of neutrophils, which increased the susceptibility of thermally-injured mice to sepsis [305]. This dual role of the SNS in sepsis appears to depend on the innate immune effector mechanisms necessary to eliminate the infectious agent. Thus, adrenergic agents used extensively in intensive-care units to stabilize blood pressure during sepsis, may either support or inhibit bacterial clearance. In order to optimize treatment of patients with sepsis it will be important to understand how the SNS, as well as the HPA axis, regulates bacterial clearance. Comprehending these important neural-immune-bacterial interactions should lead to better manipulation of stress hormones during sepsis.

Clearly targeting specific subpopulation of AR offers a unique opportunity for pharmacologic manipulation of cytokine production during septic shock, given the anti-inflammatory effects of α2-AR antagonists and β2-AR agonists, either alone or in combination. These classes of adrenergic drugs regulate Th1 and Th2 cytokine production, particularly inhibiting TNF-α and IL-12, and augmenting IL-10 production. Pretreatment of healthy human volunteers with EPI prior to endotoxin challenge in vivo inhibits circulating TNF-α levels and increases IL-10 release [307]. Furthermore, isoproterenol suppresses NO production by macrophages and prevents the LPS-induced suppression of vascular contractility to NE in the thoracic aorta ex vivo [308,309]. These findings confirm studies in LPS-treated mice showing: (1) β-AR blockade pretreatment increases; (2) β-AR agonist pretreatment inhibits; and (3) α2-AR blockade with the highly selective α2-antagonist, CH 38083 reduces plasma TNF-α. Pretreatment with chlorisondamine (a sympathetic ganglionic blocker) prevents the CH 38083-induced effect, suggesting that the α2-AR effects are mediated presynaptically [reviewed by 310]. Further, the CH 38083-induced reduction in TNF-α is blocked with addition of the nonselective β-blocker, propranolol, and the β1-selective antagonist, atenolol, but not the β2-selective antagonist, ICI 118,551, suggesting the effect of the α2-antagonist is mediated by β1-AR [310]. Together, these data suggest a dual role for SNS activation after LPS administration: (1) reduction of TNF-α through β-AR, and (2) inhibition of NE release through presynaptic α2-AR. Other investigators have demonstrated in vitro that macrophages participate directly in the β-AR-mediated inhibition of TNF-α through the modulation of IL-10 [257,311314].

In endotoxemic mice, IL-10-induces suppression of TNF-α production and down-regulates an inflammatory response, which reduces their mortality [315]. However, the anti-inflammatory effects of β-AR stimulation are not dependent on IL-10 [312]. In contrast, IL-10 regulation by α-AR stimulation may indeed influence the inflammatory response. Isoproterenol and EPI increases LPS-induced IL-10 production by macrophages in vitro and β-blockade reduces LPS-induced plasma IL-10 in mice, in vivo [307,316]. IL-10 is also an indicator of the Th2 or humoral immune response, as IL-10 drives the β-AR-induced shift towards a Th2 response after LPS treatment. Further, β-AR stimulation inhibits the production of the Th1 cytokines IL-12 and IFN-γ after immunization with LPS [312], or in activated monocyte and dendritic cell cultures [317]. The inhibition of IL-12 and IFN-γ in culture required much lower concentrations of agonist than is required to inhibit TNF-α production. These data suggest that in addition to the potential benefit of adrenergic therapy in sepsis, therapeutically manipulating catecholamine regulation of cytokine production in clinical diseases that exhibit Th1/Th2 predominance may prove beneficial.

In contrast to the inhibitory effects of β2-AR stimulation, in vitro studies demonstrate α2-AR activation stimulates TNF-α production by macrophages [175,257], however, reports of in vivo proinflammatory effects of α2-AR stimulation are limited. Fessler et al. [318] have reported increased mortality in LPS treated rats that are previously given UK 14304 (α2-agonist); however, plasma TNF-α levels are not measured in these animals. In contrast, pretreating with an α1-antagonist, rauwolscine, protects all rats from an LD100 dose of LPS [318]. Blocking α2-AR reduces circulating TNF-α, liver enzymes, bilirubin, prevents bowel hemorrhage and increases arterial pressure, suggesting that many tissues are targets for α2-AR stimulation.

Pharmacological cardiovascular support human septic shock includes the administration of α-AR agonists to maintain perfusion pressure, β-AR agonists to improve cardiac output and, thereby, oxygen delivery; and dopamine-receptor agonists to augment renal and mesenteric perfusion [for more details see 293]. Despite early claims of improved outcome, more recent studies regarding the use of dobutamine suggest poorer survival and increased end-organ injury [319]. Moreover, in humans, it is the β2- not β1-ARs that appear to control monocyte/macrophage cytokine profiles. Therefore, currently, a clear recommendation for a specific catecholamine regimen in septic shock is impossible. Systemic examination will be necessary to determine the correct combination of adrenergic agents to acquire the optimal balance between cardiovascular support and immunomodulation during septic shock.

Hensler et al. [320] have reported low preoperative macrophage IL-12 production precedes the onset of sepsis, which underscores the associative data that immunosuppression predisposes patients undergoing major surgery or trauma to sepsis. Additionally, catecholamines can dramatically increase the growth of gram-negative bacteria such as Escherichia coli and Yersinia enterocolitica in vitro [321]. Thus, an early intervention with adrenergic drugs that suppresses IL-12 production and/or possibly increases bacterial growth may be harmful.

Anti-TNF-α strategies demonstrate that patients with high systemic levels of IL-6 might benefit most from anti-cytokine therapy [294]. Therefore, the benefits of immune therapy would be optimized based on the individual inflammatory response condition. Thus, it is proposed that immune stimulatory protocols should prove beneficial for patients showing hyporeactive immune response conditions, whereas patients with a hyperactive immune system may selectively benefit from anti-inflammatory therapies [320]. In conclusion, patient stratification, timing issues, and the type of intervention, including adrenergic agents, are important factors in successful therapeutic strategies in sepsis and its complications. The role for targeted AR interventions and the temporal relationship between endotoxin administration and catecholamine-induced alterations in cytokine production in vivo may lead to clinical studies that maximize the benefits and minimize unwanted side effects of stimulating AR in the treatment of sepsis.

7.3. Major injury

Suppression of Th1 cytokines and potentiation of Th2 cytokine production impair lymphocyte proliferation, reduce delayed-type hypersensitivity skin test response, lower cell surface MHC class II antigen expression, and suppress neutrophil functions (chemotaxis, phagocytosis and oxygen radical production) correlate with trauma and major surgery. Serious traumatic injury, major burns or major surgical procedures often lead to severe immunosuppression, which contributes to infectious complications, the most common cause of late death after trauma. Activation of the neuroendocrine pathways is an essential component of the adaptive process to trauma, brain injury, and major surgery. After extracerebral brain injury there is a biphasic response, with an early increase in sympathetic outflow followed by an altered stimulation of the HPA axis during the ebb phase, the first phase is followed by a decrease in both responses [322]. The intensity of these changes (particularly with catecholamines and cortisol plasma levels) correlates with the severity of both cerebral and extracerebral injury and is a predictor of an unfavorable prognosis [322325]. Cellular immunity is suppressed in patients with traumatic major injury and in animal models of burn injury, as there is a reduction in production of IFN-γ and IL-12 and increase in IL-10, i.e., a Th2 shift [326]. Further, TNF-α production is reduced in LPS-stimulated whole blood after trauma [327], whereas the production of type 1 cytokines, such as IFN-γ, TNF-α, and IL-2, are down-regulated on postoperative day 1 following conventional but not laparoscopic surgery [328]. SNS activation may drive the increase in systemic IL-10 indicated in the immunosuppression after injury [329]. Thus, high levels of systemic IL-10 documented in patients experiencing a “sympathetic storm” due to acute accidental or iatrogenic brain trauma correlated with the high incidence of infection. β-AR blockade by propranolol dose dependently inhibited catecholamine-induced release of IL-10 and prevented the increase of circulating IL-10 in the rat model [329]. Similarly, in a mouse model of focal cerebral ischemia, blocking the SNS but not the HPA axis prevented the defective IFN-γ response and the life-threatening spontaneous systemic bacterial infections in ischemic mice [330]. These data suggest that catecholamine secretion triggered by major injury may contribute to the severe immunosuppression observed in these patients. These observations may have two important implications: 1) they provide direct evidence that the sympathoadrenal system coupled with a systemic immunosuppressive response triggers a high incidence of infections and complications; and 2) they emphasize the importance of neurotransmitter/hormone-associated modulation of immunity [331].

7.4. Sympathetic regulation of autoimmune diseases

Inflammatory autoimmune diseases have dysregulated Th1 versus Th2 immune responses and altered IL-12/TNF-α versus IL-10 balance. An excess of IL-12 and TNF-α production is seen in rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes mellitus, autoimmune thyroid disease (ATD) and Crohn’s disease resulting in the cytokine balance being skewed toward Th1 cell-mediated immune response and as a result the Th2 activity and the production of IL-10 are deficient [277,332334]. This is a critical factor determining the proliferation and differentiation of Th1-related autoreactive cellular immune responses in these disorders [335]. In contrast, systemic lupus erythematosus (SLE) is associated with excessive production of IL-10 and a deficiency in IL-12 and TNF-α production, which skews cytokine production toward a Th2 profile [336,337].

The influence of SNS in autoimmune disorders is complex [332,338340]. Considering the Th2-driving effects of catecholamines via β2-AR receptors under normal conditions, a hypoactive SNS may facilitate or sustain the Th1 shift in MS or RA, whereas SNS hyperactivity may intensify the Th2 shift and induce or facilitate flares of SLE. This hypothesis is supported by several animal studies and certain clinical observations. F344 rats, which have a hyperactive stress system, are extremely resistant to experimental induction of Th1-mediated autoimmune states, including arthritis, uveitis, and experimental allergic encephalomyelitis (EAE) [332]. Similarly, remission of Th1 type-mediated autoimmune diseases, such as RA, MS, type 1 diabetes mellitus, and ATD, possibly via the increased cortisol and probably catecholamine levels may suppress proinflammatory and potentiate anti-inflammatory cytokine production in women in the third trimester of pregnancy [341,342]. In contrast, increased SNS activity seems to drive the higher Th1 vs. Th2 cytokine profiles in the Lewis rat model for RA. Treatment with a β-AR agonist or an α-AR antagonist exacerbates disease pathology. However, combined treatment with these compounds appears to balance Th1/Th2 drive and reduce disease severity [334], suggesting that the role of the SNS is more complex, involving both promotional and inhibitory mechanisms under conditions where chronic inflammation plays a major role in disease pathology. Through a reciprocal mechanism, Th2-mediated autoimmune disorders, such as SLE, during stress or pregnancy, may flare under conditions of high cortisol and catecholamines to drive IL-10 production [332,333]. There are different opinions [343345] regarding dysregulation between the nonspecific and specific immune responses in pregnancy. However, Lewis rats, which exhibit dysregulated stress systems, are extremely prone to develop experimentally-induced Th1-mediated states, such as arthritis, uveitis, or EAE [332]. Similarly, patients with Th1 type-mediated autoimune diseases such as RA, MS, and ATD have heightened, blunted or dysregulated stress activity. This might include the postpartum period, the period that follows cessation of chronic stress or a rebound effect upon relief from stressors [281,332,339].

Several lines of evidence suggest that in MS and in EAE, the experimental model for MS, the sympathetic-immune interface is defective. In progressive MS, sympathetic skin responses decrease and lymphocyte β-AR expression increase [346]. While the density of β-AR on monocytes, B cells, and CD41 cells remain unchanged, receptor numbers on CD8+ T cells increase 2- to 3-fold, compared with age-matched controls [338,346,347]. Increased β-AR may result from a compensatory response to reduced concentration of NE in secondary lymphoid tissues in EAE [348], and suggests an adaptive response to lower NE availability in the spleen observed in MS [338]. An “up-regulating” effect of cortisol or IL-1 on β-AR receptor expression may further contribute to this effect [349].

The “protective” role of the SNS in EAE pathogenesis is further supported by observations that chemical sympathectomy augments disease severity [229], even in the presence of elevated circulating glucocorticoids. Splenic NE content increases at peak disease an effect that is more difficult to interpret [350], but may suggest reduced NE release which would further support protection by the SNS in EAE. NE turnover studies are needed to confirm this hypothesis. Additionally, isoproterenol or terbutaline, β-AR and β2-AR agonists, respectively, or prazosin, an α1-AR blocker are reported to suppress chronic/relapsing EAE in Lewis rats [351,352]. These drug actions may result from receptor-mediated changes in production of type 1 cytokines. This is further substantiated by recent evidence that the drug rolipram, a selective inhibitor of phosphodiesterase type IV and consequently the production of type 1/proinflammatory cytokines, such as TNF-α, IFN-γ, and IL-12, ameliorates EAE.

The role of SNS in RA is less clear as the SNS appears to play both a permissive and inhibitory role in the disease pathogenesis. Clinically, there are multiple reports of autonomic dysregulation in both juvenile chronic and RA, such as increased resting heart rates and elevated levels of urinary NE metabolites [353] and augmented SNS reflex responses to orthostatic stress [354], the valsalva maneuver [354], deep breathing [355], pupil size [355] and perspiration [356]. These reports suggest increased sympathetic tone. If the sympathetic-to-immune modulatory effects were solely mediated via β-AR stimulation, heightened SNS tone should afford protection by shifting the immune response toward humoral immunity. However, denervation studies in animal models for RA report mixed results depending on whether sympathetic denervation is local or systemic. Systemic depletion of NE using guanethidine, reserpine, or 6-OHDA treatment in rodent models of RA attenuates inflammation and joint destruction [357362]. Additionally, administration of propranolol, a β-AR antagonist, or regional sympathetic blockade with guanethidine reduces symptoms associated with RA in clinical studies [359]. Unlike other Th1-driven autoimmune diseases, these studies suggest a promiscuous role for SNS in disease pathology.

Based on findings from our laboratory using an animal model for RA, the reasons for this difference in sympathetic-immune modulatory effects in RA are 4-fold. First, it is important to realize that the SNS regulates immune events that take place in secondary lymphoid organs as well as immune responses that occur locally in the inflamed joint. In the former site, sympathetic regulates Th1 vs. Th2 drive through clonal expansion of autoreactive immune cell, and affects immune cell migration and blood flow through secondary lymphoid organs. In the latter site, the SNS regulates the movement of antigen-presenting cells such as dendritic cells from the inflamed site into draining lymph nodes, and the localized inflammatory response including blood flow, vascular permeability, influx of inflammatory cells, and release of inflammatory mediators. Additionally, sympathetic nerves in the inflamed joint interact with substance P-containing nerves that convey pain and modulate local immune responses. Levine et al. [363] using an adjuvant-induced arthritis (AA) model in Sprague-Dawley rats has shown that chronic administration of the β-AR blocker, propranolol, attenuates the severity of joint injury. In contrast, selective denervation of the lymph nodes that drain the hind limbs (the popliteal and inguinal) prior to immunization to induce experimental arthritis exacerbates the disease [364]. This latter finding is consistent with a protective role of the SNS, likely afforded by shifting the immune response toward a humoral response based on circulating and immune organ cytokine profiles [334,365] or by preventing the autoreactive immune cell populations from homing to the joint.

A second factor important in understanding SNS regulation in RA is the multiple roles of the SNS that occur across the progression of the disease. This is underscored by studies in animal models for RA where the timing of sympathetic manipulation, either via pharmacological agents or denervation of sympathetic nerves, relative to the arthrogenic antigen challenge differs. Continuous infusion of EPI or salbutamol (β2-AR agonists) beginning 2 days before and continuing through day 28 after immunization with adjuvant increases the severity of AA, respectively [366]. Consistent with these findings, continuous infusion of propranolol and ICI118 551 (β-AR or β2-AR antagonists, respectively) over the same time frame relative to challenge ameliorates disease severity. These data indicate that heightened SNS activity prior to exposure to the arthrogenic antigen and continuing through disease progression is detrimental. Conversely, treatment with β2-AR agonists, like salbutamol, beginning at disease onset and continuing every other day for 10 days potently suppresses disease processes to prevent joint damage in established collagen-induced arthritis [367]. Malfait et al. [367] attribute these differences in disease outcome to the way the drug salbutamol was administered, continuous infusion rather than individual injections and to differences between the animal models used in these studies. However, studies from our laboratories suggest that differences in outcome may be due to the timing of adrenergic drug treatment relative to the adjuvant challenge and the disease course. Treatment of AA rats with terbutaline, a β2-AR agonist, started at the time of adjuvant injection to induce disease, exacerbated disease severity; however, a directionally opposite effect of terbutaline treatment occurs when treatment begins at the time of disease onset significantly reduced disease severity [365]. Collectively, these findings suggest that at different time frames across the development and progression of AA, the SNS may differentially modulates afferent phase of the immune response and chronic inflammation once the disease has been established.

Changes in AR expression with development and progression of AA are the third factor that may contribute to controversies in the reported adrenergic drug effects on severity of inflammatory arthritis. The surprisingly small effects of sympathectomy and β-AR drug treatments on disease severity that have been previously reported [362,364] may be explained by altered AR expression and/or signaling capacity with disease progression. β2-AR numbers and functional responses to agonists in peripheral blood mononuclear cells (PBMC) are lower in RA patients [368370]. The density of β-AR is lower in synovial fluid lymphocytes and CD8+, but not CD4+, lymphocytes in RA patients [370] compared with cells from healthy donors. Altered β-AR expression may explain diminished catecholamines-stimulated OKT3 (T cell mitogen)-induced PBMC activation. These findings suggest impaired SNS regulation of cellular proliferation, effects that are expected to contribute to the pathogenesis of RA. Reduced PBMC expression and activity of G-protein receptor-coupled kinase, important for regulating β2-AR cell surface expression, may also influence disease progression in RA patients [371]. Defective β2-AR signaling is also seen in PBMC from children with active systemic and polyarticular juvenile RA as the disease develops, reflected by impaired β2-AR agonist-stimulated cAMP production [372]. Collectively, these studies indicate dysregulated sympathetic-immune signaling in immune target cells in RA.

Concomitant with altered functional β2-AR, α1-AR are induced abnormally on peripheral blood monocytes in juvenile RA patients [373]. Stimulation of these α1-ARs increases the production of IL-6 [373], a proinflammatory cytokine. Similarly, AA rats treated with an α-AR antagonist reduces the production of proinflammatory cytokines [374]. Increased sympathetic activation (and/or circulating cortisol) may induce monocyte α1-AR expression, since treatment of monocytes in vitro with a β2-AR agonist or dexamethasone can induce monocyte α1-AR expression [375]. Disease-induced AR subtype switching and impaired β2-AR signaling capacity sensitivity to catecholamines likely drives the inflammatory response with consequent disease exacerbation, since reduced β-AR and increased α-AR stimulation would be disinhibitory and excitatory, respectively [314,317,257,307,376378]. For example, β2-AR stimulation inhibits TNF-α and IL-12 and increases IL-10 production, so impaired receptor function promotes TNF-α production. Similarly, α1- and α2-AR stimulation increases TNF-α production.

While the role of α-AR in autoimmunity is not entirely clear, intriguing evidence exists for this class of receptors to plays a profound role in models of RA and MS. The effects of treatment with drugs that interact with α-AR on disease pathology are dependent on time of treatment relative to disease course [365]. Treating rats with α- or α2-antagonists from adjuvant challenge through severe disease increases disease severity [363, 365,366]. However, it α-, α1- or α2-AR antagonist treatment begins at disease onset, disease pathology is significantly ameliorated ([365, 366] Lorton and Lubahn, unpublished observations). Based on these data, both α1 and α2-AR mediate sympathetic influences on disease outcome. Mechanistically, α-AR antagonists may alter antigen processing and apoptosis within the secondary lymphoid organs during initiation of the disease via blocked interaction of NE with α-AR on sympathetic nerve terminals and/or on cells of the immune system. α2-AR antagonists acting through presynaptic α2-AR, would reduce NE release from sympathetic nerves and lower NE available for binding to β2-AR expressed by immune target cells. As mentioned above during the effector phase, inhibiting α-AR signaling inhibits inflammatory cytokine production in arthritic joints. Additionally, α1- and α2-antagonists could impact disease pathology by targeting the smooth muscle and endothelial cells in the vasculature by affecting immune cell migration from secondary lymphoid organs, homing of immune cells into the arthritic joints, plasma extravasation and edema. Further studies are needed to elucidate the mechanisms of α1- and α2-AR antagonists on the inflammatory and disease processes of RA.

Interestingly, a study from our laboratory reveals a striking reorganization of NA nerves and immune cell populations in secondary lymphoid organs as AA develops [379; reviewed in 365]. NA innervation of splenic white pulp (site of antigen processing and clonal expansion of T cells) is reduced, particularly in white pulps most distal from the point of nerve entry into the spleen and suggestive a peripheral neuropathy. In contrast near regions where the nerves enter the spleen splenic white pulps are hyperinnervated by sympathetic nerves. Taken together, these findings suggest an injury-sprouting response, with a dying back of sympathetic nerves in regions distant to the nerve entry regions as the disease progresses and a subsequent attempt to replace, reorganize, and repair lost innervation with a sprouting response at nerve entry points. Additionally, we report an increase in the sympathetic innervation of the red pulp, a region that normally is sparsely supplied with NA nerves and where activated immunocytes reside in AA rats with severe disease. The red pulp also is a site of immune cell exiting from the spleen. Reduced sympathetic innervation and NE concentrations in the spleen also precedes the onset of other autoimmune diseases in other animal models, including the New Zealand mouse strains as models for hemolytic anemia and lupus-like syndrome [114,380], and EAE [348]. However, the growth of sprouting NA nerves into the red pulp with severe disease appears to be unique to the AA model.

Altered nerve distribution and density in spleens of AA rats occurs concomitantly with changes in ED3+ macrophages and CD4+ T cells, which are target cells of sympathetic nerves. The density of these immune cell populations are reduced the white pulp and increased in the red pulp. This disease-mediated effect is further exacerbated by chemical sympathectomy. The density of IgM+ B lymphocytes the splenic follicles [7] throughout the spleens of AA rats also decreases with disease development, but does not appear to be dependent on changes in NA innervation, since chemical sympathectomy did not have an effect of numbers of IgM+ cells seen with disease development. These findings underscore the disease-associated pathology of sympathetic nerves in secondary lymphoid organs. Altered sympathetic nerve density and distribution may reflect changes in the activational states and redistribution of immune target cells as the disease progresses, and may, in turn, alter antigen processing and clonal expansions of future antigenic challenges. Given that NA nerves, through β-AR mechanisms, promote Th2 cell and impede Th1 cell development, a loss of these nerve fibers in the splenic white pulp may support aggressive cell-mediated immune responses. The abnormal NA innervation of the red pulp may facilitate SNS signaling of immune system effector cells residing in this compartment prior to homing to arthritic joints. These signals received in the red pulp may promote joint inflammation once they arrive in the arthritic joints.

Antigen- and hydrogen peroxide-induced arthritic animals [381] and RA patients [reviewed in 382,383] have markedly reduced sympathetic innervation of the synovium in affected joints. Additionally, bradykinin, a potent mediator of inflammation, stimulates the release of transmitters (specifically, PGE2) from these nerves, which promotes plasma extravasation and thus inflammation [384]. Bradykinin-induced neurotransmitter release is not dependent on increased activity of NA neurons or release of NE from these nerve terminals [385]. From these findings it appears that NA nerves can have two distinct actions: classical transmission regulated by impulse activity to release NE and locally-induced release of non-adrenergic transmitters, both of which modulate inflammatory processes [384]. In summary, the SNS’s ability to modulate immune functions is compromised in this disease. The complexities of adrenergic drug effects on severity of inflammatory arthritis appear to result from altered AR expression with disease progression, differential outcomes from sympathetic regulation of different target organs, both in lymphoid organs and affected joints, the relationship between onset of drug delivery and timing of drug treatments relative to the arthritis inducing antigen challenge, and the reorganization of sympathetic nerves and immune cell populations in SNS target organs.

7.5. SNS and Tumor growth

Based on both clinical and animal research, inhibited tumor growth and better patient prognosis is positively associated with good social support. While the mechanisms for this association are not clear, we do know that interactions between the nervous, neuroendocrine, and immune system are important [386,387], and that positive social interactions positively influences these systems to buffer against tumor growth. Psychosocial factors alter the activity or stress-reactive areas of the brain to affect tumor immunity, which in turn significantly affects the progression of certain types of cancer [reviewed in 388,389]. These anti-cancer effects are mediated through innate and acquired immune-directed killing mechanisms. NK cells of innate immunity targets tumor cells that express low levels of MHC I [390398]. In metastatic breast cancer, in vitro NK cell activity is a good predictor of the rate of disease progression [399]. Anti-tumor defense is also provided by the functions of macrophages and granulocytes [400]. Specific killing of tumor cell is directed by CD4+ T cells or antibody-complement-mediated immune mechanisms [401,402].

Evidence for psychosocial effects on tumor immunity has been provided from clinical and animal research. Higher stress levels predict lower NK cytotoxicity, mitogen- induced T cell and IFN-γ stimulation responses [403410]. Negative psychosocial factors and chronic stress interact to negatively affect tumor immunity, whereas psychosocial interventions for stress reduction and improved coping strategies positively impact immunity [407,411,412; reviewed in 413]. In contrast, acute psychological stress increased circulating NK cell numbers and/or NK cell activity [414416], but this effect may be followed by reduction below baseline shortly after stress [416]. Interpersonal difficulties, depression, anxiety and chronic stress reduce the number and function of circulating NK cells [417423]; effects associated with elevated cortisol levels [424]. Positive supportive relationships mitigate the stress-induced immunosuppression [418], such as chronic stress-induced reduction in NK cell number and functions in cancer patients [425]. Negative emotions are linked to lower lymphocytic infiltration into the tumor site, increased tumor growth, and poorer disease outcome [426]. In studies with rats inoculated with a mammary carcinoma stress-associated increases in tumor growth occur concomitantly with suppressed NK cell activity and reduced survival time [427,428].

Psychosocial factors alter SNS activity to affect tumor immunity and disease outcome. Catecholamines, the major neurotransmitter released by the SNS, modulate immune functions important for tumor immunity, including Th and B cell functions, NK cell trafficking and activity and T suppressor cell cytotoxicity [429434]. In a model where mammary tumors are subcutaneously injected into a sympathetically denervated ear, tumor growth is attenuated approximately 50% over that observed in the contralateral, sympathetically intact ear [435]. Evidence of increased lymphocyte metabolism in draining lymph nodes supports immunological mechanisms as being responsible, at least in part, for this result. In this study, sympathectomy had no effect on tumor metastases. In another animal study, treatment with deprenyl, a monoamine oxidase B inhibitor, reduces age-related spontaneous mammary and pituitary tumor growth [438]. Lower spontaneous tumor growth by deprenyl treatment is linked to partially restoration of the age-related decline in splenic NA innervation and improvement in immune function in old rats [436437]. Not all studies, however, report attenuated tumor growth under conditions of reduced sympathetic activity. Brenner et al. [439] have reported an increase in the number of pulmonary metastases in mice chemically sympathectomized before injection of alveolar carcinoma cells, an effect that is independent of changes in NK cell activity. No effects of sympathectomy occur when mice are denervated after injection of cancer cells. In vivo priming of the immune system with irradiated carcinoma cells abrogates tumor cell metastases, but, NA denervation before priming has no effect on T cell-mediated attenuation in metastases. These findings suggest NA innervation can decrease tumor metastases through non-immunological mechanisms. They also emphasize a complex modulatory role for the SNS in cancer cell migration and proliferation. From these studies it is clear that a number of factors are important in determine effects of sympathetic activity on tumor immunity, including timing of manipulations relative to tumor inoculation, the tumor type, and the species/strains and the age of the animals used for study.

It is becoming increasingly clear that the SNS, along with the hypothalamic-pituitary adrenal axis, mediate the effects of psychological stress on breast cancer progression. Studies by Ben-Eliyahu and coworkers report that sympathetic ganglionic blockade, adrenal demedullation, or administration of a non-selective β-blocker either ameliorated or attenuated swim stress-induced increases in MADB106 tumor metastases compared with non-stressed animals [440]. Similarly, the tumor enhancing effects of social confrontation and hypothermic stress are lowered by adrenal demedullation or β-AR blockade [441]. These studies indicate that adrenal catecholamines are responsible for some of the stress-induced increases in MADB106 metastasis, via a β-AR-mediated mechanism. The effects of acute stress on MADB106 metastasis are mimicked be treatment with β-AR agonists, blocked by nadolol, and not observed in NK-depleted rats [442], suggesting that catecholamines interact with β-AR on NK cells to exert SNS effects on tumor metastases. Conversely, stress-induced effects on NK cell activity and tumor metastasis are not directly mediated via the HPA axis, since corticosterone administration (1, 3, or 9 mg/kg, sc) does not alter tumor metastasis [reviewed in 443].

The SNS mediates the effects of housing stress and social isolation on tumor development for several types of cancer in mice [444]. In syngeneic C57Bl/6 and BALB/c mice, social isolation reduces host resistance to B16 melanoma and Meth A fibrosarcoma growth, respectively. Oral administration of propranolol, a non-selective β-adrenergic antagonist, but not chronic treatment with corticosterone, completely abrogates the effect of social isolation on tumor growth. These data support the SNS as a modulator of the effects of social isolation stress on the progression of several types of cancer.

Of clinical relevance, prostate cancer patients receive selective α1-AR antagonists to reduce the discomfort associated with urethral blockage by relaxing smooth muscles in the prostate gland and urethra. Several studies in mice suggest that treatment with quinazoline-derived α1-AR antagonists, doxazosin and terazosin, which are long-lasting drugs used to provide acute relief of the obstructive symptoms, also reduce tumor growth [445]. The in vivo mechanism(s) is(are) not clear, however, culturing prostate cells with α1-AR antagonist reduces cellular proliferation and increases the rate of apoptosis [445,446] via an α1-AR independent mechanism [447]. Sympathetic activation may also regulate prostate cancer progression via β-AR-mediated mechanisms. In rat prostate cells, β-AR agonists stimulate adenylyl cyclase and raise cAMP levels [448,449], and in human prostate cancer cell lines elevated cAMP production drives their differentiation toward a neuroendocrine phenotype [450,451], an effect reversed by removing the cAMP-generating agents [451]. These findings are important because tumor grade, loss of androgen sensitivity, autocrine and paracrine activity, and poor prognosis all correlate with appearance of this neuroendocrine phenotype in human prostate cancer cells. Therefore, adrenergic regulation via β-AR on prostate tumor cells by the SNS could play an important role in advanced prostate cancer.

8. Summary and Concluding Remarks

The SNS innervates all primary and secondary immune organs (bone marrow, thymus, spleen, lymph nodes and gut-associated lymphoid tissue) and release catecholamines to modulate immune functions. Further, some immune cells synthesize, store and release catecholamines that can act as autocrine and/or paracrine factors to regulate immune function. The literature summarized and discussed above overwhelmingly indicates that even small effects of catecholamines on immune responses can have significant effects on health outcomes. The mechanisms for neuroimmune modulation via the SNS are not well delineated. Further research is necessary to expand our knowledge of the role the SNS in modulating cellular- and humoral-mediated immune responses to advance the development of therapeutic interventions.

Sympathetic innervation of lymphoid tissues meets the criteria for neurotransmission with immune cells as target cells. These criteria include demonstrating the presence of NA nerve fibers in lymphoid organs, the release of NE from the NA nerve terminals in these organs, and the expression of AR on lymphoid cells, which respond functionally to stimulation. Electron microscopic studies reveal direct contacts between NA nerve terminals and lymphocytes and macrophages in the PALS and marginal zone of the spleen. Whether direct contacts function in neurotransmission is not clear. As in many peripheral sympathetic target organs, NE released from NA terminals diffuses away from the terminal site to interact with AR expressed on adjacent and distant target cells; this is called paracrine release. The release of NE is modulated presynaptically by negative feedback of NE acting on α2A/C-AR. Thus, the SNS is a “hardwired” integrative and regulatory pathway between the CNS and the immune system. Sympathetic-immune interactions are complex. The spleen and lymph nodes are the sites of interest for the majority of studies examining sympathetic modulation of immune functions in basic research using animals. The SNS’s role in regulating immune functions in primary lymphoid organ and mucosal lymphoid tissues is a neglected area if research thus far. There is enough data to support a functional role of the SNS in regulating the functions of primary lymphoid organs (bone marrow and the thymus) and mucosal-associated lymphoid tissue.

Catecholamines released by increased sympathetic activation “fine tune” immune responses. The release of co-localized transmitters or modulators, such as NPY and ATP, also bind to selective receptors on immune target cells, and can modify NA signaling [452]. The influence of co-released neurotransmitters on immune response also is poorly understood. The effects of catecholamines allow for quick adjustments of immune responsiveness (within minutes). Peripheral catecholamines also may tonically inhibit certain immune functions (i.e., type 1 proinflammatory cytokine production). One important effect of catecholamine release from sympathetic nerves is to opposing effects on cellular and humoral immunity. Stimulation of β-AR on Th1 inhibits the production of Th1 type cytokines. The production of cytokines by Th2 cells, which do not appear to express β-AR, is potentiated by reduced production of Th1-type cytokines. Additionally, catecholamines can affect other effector cells to suppress cellular and boost humoral immunity. Sympathetically-driven humoral responses can be either beneficial or detrimental to disease progress depending on the pathological condition. For example, heightened sympathetic activation of an autoimmune disease mediated by a Th2 bias would predict a poorer outcome. Under normal conditions, the above-mentioned Th2-driving force of catecholamines, in concert with the Th2-driving effect of glucocorticoids, serves as an important feedback mechanism to reduce excessive Th1 proinflammatory cytokine production and prevent bystander damage after the antigen has been cleared. Catecholamines also regulate the production of pro- and anti-inflammatory cytokines produced by macrophages locally at sites of inflammation, and therefore play a regulatory role in innate immune responses.

An abnormal increase or decrease in SNS activity, defects in normal sympathetic-to-immune signaling, or changes in NA innervation of immune organs and chronically inflamed tissues can contribute to the pathophysiology of immune-mediated diseases, particular under conditions where chronic inflammation and/or a selection of Th1 (type 1) versus Th2 (type 2) responses play a significant role in disease pathogenesis. Examples of such diseases include, but are not limited to, some types of infections, major injury and its complications, autoimmune/inflammatory diseases, allergic (atopic) reactions, and cancer. Specific immune responses regulated by the SNS are influenced by many factors including the nature, clearance and presence or absence of an antigen. In addition, the timing of sympathetic activation relative to the antigen challenge, the presence and expression level of AR subtypes on the immune cells (e.g., β2- versus α2-ARs), the type of G-protein coupled to the β2-AR (e.g., Gs versus Gi) and second messenger system activated and the stage of activation/differentiation of the immune cell. Research directed at gaining a better understanding of the dynamic changes in AR subtype expression on different immune cells, the mechanisms for altered AR coupling to G proteins with consequent effects on intracellular pathways used to mediate the actions of catecholamines, and the conditions necessary for these changes to take place is clearly needed. Better knowledge and understanding of the physiology and pathophysiology of sympathetic-immune interactions will be critical for development of novel therapeutic approaches aimed at ameliorating diseases in which abnormal immune functions contribute to, or drive, the underlying pathophysiology of the disease. Patients with infections that are difficult to clear and certain types of cancer may benefit from β2-AR antagonist or α2-AR agonist interventions, which would boost cell-mediated immune responses; while Th1-mediated autoimmune diseases, such as RA and MS, may benefit from β2-AR agonists and/or α2-AR antagonists treatment that would dampen chronic inflammation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Bjurholm A, Kreicbergs A, Terenius L, Goldstein M, Schultzberg M. Neuropeptide Y-, tyrosine hydroxylase- and vasoactive intestinal polypeptide-immunoreactive nerve in bone and surrounding tissues. J. Auton. Nerv. Syst. 1988;25:119–125. doi: 10.1016/0165-1838(88)90016-1. [DOI] [PubMed] [Google Scholar]
  • 2.Tabarowski Z, Gibson-Berry K, Felten SY. Noradrenergic and peptidergic innervation of the mouse femur bone marrow. Acta Histochem. 1996;98:453–457. doi: 10.1016/S0065-1281(96)80013-4. [DOI] [PubMed] [Google Scholar]
  • 3.Felten DL, Gibson-Berry K, Wu JH. Innervation of bone marrow by tyrosine hydroxylase-immunoreactive nerve fibers and hemopoiesis-modulating activity of a β-adrenergic agonist in mouse. Mol. Biol. Hematopoiesis. 1996;5:627–636. [Google Scholar]
  • 4.Felten DL, Felten SY, Carlson SL, Olschowka JA, Livnat S. Noradrenergic and peptidergic innervation of lymphoid tissue. J. Immunol. 1985;135:755s–765s. [PubMed] [Google Scholar]
  • 5.Bellinger DL, Felten SY, Ackerman KD, Lorton D, Madden KS, Felten DL. Noradrenergic sympathetic innervation of lymphoid organs during development, aging, and in immune disease. In: Amenta F, editor. Aging of the Autonomic Nervous System. Boca Raton: CRC Press; 1992. pp. 243–284. [Google Scholar]
  • 6.Felten SY, Felten DL. The innervation of lymphoid tissue. In: Ader R, Felten DL, Cohen N, editors. Psychoneuroimmunology. 2nd edition. New York: Academic Press; 1991. pp. 27–69. [Google Scholar]
  • 7.Bellinger DL, Lorton D, Lubahn C, Felten DL. Innervation of lymphoid organs – association of nerves with cells of the immune system and their implications in disease. In: Ader R, Felten DL, Cohen N, editors. Psychoneuroimmunology. 3rd edition. New York: Academic Press; 2001. pp. 55–111. [Google Scholar]
  • 8.Yamazaki K, Allen TD. Ultrastructural morphometric study of efferent nerve terminals on murine bone marrow stromal cells, and the recognition of a novel anatomical unit: the “neuro-reticular complex”. Am. J. Anat. 1990;187:261–276. doi: 10.1002/aja.1001870306. [DOI] [PubMed] [Google Scholar]
  • 9.Afan AM, Broome CS, Nicholls SE, Whetton AD, Miyan JA. Bone marrow innervation regulates cellular retention in the murine haemopoietic system. Br. J. Haematol. 1997;98:569–577. doi: 10.1046/j.1365-2141.1997.2733092.x. [DOI] [PubMed] [Google Scholar]
  • 10.Garcia JF, van Dyke DC. Response of rats of various ages to erythropoietin. Proc. Soc. Exp. Biol. Med. 1961;106:585–588. doi: 10.3181/00379727-106-26410. [DOI] [PubMed] [Google Scholar]
  • 11.Maestroni GJ, Cosentino M, Marino F, Togni M, Conti A, Lecchini S, Frigo G. Neural and endogenous catecholamines in the bone marrow. Circadian association of norepinephrine with hematopoiesis? Exp. Hematol. 1998;26:1172–1177. [PubMed] [Google Scholar]
  • 12.Marino F, Cosentino M, Bombelli R, Ferrari M, Maestroni GJ, Conti A, Lecchini S, Frigo G. Measurement of catecholamines in mouse bone marrow by means of HPLC with electrochemical detection. Haematologica. 1997;82:392–394. [PubMed] [Google Scholar]
  • 13.Besedovsky HO, del Rey AE, Sorkin E, Da Prada M, Keller HH. Immunoregulation mediated by the sympathetic nervous system. Cell. Immunol. 1979;48:346–355. doi: 10.1016/0008-8749(79)90129-1. [DOI] [PubMed] [Google Scholar]
  • 14.Madden KS, Sanders VM, Felten DL. Catecholamine influences and sympathetic neural modulation of immune responsiveness. Annu. Rev. Pharmacol. Toxicol. 1995;35:417–448. doi: 10.1146/annurev.pa.35.040195.002221. [DOI] [PubMed] [Google Scholar]
  • 15.Tang Y, Shankar R, Gamelli R, Jones S. Dynamic norepinephrine alterations in bone marrow: evidence of functional innervation. J. Neuroimmunol. 1999;96:182–189. doi: 10.1016/s0165-5728(99)00032-6. [DOI] [PubMed] [Google Scholar]
  • 16.Maestroni GJ, Conti A. Modulation of hematopoiesis via alpha 1-adrenergic receptors on bone marrow cells. Exp. Hematol. 1994;22:313–320. [PubMed] [Google Scholar]
  • 17.Lipski S. Effects of beta-adrenergic stimulation on bone-marrow function in normal and sublethally irradiated mice. I. The effect of isoproterenol on cAMP content in bone-marrow cells in vivo and in vitro. Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med. 1976;29:359–366. doi: 10.1080/09553007614550411. [DOI] [PubMed] [Google Scholar]
  • 18.Lipski S. Effect of beta-adrenergic stimulation by isoproterenol on proliferation and differentiation of mouse bone marrow cells in vivo. Pol. J. Pharmacol. Pharm. 1980;32:281–287. [PubMed] [Google Scholar]
  • 19.Beckman B, Mirand E, Fisher JW. Effects of beta adrenergic agents and prostaglandin E1 on erythroid colony (CFU-E) growth and cyclic AMP formation in Friend erythroleukemic cells. J. Cell. Physiol. 1980;105:355–361. doi: 10.1002/jcp.1041050218. [DOI] [PubMed] [Google Scholar]
  • 20.Byron JW. Evidence for a beta-adrenergic receptor initiating DNA synthesis in haemopoietic stem cells. Exp. Cell. Res. 1972;71:228–232. doi: 10.1016/0014-4827(72)90283-2. [DOI] [PubMed] [Google Scholar]
  • 21.Byron JW, Fox M. Adrenergic receptor blocking agents modifying the radioprotective action of T.A.B. Brit. J. Radiol. 1969;42:400. 1. [PubMed] [Google Scholar]
  • 22.Dresch C, Minc J, Poirier O, Bouvet D. Effect of beta adrenergic agonists on beta blocking agents on hemopoiesis in human bone marrow. Biomedicine. 1981;34:93–98. [PubMed] [Google Scholar]
  • 23.Sudo N, Yu XN, Sogawa H, Kubo C. Restraint stress causes tissue-specific changes in the immune cell distribution. Neuroimmunomodulation. 1997;4:113–119. doi: 10.1159/000097329. [DOI] [PubMed] [Google Scholar]
  • 24.Maestroni GJ. Adrenergic regulation of haematopoiesis. Pharmacol Res. 1995;32:249–253. doi: 10.1016/s1043-6618(05)80012-x. [DOI] [PubMed] [Google Scholar]
  • 25.Maestroni GJ, Conti A, Pedrinis E. Effect of adrenergic agents on hematopoiesis after syngenic bone marrow transplantation in mice. Blood. 80;1992:1178–1182. [PubMed] [Google Scholar]
  • 26.Maestroni GJ, Conti A. Noradrenergic modulation of lymphohematopoiesis. Int. J. Immunopharmacol. 1994;16:117–122. doi: 10.1016/0192-0561(94)90067-1. [DOI] [PubMed] [Google Scholar]
  • 27.Tran MA, Dang Tran LAC, Lafontan M, Montastruc P. Adrenergic neurohumoral influences on FFA release from bone marrow adipose tissue. J. Pharmacol. 1985;16:171–179. [PubMed] [Google Scholar]
  • 28.Benestad HB, Strom-Gundersen I, Iversen PO, Haug E, Nja A. No neuronal regulation of murine bone marrow function. Blood. 1998;91:1280–1287. [PubMed] [Google Scholar]
  • 29.Besedovsky HO, del Rey AE, Sorkin E, Burri R, Honegger CG, Schlumpf M, Lichtensteiger W. T lymphocytes affect the development of sympathetic innervation of mouse spleen. Brain Behav. Immun. 1987;1:185–193. doi: 10.1016/0889-1591(87)90020-1. [DOI] [PubMed] [Google Scholar]
  • 30.Bulloch K, Pomerantz W. Autonomic nervous system innervation of thymic-related lymphoid tissue in wild type and nude mice. J. Comp. Neurol. 1984;228:57–68. doi: 10.1002/cne.902280107. [DOI] [PubMed] [Google Scholar]
  • 31.Bellinger DL, Felten SY, Felten DL. Maintenance of noradrenergic sympathetic innervation of the involuted thymus of the aged Fischer 344 rats. Brain Behav. Immun. 1988;2:133–150. doi: 10.1016/0889-1591(88)90014-1. [DOI] [PubMed] [Google Scholar]
  • 32.Felten DL, Felten SY. Innervation of the thymus. In: Kendal MD, Ritter MA, editors. Thymus Update. London: Harwood Academic Publishers; 1989. pp. 73–88. [Google Scholar]
  • 33.Ackerman KD, Bellinger DL, Felten SY, Felten DL. Ontogeny and senescence of noradrenergic innervation of the rodent thymus and spleen. In: Ader R, Felten DL, Cohen N, editors. Psychoneuroimmunology. 2nd edition. New York: Academic Press; 1991. pp. 71–125. [Google Scholar]
  • 34.Kurz B, Feindt J, Von Gaudecker B, Kranz A, Loppnow H, Mentlein R. Beta-adrenoceptor-mediated effects in rat cultured thymic epithelial cells. Br. J. Pharmacol. 1997;120:1041–1408. doi: 10.1038/sj.bjp.0701045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kendall MD, Al-Shawaf AA. Innervation of the rat thymus gland. Brain Behav. Immun. 1991;5:9–28. doi: 10.1016/0889-1591(91)90004-t. [DOI] [PubMed] [Google Scholar]
  • 36.Vizi ES, Orso E, Osipenko ON, Hasko G, Elenkov IJ. Neurochemical, electrophysiological and immunocytochemical evidence for a noradrenergic link between the sympathetic nervous system and thymocytes. Neuroscience. 1995;68:1263–1276. doi: 10.1016/0306-4522(95)00215-5. [DOI] [PubMed] [Google Scholar]
  • 37.de Leeuw FE, Jansen GH, Batanero E, van Wichen DF, Huber J, Schuurman HJ. The neural and neuroendocrine component of the human thymus. I. Nerve-like structures. Brain Behav. Immun. 1992;6:234–248. doi: 10.1016/0889-1591(92)90046-q. [DOI] [PubMed] [Google Scholar]
  • 38.Kranz A, Kendall MD, Von Gaudecker B. Studies on rat and human thymus to demonstrate immunoreactivity of calcitonin gene-related peptide, tyrosine hydroxylase and neuropeptide Y. J. Anat. 1997;191:441–450. doi: 10.1046/j.1469-7580.1997.19130441.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Nance DM, Hopkins DA, Bieger D. Re-investigation of the innervation of the thymus gland in mice and rats. Brain Behav. Immun. 1987;1:134–147. doi: 10.1016/0889-1591(87)90016-x. [DOI] [PubMed] [Google Scholar]
  • 40.Tollefson L, Bulloch K. Dual-label retrograde transport: CNS innervation of the mouse thymus distinct from other mediastinum viscera. J. Neurosci. Res. 1990;25:20–28. doi: 10.1002/jnr.490250104. [DOI] [PubMed] [Google Scholar]
  • 41.Marchetti B, Morale MC, Paradis P, Bouvier M. Characterization, expression, and hormonal control of a thymic beta-2-adrenergic receptor. Am. J. Physiol. 1994;267:E718–E731. doi: 10.1152/ajpendo.1994.267.5.E718. [DOI] [PubMed] [Google Scholar]
  • 42.Leposavic G, Micic M, Ugresic N, Bogojevic M, Isakovic K. Components of sympathetic innervation of the rat thymus during late foetal and postnatal development: histofluorescence and biochemical study. Sympathetic innervation of the rat thymus. Thymus. 1992;19:77–87. [PubMed] [Google Scholar]
  • 43.Williams JM, Felten DL. Sympathetic innervation of murine thymus and spleen: a comparative study. Anat. Rec. 1981;199:531–542. doi: 10.1002/ar.1091990409. [DOI] [PubMed] [Google Scholar]
  • 44.Al-Shawaf AA, Kendall MD, Cowen T. Identification of neural profiles containing vasoactive intestinal polypeptide, acetylcholinesterase and catecholamines in the rat thymus. J. Anat. 1991;174:131–143. [PMC free article] [PubMed] [Google Scholar]
  • 45.Cavallotti C, Artico N, Cavallotti D. Occurrence of adrenergic nerve fibers and noradrenaline in thymus gland of juvenile and aged rats. Immunol. Lett. 1999;70:53–62. doi: 10.1016/s0165-2478(99)00127-3. [DOI] [PubMed] [Google Scholar]
  • 46.Müller S, Weihe E. Interrelation of peptidergic innervation with mast cells and ED1-positive cells in rat thymus. Brain Behav. Immun. 1991;5:55–72. doi: 10.1016/0889-1591(91)90007-w. [DOI] [PubMed] [Google Scholar]
  • 47.Brouxhon SM, Prasad AV, Joseph SA, Felten DL, Bellinger DL. Localization of corticotropin-releasing factor in primary and secondary lymphoid organs of the rat. Brain Behav. Immun. 1998;12:107–122. doi: 10.1006/brbi.1998.0520. [DOI] [PubMed] [Google Scholar]
  • 48.Elenkov IJ, Vizi ES. Presynaptic modulation of release of noradrenaline from the sympathetic nerve terminals in the rat spleen. Neuropharmacology. 1991;30:1319–1324. doi: 10.1016/0028-3908(91)90029-b. [DOI] [PubMed] [Google Scholar]
  • 49.Hasko G, Elenkov IJ, Vizi ES. Presynaptic receptors involved in the modulation of release of noradrenaline from the sympathetic nerve terminals of the rat thymus. Immunol. Lett. 1995;47:133–137. doi: 10.1016/0165-2478(95)00085-j. [DOI] [PubMed] [Google Scholar]
  • 50.Docherty JR. Subtypes of functional alpha1- and alpha2-adrenoceptors. Eur. J. Pharmacol. 1998;361:1–15. doi: 10.1016/s0014-2999(98)00682-7. [DOI] [PubMed] [Google Scholar]
  • 51.Wonnacott S. Presynaptic nicotinic Ach receptors. Trends Neurosci. 1998;20:92–98. doi: 10.1016/s0166-2236(96)10073-4. [DOI] [PubMed] [Google Scholar]
  • 52.Vizi ES, Lendvai B. Modulatory role of presynaptic nicotinic receptors in synaptic and nonsynaptic chemical communication in the central nervous system. Brain Res. Brain. Res. Rev. 1999;30:219–235. doi: 10.1016/s0165-0173(99)00016-8. [DOI] [PubMed] [Google Scholar]
  • 53.Madden KS. Catecholamines, sympathetic nerves, and immunity. In: Ader R, Cohen N, Felten DL, editors. Psychoneuroimmunology. 3rd edition. New York: Academic Press; 2001. pp. 197–216. [Google Scholar]
  • 54.Fuchs BA, Albright JW, Albright JF. Beta-adrenergic receptors on murine lymphocytes: density varies with cell maturity and lymphocyte subtype and is decreased after antigen administration. Cell. Immunol. 1988;114:231–245. doi: 10.1016/0008-8749(88)90318-8. [DOI] [PubMed] [Google Scholar]
  • 55.Radojcic T, Baird S, Darko D, Smith D, Bulloch K. Changes in beta-adrenergic receptor distribution on immunocytes during differentiation: an analysis of T cells and macrophages. J. Neurosci. Res. 1991;30:328–335. doi: 10.1002/jnr.490300208. [DOI] [PubMed] [Google Scholar]
  • 56.Morale MC, Gallo F, Batticane N, Marchetti B. The immune response evokes up-and downmodulation of beta 2-adrenergic receptor messenger RNA concentration in the male rat thymus. Mol. Endocrinol. 1992;6:1513–1524. doi: 10.1210/mend.6.9.1359402. [DOI] [PubMed] [Google Scholar]
  • 57.Von Patay B, Loppnow H, Feindt J, Kurz B, Mentlein R. Catecholamines and lipopolysaccharide synergistically induce the release of interleukin-6 from thymic epithelial cells. J. Neuroimmunol. 1998;86:182–189. doi: 10.1016/s0165-5728(98)00051-4. [DOI] [PubMed] [Google Scholar]
  • 58.Singh U, Owen JJT. Studies on the effect of various agents on the maturation of thymus stem cells. Eur. J. Immunol. 1975;5:286–288. doi: 10.1002/eji.1830050414. [DOI] [PubMed] [Google Scholar]
  • 59.Singh U, Owen JJT. Studies on the maturation of thymus stem cells. The effects of catecholamines, histamine, and peptide hormones on the expression of T allo-antigens. Eur. J. Immunol. 1976;6:59–62. doi: 10.1002/eji.1830060113. [DOI] [PubMed] [Google Scholar]
  • 60.Singh U. Effect of catecholamines on lymphopoiesis in foetal mouse thymic explants. Eur. J. Immunol. 1979;14:757–759. [PMC free article] [PubMed] [Google Scholar]
  • 61.Singh U. Effect of catecholamines on lymphopoiesis in foetal mouse thymic explants. J. Anat. 1979;129:279–292. [PMC free article] [PubMed] [Google Scholar]
  • 62.Singh U. Lymphopoiesis in the nude foetal mouse thymus following sympathectomy. Cell. Immunol. 1985;93:222–228. doi: 10.1016/0008-8749(85)90402-2. [DOI] [PubMed] [Google Scholar]
  • 63.Singh U. Effect of sympathectomy on the maturation of foetal thymocytes grown within the anterior eye chambers in mice. Adv. Exp. Med. Biol. 1985;186:349–356. doi: 10.1007/978-1-4613-2463-8_42. [DOI] [PubMed] [Google Scholar]
  • 64.Durant S. In vivo effects of catecholamines and glucocorticoids on mouse thymic cAMP content and thymolysis. Cell. Immunol. 1986;102:136–143. doi: 10.1016/0008-8749(86)90332-1. [DOI] [PubMed] [Google Scholar]
  • 65.Cook-Mills JM, Cohen RL, Perlman RL, Chambers DA. Inhibition of lymphocyte activation by catecholamines: evidence for a non-classical mechanism of catecholamine action. Immunology. 1995;85:544–549. [PMC free article] [PubMed] [Google Scholar]
  • 66.Anderson KL, Anderson G, Michell RH, Jenkinson EJ, Owen JJ. Intracellular signaling pathways involved in the induction of apoptosis in immature thymic T lymphocytes. J. Immunol. 1996;156:4083–4091. [PubMed] [Google Scholar]
  • 67.McConkey DJ, Orrenius S, Jondal M. Agents that elevate cAMP stimulate DNA fragmentation in thymocytes. J. Immunol. 1990;145:1227–1230. [PubMed] [Google Scholar]
  • 68.Suzuki K, Tadakuma T, Kizaki H. Modulation of thymocyte apoptosis by isoproterenol and prostaglandin E2. Cell. Immunol. 1991;134:235–240. doi: 10.1016/0008-8749(91)90346-d. [DOI] [PubMed] [Google Scholar]
  • 69.Morgan JI, Wigham CG, Perris AD. The promotion of mitosis in cultured thymic lymphocytes by acetylcholine and catecholamines. J. Pharm. Pharmacol. 1984;36:511–515. doi: 10.1111/j.2042-7158.1984.tb04441.x. [DOI] [PubMed] [Google Scholar]
  • 70.Delrue-Perollet C, Li KS, Vitiello S, Neveu PJ. Peripheral catecholamines are involved in the neuroendocrine and immune effects of LPS. Brain Behav. Immun. 1995;9:149–162. doi: 10.1006/brbi.1995.1014. [DOI] [PubMed] [Google Scholar]
  • 71.Tsao CW, Cheng JT, Shen CL, Lin YS. 6-Hydroxydopamine induces thymocyte apoptosis in mice. J. Neuroimmunol. 1996;65:91–95. doi: 10.1016/0165-5728(95)00166-2. [DOI] [PubMed] [Google Scholar]
  • 72.Kostrzewa RM, Jacobwitz DM. Pharmacological actions of 6-hydroxydopamine. Pharmacol. Rev. 1974;26:199–288. [PubMed] [Google Scholar]
  • 73.Rauski A, Kosec D, Vidic-Dankovic B, Plecas-Solarovic B, Leposavic G. Effects of beta-adrenoceptor blockade on the phenotypic characteristics of thymocytes and peripheral blood lymphocytes. Int. J. Neurosci. 2003;113:1653–1673. doi: 10.1080/00207450390245216. [DOI] [PubMed] [Google Scholar]
  • 74.Rauski A, Kosec D, Vidic-Dankovic B, Radojevic K, Plecas-Solarovic B, Leposavic G. Thymopoiesis following chronic blockade of beta-adrenoceptors, Immunopharmacol. Immunotoxicol. 2003;25:513–528. doi: 10.1081/iph-120026437. [DOI] [PubMed] [Google Scholar]
  • 75.Blanco A, Artacho-Perula E, Flores-Acuna R, Moyano R, Monterde JG. Quantitative changes in the normal and apoptotic thymocytes of pigs treated with anabolic doses of the beta2 adrenergic agonist clenbuterol. Vet. Immunol. Immunopathol. 2003;96:111–115. doi: 10.1016/s0165-2427(03)00162-4. [DOI] [PubMed] [Google Scholar]
  • 76.Madden KS, Felten DL. Beta-adrenoceptor blockade alters thymocyte differentiation in aged mice. Cell. Mol. Biol. (Noisy-le-grand) 2001;47:189–196. [PubMed] [Google Scholar]
  • 77.Madden KS, Bellinger DL, Felten SY, Snyder E, Maida ME, Felten DL. Alterations in sympathetic innervation of thymus and spleen in aged mice. Mech. Ageing Dev. 1997;94:165–175. doi: 10.1016/s0047-6374(96)01858-1. [DOI] [PubMed] [Google Scholar]
  • 78.Plecas-Solarovic B, Hristic-Zivkovic I, Radojevic K, Kosec D, Leposavic G. Chronic alpha1-adrenoreceptor blockade produces age-dependent changes in rat thymus structure and thymocyte differentiation. Histol. Histopathol. 2005;20:833–841. doi: 10.14670/HH-20.833. [DOI] [PubMed] [Google Scholar]
  • 79.Colic M, Cupic V, Pavicic L, Vucevic D, Varagic VM. Xylazine, an alpha 2-adrenergic agonist, modulates proliferation of rat thymocytes in vivo and in vitro. Methods Find. Exp. Clin. Pharmacol. 2000;22:557–562. [PubMed] [Google Scholar]
  • 80.Cupic V, Colic M, Jandric D, Milojkovic B, Varagic VM. Xylazine, an alpha 2-adrenergic agonist, induces apoptosis of rat thymocytes and a thymocyte hybridoma line in vitro. Methods Find. Exp. Clin. Pharmacol. 2003;25:5–10. [PubMed] [Google Scholar]
  • 81.Le JM, Fredrickson G, Reis LF, Diamantstein T, Hirano T, Kishimoto T, Vilcek J. Interleukin 2-dependent and interleukin 2-independent pathways of regulation of thymocyte function by interleukin 6. Proc. Natl. Acad. Sci. U.S.A. 1988;85:8643–8647. doi: 10.1073/pnas.85.22.8643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Weihe E, Nohr D, Michel S, Muller S, Zentel HJ, Fink T, Krekel J. Molecular anatomy of the neuro-immune connection. Int. J. Neurosci. 1991;59:1–23. doi: 10.3109/00207459108985446. [DOI] [PubMed] [Google Scholar]
  • 83.Kaliner M, Orange RP, Austen KF. Immunological release of histamine and slow reacting substance of anaphylaxis from human lung. VI. Enhancement of cholinergic and alpha adrenergic stimulation. J. Exp. Med. 1972;136:556–670. doi: 10.1084/jem.136.3.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Tomita Y, Patterson R, Suszko IM. Respiratory mast cells and basophilic cells. II. Effects of pharmacological agents on 3’5’-adenosine monophosphate content and on antigen-induced histamine release. Int. Arch. Allergy Appl. Immunol. 1974;47:261–272. [PubMed] [Google Scholar]
  • 85.Bellinger DL, Felten SY, Lorton D, Felten DL. Origin of noradrenergic innervation of the spleen in rats. Brain Behav. Immun. 1989;3:291–311. doi: 10.1016/0889-1591(89)90029-9. [DOI] [PubMed] [Google Scholar]
  • 86.Nance DM, Burns J. Innervation of the spleen in the rat: evidence for absence of afferent innervation. Brain Behav. Immun. 1989;3:281–290. doi: 10.1016/0889-1591(89)90028-7. [DOI] [PubMed] [Google Scholar]
  • 87.Chen SH, Itoh M, Sun W, Miki T, Takenuchi Y. Localization of sympatheatic and parasympathetic neurons innervating pancreas and spleen in the cat. J. Auton. Nerv. Syst. 1996;59:12–16. doi: 10.1016/0165-1838(95)00136-0. [DOI] [PubMed] [Google Scholar]
  • 88.Trudrung P, Furness JB, Pompolo S, Messenger JP. Locations and chemistries of sympathetic nerve cells that project to the gastrointestinal tract and spleen. Arch. Histol. Cytol. 1994;57:139–150. doi: 10.1679/aohc.57.139. [DOI] [PubMed] [Google Scholar]
  • 89.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. doi: 10.1016/0306-4522(82)90135-x. [DOI] [PubMed] [Google Scholar]
  • 90.Ackerman KD, Felten SY, Dijkstra CD, Livnat S, Felten DL. Parallel development of noradrenergic innervation and cellular compartmentation in the rat spleen. Exp. Neurol. 1989;103:239–255. doi: 10.1016/0014-4886(89)90048-4. [DOI] [PubMed] [Google Scholar]
  • 91.Ackerman KD, Madden KS, Livnat S, Felten SY, Felten DL. Neonatal sympathetic denervation alters the development of in vitro spleen cell proliferation and differentiation. Brain Behav. Immun. 1991;5:235–261. doi: 10.1016/0889-1591(91)90021-2. [DOI] [PubMed] [Google Scholar]
  • 92.Fillenz M. Innervation of the cat spleen. Proc. Physiol. Soc. 1966:25–26. 2P–3P. doi: 10.1098/rspb.1970.0005. [DOI] [PubMed] [Google Scholar]
  • 93.Fillenz M. Innervation of blood vessels of lung and spleen. In: Karger B, editor. Sympathetic electric activation and innervation of blood vessels. London: Cambridge; 1966. pp. 56–59. [Google Scholar]
  • 94.Romano T, Felten SY, Olschowka JA, Felten DL. Noradrenergic and peptidergic innervation of lyphoid organs in the beluga, Delphinapterus leucas: an anatomical link between the nervous and immune systems. J. Morphol. 1994;221:243–259. doi: 10.1002/jmor.1052210302. [DOI] [PubMed] [Google Scholar]
  • 95.Bellinger DL, Ackerman KD, Felten SY, Felten DL. A longitudinal study of age-related loss of noradrenergic nerves and lymphoid cells in the rat spleen. Exp. Neurol. 1992;116:295–311. doi: 10.1016/0014-4886(92)90009-f. [DOI] [PubMed] [Google Scholar]
  • 96.Ackerman KD, Felten SY, Bellinger DL, Felten DL. Noradrenergic sympathetic innervation of the spleen: III. Development of innervation in the rat spleen. J. Neurosci. Res. 1987;18:49–54. 123–125. doi: 10.1002/jnr.490180109. [DOI] [PubMed] [Google Scholar]
  • 97.Bellinger DL, Felten SY, Collier TJ, Felten DL. Noradrenergic sympathetic innervation of the spleen: IV. Morphometric analysis in adult and aged F344 rats. J. Neurosci. Res. 1987;18:55–63. doi: 10.1002/jnr.490180110. [DOI] [PubMed] [Google Scholar]
  • 98.Felten DL, Ackerman KD, Wiegand SJ, Felten SY. Noradrenergic sympathetic innervation of the spleen: I. Nerve fibers associate with lymphocytes and macrophages in specific compartments of the splenic white pulp. J. Neurosci. Res. 1987;18:28–36. 118–126. doi: 10.1002/jnr.490180107. [DOI] [PubMed] [Google Scholar]
  • 99.Felten SY, Olschowka JA. Noradrenergic sympathetic innervation of the spleen. II Tyrosine hydroxylase (TH)-positive nerve terminals from synaptic-like contacts on lymphocytes in the splenic white pulp. J. Neurosci. Res. 1987;18:37–48. doi: 10.1002/jnr.490180108. [DOI] [PubMed] [Google Scholar]
  • 100.Stevens-Felten SY, Bellinger DL. Noradrenergic and peptidergic innervation of lymphoid organs. Chem. Immunol. 1997;69:99–131. doi: 10.1159/000058655. [DOI] [PubMed] [Google Scholar]
  • 101.Bellinger DL, Lorton D, Hamill RW, Felten SY, Felten DL. Acetylcholinesterase staining and choline acetyltransferase activity in the young adult rat spleen: lack of evidence for cholinergic innervation. Brain Behav. Immun. 1993;7:191–204. doi: 10.1006/brbi.1993.1021. [DOI] [PubMed] [Google Scholar]
  • 102.Fried G, Terenius L, Brodin E, Efendic S, Docray G, Fahrenkrug J, Goldstein M, Hokfelt T. Neuropeptide Y, enkephalin and noradrenaline coexist in sympathetic neurons innervating the bovine spleen. Biochemical and immunohistochemical evidence. Cell. Tissue Res. 1986;243:495–508. doi: 10.1007/BF00218056. [DOI] [PubMed] [Google Scholar]
  • 103.Romano TA, Felten SY, Felten DL, Olschowka JA. Neuropeptide-Y innervation of the rat spleen: another potential immunomodulatory neuropeptide. Brain Behav. Immun. 1991;5:116–131. doi: 10.1016/0889-1591(91)90011-x. [DOI] [PubMed] [Google Scholar]
  • 104.Ueda H, Abe M, Takehana K, Iwasa K, Hiraga T. Ultrastructure of the red pulp in spleen innervation in horse and pig. Acta Anatomica. 1991;141:151–158. doi: 10.1159/000147115. [DOI] [PubMed] [Google Scholar]
  • 105.Elfvin JG, Johansson J, Hoijer AS, Aldskogius H. The innervation of the splenic capsule in the guinea pig: an immunohistochemical and ultrastructural study. J. Anat. 1994;185:267–278. [PMC free article] [PubMed] [Google Scholar]
  • 106.Carlson SL, Albers KM, Beiting DJ, Parish M, Conner JM, Davis BM. NGF modulates sympathetic innervation of lymphoid tissues. J. Neurosci. 1995;15:5892–5899. doi: 10.1523/JNEUROSCI.15-09-05892.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bassett K, Molinaro CA, Millar AB, Flowers S, Perez SD, Karsten A, Bellinger DL. Age-related changes in splenic neurotrophin content: a possible cause for declining sympathetic innervation in old rats. West. Am. Fed. Med. Res. 2005:S55–S360. [Google Scholar]
  • 108.Lorton D, Lubahn C, Schaller J, Bellinger DL. Noradrenergic (NA) nerves in spleens from rats with adjuvant arthritis (AA) undergo an injury and sprouting response that parallel changes in NGF-positive cells and NGF tissue levels. Brain Behav. Immun. 2003;17:186–187. [Google Scholar]
  • 109.Giron LTJ, Crutcher KA, Davis JN. Lymph nodes-a possible site for sympathetic neuronal regulation of immune responses. Ann. Neurol. 1980;8:520–525. doi: 10.1002/ana.410080509. [DOI] [PubMed] [Google Scholar]
  • 110.Fink T, Weihe E. Multiple neuropeptides in nerves supplying mammalian lymph nodes: messenger candidates for sensory and autonomic neuroimmunomodulation? Neurosci. Lett. 1988;19:39–44. doi: 10.1016/0304-3940(88)90783-5. [DOI] [PubMed] [Google Scholar]
  • 111.Felten DL, Livnat S, Felten SY, Carlson SL, Bellinger DL, Yeh P. Sympathetic innervation of lymph nodes in mice. Brain Res. Bull. 1984;13:693–699. doi: 10.1016/0361-9230(84)90230-2. [DOI] [PubMed] [Google Scholar]
  • 112.Ackerman KD, Felten SY, Bellinger DL, Livnat S, Felten DL. Noradrenergic sympathetic innervation of spleen and lymph nodes in relation to specific cellular compartments. In: Cinader B, Miller RG, editors. Progress in Immunology IV. Orlando: Academic Press; 1987. pp. 588–600. [Google Scholar]
  • 113.Felten DL, Felten SY, Bellinger DL, Carlson SL, Ackerman KD, Madden KS, Olschowka 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. [DOI] [PubMed] [Google Scholar]
  • 114.Bellinger DL, Ackerman KD, Felten SY, Lorton D, Felten DL. Noradrenergic sympathetic innervation of thymus, spleen, and lymph nodes: Aspects of development, aging, and plasticity in neural-immune interactions. In: Nistico G, editor. Proceedings of a Symposium on Interactions between the Neuroendocrine and Immune Systems. Milano Rome: Pythagora Press; 1989. pp. 35–66. [Google Scholar]
  • 115.Novotny GE, Kliche KO. Innervation of lymph nodes: a combined silver impregnation and electron-microscopic study. Acta Anat. (Basel) 1986;127:243–248. doi: 10.1159/000146293. [DOI] [PubMed] [Google Scholar]
  • 116.Novotny GEK. Ultrastructural analysis of lymph node innervation in the rat. Acta Anat. 1988;133:57–61. doi: 10.1159/000146615. [DOI] [PubMed] [Google Scholar]
  • 117.Bienenstock J, Befus D, McDermott M, Mirski S, Rosenthal K. Regulation of lymphoblast traffic and localization in mucosal tissues, with emphasis on IgA. Fed. Proc. 1983;42:3213–3217. [PubMed] [Google Scholar]
  • 118.Husband AJ. Kinetics of extravasation and redistribution of IgA-specific antibody-containing cells in the intestine. J. Immunol. 1982;128:1355–1359. [PubMed] [Google Scholar]
  • 119.Jeurissen SH, Duijvestijn AM, Sontag Y, Kraal G. Lymphocyte migration into the lamina propria of the gut is mediated by specialized HEV-like blood vessels. Immunology. 1987;62:273–277. [PMC free article] [PubMed] [Google Scholar]
  • 120.Crivellato E, Soldano F, Travan L, Fusaroli P, Mallardi F. Apposition of enteric nerve fibers to plasma cells and immunoblasts in the mouse small bowel. Neurosci. Lett. 1998;241:123–126. doi: 10.1016/s0304-3940(98)00004-4. [DOI] [PubMed] [Google Scholar]
  • 121.Ueyama T, Kozuki K, Houtani T, Ikeda M, Kitajiri M, Yamashita T, Kumazawa T, Nagatsu I, Sugimoto T. Immunolocalization of tyrosine hydroxylase and vasoactive intestinal polypeptide in nerve fibers innervating human palatine tonsil and paratonsillar glands. Neurosci. Lett. 1990;116:70–74. doi: 10.1016/0304-3940(90)90388-p. [DOI] [PubMed] [Google Scholar]
  • 122.Yamashita T, Kumazawa H, Kozuki K, Amano H, Tomoda K, Kumazawa T. Autonomic nervous system in human palatine tonsil. Acta Otolaryngol. Suppl. 1984;416:63–71. [PubMed] [Google Scholar]
  • 123.Felten DL, Overhage JM, Felten SY, Schmedtje JF. Noradrenergic sympathetic innervation of lymphoid tissue in the rabbit appendix: further evidence for a link between the nervous and immune systems. Brain Res. Bull. 1981;7:595–612. doi: 10.1016/0361-9230(81)90010-1. [DOI] [PubMed] [Google Scholar]
  • 124.Jesseph JM, Felten DL. Noradrenergic innervation of the gut-associated lymphoid tissues (GALT) in the rabbit. Anat. Rec. 1984;208:81A. [Google Scholar]
  • 125.Nohr D, Weihe E. The neuroimmune link in the bronchus-associated lymphoid tissue (BALT) of cat and rat: peptides and neural markers. Brain Behav. Immun. 1991;5:84–101. doi: 10.1016/0889-1591(91)90009-y. [DOI] [PubMed] [Google Scholar]
  • 126.Bidart JM, Motte Ph, Assicot M, Bohuon C, Bellet D. Catechol-O-methyltransferase activity and aminergic binding sites distribution in human peripheral blood lymphocyte subpopulations. Clin. Immunol. Immunopathol. 1983;26:1–9. doi: 10.1016/0090-1229(83)90167-8. [DOI] [PubMed] [Google Scholar]
  • 127.Bishopric NH, Mcohen HJ, Lefkowitz RJ. Beta-adrenergic receptors in lymphocyte subpopulations. J. Allergy Clin. Immunol. 1980;65:29–33. doi: 10.1016/0091-6749(80)90173-6. [DOI] [PubMed] [Google Scholar]
  • 128.Khan MM, Sansoni P, Silverman ED, Engleman EG, Melmon KL. Beta-adrenergic receptors on human suppressor, helper, and cytolytic lymphocytes. Biol. Psychiatry. 1986;35:1137–1142. doi: 10.1016/0006-2952(86)90150-4. [DOI] [PubMed] [Google Scholar]
  • 129.Krawietz W, Werdan K, Schober M, Erdmann E, Rindfleisch GE, Hannig K. Different numbers of beta-receptors in human lymphocyte subpopulations. Biochem. Pharmacol. 1982;31:4613–4620. doi: 10.1016/0006-2952(82)90252-0. [DOI] [PubMed] [Google Scholar]
  • 130.Loveland BE, Jarrott B, McKenzie IF. The detection of beta-adrenoceptors on murine lymphocytes. Interntl J. Immunopharmacol. 1981;3:45–55. doi: 10.1016/0192-0561(81)90044-8. [DOI] [PubMed] [Google Scholar]
  • 131.Pochet R, Delespesse G. Beta-adrenoceptors display different efficiency on lymphocyte subpopulations. Biochem. Pharmacol. 1983;32:1651–1655. doi: 10.1016/0006-2952(83)90344-1. [DOI] [PubMed] [Google Scholar]
  • 132.Pochet R, Delespesse G, Gausset PW, Collet H. Distribution of beta-adrenergic receptors on human lymphocyte subpopulations. Clin. Exp. Immunol. 1979;38:578–584. [PMC free article] [PubMed] [Google Scholar]
  • 133.Van Tits LJ, Michel MC, Grosse-Wilde H, Happel M, Eigler FW, Soliman A, Brodde O-E. Catecholamines increase lymphocyte beta-2-adrenergic receptors via a beta-2-adrenergic, spleen-dependent process. Am. J. Physiol. 1990;258:E191–E202. doi: 10.1152/ajpendo.1990.258.1.E191. [DOI] [PubMed] [Google Scholar]
  • 134.Westly HJ, Kelley KW. Down-regulation of glucocorticoid and beta-adrenergic receptors on lectin-stimulated splenocytes. Proc. Soc. Exp. Biol. Med. 1987;185:211–218. doi: 10.3181/00379727-185-42537. [DOI] [PubMed] [Google Scholar]
  • 135.Williams LT, Snyderman R, Lefkowitz RJ. Identification of beta adrenergic receptors in human lymphocytes by (–)3H-alprenolol binding. J. Clin. Invest. 1976;57:149–155. doi: 10.1172/JCI108254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Bourne HR, Melmon KL. Adenyl cyclase in human leukocytes: Evidence for activation by separate beta adrenergic and prostaglandin receptors. J. Pharmacol. Exp. Ther. 1971;178:1–7. [PubMed] [Google Scholar]
  • 137.Conolly ME, Greenacre JK. The beta-adrenoceptor of the human lymphocyte and human lung parenchyma. Br. J. Pharmacol. 1977;59:17–23. doi: 10.1111/j.1476-5381.1977.tb06971.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Meurs H, Van Den Bogaard W, Kauffman HF, Bruynzeel PL. Characterization of~(—)-[3H]dihydroalprenolol binding to intact and broken cell preparations of human peripheral blood lymphocytes. Eur. J. Pharmacol. 1982;85:185–194. doi: 10.1016/0014-2999(82)90464-2. [DOI] [PubMed] [Google Scholar]
  • 139.Ramer-Quinn DS, Baker RA, Sanders VM. Activated Th1 and Th2 cells differentially express the beta-2-adrenergic receptor: A mechanism for selective modulation of Th1 cell cytokine production. J. Immunol. 1997;159:4857–4867. [PubMed] [Google Scholar]
  • 140.Sanders VM, Baker RA, Ramer-Quinn DS, Kasprowicz DJ, Fuchs BA, Street NE. Differential expression of the beta-2-adrenergic receptor by Th1 and Th2 clones: implication for cytokine production and B cell help. J. Immunol. 1997;158:4200–4210. [PubMed] [Google Scholar]
  • 141.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. [DOI] [PubMed] [Google Scholar]
  • 142.Kohm AP, Mozaffarian A, Sanders VM. B cell receptor- and beta 2-adrenergic receptor-induced regulation of B7-2 (CD86) expression in B cells. J. Immunol. 2002;168:6314–6322. doi: 10.4049/jimmunol.168.12.6314. [DOI] [PubMed] [Google Scholar]
  • 143.Sanders VM, Munson AE. Role of alpha adrenoceptor activation in modulating the murine primary antibody response in vitro. J. Pharmacol. Exp. Ther. 1985;232:395–400. [PubMed] [Google Scholar]
  • 144.Cazaux CA, Sterin-Borda L, Gorelik G, Cremaschi GA. Down-regulation of beta-adrenergic receptors induced by mitogen activation of intracellular signaling events in lymphocytes. F.E.B.S. Lett. 1995;364:120–124. doi: 10.1016/0014-5793(95)00366-h. [DOI] [PubMed] [Google Scholar]
  • 145.Madden KS, Felten SY, Felten DL, Sundaresan PR, Livnat S. Sympathetic neuronal modulation of the immune system. I. Depression of T cell immunity in vivo and vitro following chemical sympathectomy. Brain Behav. Immun. 1989;3:72–89. doi: 10.1016/0889-1591(89)90007-x. [DOI] [PubMed] [Google Scholar]
  • 146.Bartik MM, Bauman GP, Brooks WH, Roszman TL. Costimulatory signals modulate the antiproliferative effects of agents that elevate cAMP in T cells. Cell. Immunol. 1994;158:116–130. doi: 10.1006/cimm.1994.1261. [DOI] [PubMed] [Google Scholar]
  • 147.Brodde OE, Daul A, O'Hara N. Beta-adrenoceptor changes in human lymphocytes, induced by dynamic exercise. Naunyn Schmiedebergs Arch Pharmacol. 1984;325:190–192. doi: 10.1007/BF00506201. [DOI] [PubMed] [Google Scholar]
  • 148.Makman MH. Properties of adenylate cyclase of lymphoid cells. Proc. Natl. Acad. Sci. U.S.A. 1971;68:885–889. doi: 10.1073/pnas.68.5.885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bauman GP, Bartik MM, Brooks WH, Roszman TL. Induction of cAMP-dependent protein kinase (PKA) activity in T cells after stimulation of the prostaglandin E2 or the beta-adrenergic receptors: relationship between PKA activity and inhibition of anti-CD3 monoclonal antibody-induced T cell proliferation. Cell. Immunol. 1994;158:182–194. doi: 10.1006/cimm.1994.1266. [DOI] [PubMed] [Google Scholar]
  • 150.DeRubertis FR, Zenser TV, Adler WH, Hudson T. Role of cyclic adenosine 3',5'-monophosphate in lymphocyte mitogenesis. J. Immunol. 1974;113:151–161. [PubMed] [Google Scholar]
  • 151.Diamantstein T, Ulmer A. The antagonistic action of cyclic GMP and cyclic AMP on proliferation of B and T lymphocytes. Immunology. 1975;28:113–119. [PMC free article] [PubMed] [Google Scholar]
  • 152.Estes G, Solomon SS, Norton WL. Inhibition of lymphocyte stimulation by cyclic and non-cyclic nucleotides. J. Immunol. 1971;107:1489–1492. [PubMed] [Google Scholar]
  • 153.Ledbetter JA, Parsons M, Martin PJ, Hansen JA, Rabinovitch PS, June CH. Antibody binding to CD5 (Tp67) and Tp44 T cell surface molecules: effects on cyclic nucleotides, cytoplasmic free calcium, and cAMP-mediated suppression. J. Immunol. 1986;137:3299–3305. [PubMed] [Google Scholar]
  • 154.Smith JW, Steiner AL, Parker CW. Human lymphocytic metabolism. Effects of cyclic and noncyclic nucleotides on stimulation by phytohemagglutinin. J. Clin. Invest. 1971;50:442–448. doi: 10.1172/JCI106511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Vischer TL. The differential effect of cyclic AMP on lymphocyte stimulation by T- or B-cell mitogens. Immunology. 1976;30:735–739. [PMC free article] [PubMed] [Google Scholar]
  • 156.Weinstein Y, Segal S, Melmon KL. Specific mitogenic activity of 8-Br-guanosine 3', 5’-monophosphate (Br-cyclic GMP) on B lymphocytes. J. Immunol. 1975;115:112–117. [PubMed] [Google Scholar]
  • 157.Cremaschi GA, Fisher P, Boege F. Beta-adrenoceptor distribution in murine lymphoid cell lines. Immunopharmacology. 1991;22:195–206. doi: 10.1016/0162-3109(91)90044-y. [DOI] [PubMed] [Google Scholar]
  • 158.Miles K, Atweh S, Otten G, Arnason BG, Chelmicka-Schorr E. Beta-adrenergic receptors on splenic lymphocytes from axotomized mice. Int. J. Immunopharmacol. 1984;6:171–177. doi: 10.1016/0192-0561(84)90014-6. [DOI] [PubMed] [Google Scholar]
  • 159.Miles K, Chelmicka-Schorr E, Atweh S, Otten G, Arnason BG. Sympathetic ablation alters lymphocyte membrane properties. J. Immunol. 1985;135(Suppl. 2):797s–801s. [PubMed] [Google Scholar]
  • 160.Kohm AP, Sanders VM. Suppression of antigen-specific Th2 cell-dependent IgM and IgG1 production following norepinephrine depletion in vivo. J. Immunol. 1999;162:5299–5308. [PubMed] [Google Scholar]
  • 161.Kasprowicz DJ, Kohm AP, Berton MT, Chruscinski AJ, Sharpe A, Sanders VM. Stimulation of the B cell receptor, CD86 (B7-2), and the beta 2-adrenergic receptor intrinsically modulates the level of IgG1 and IgE produced per B cell. J. Immunol. 2000;165:680–690. doi: 10.4049/jimmunol.165.2.680. [DOI] [PubMed] [Google Scholar]
  • 162.Blomhoff HK, Smeland EB, Beiske K, Blomhoff R, Ruud E, Bjoro T, Pfeifer-Ohlsson S, Watt R, Funderud S, Godal T, et al. Cyclic AMP-mediated suppression of normal and neoplastic B cell proliferation is associated with regulation of myc and Ha-ras protooncogenes. J. Cell. Physiol. 1987;131:426–433. doi: 10.1002/jcp.1041310315. [DOI] [PubMed] [Google Scholar]
  • 163.Cohen DP, Rothstein TL. Adenosine 3',5'-cyclic monophosphate modulates the mitogenic responses of murine B lymphocytes. Cell. Immunol. 1989;121:113–124. doi: 10.1016/0008-8749(89)90009-9. [DOI] [PubMed] [Google Scholar]
  • 164.Muthusamy N, Baluyut AR, Subbarao B. Differential regulation of surface Ig- and Lyb2-mediated B cell activation by cyclic AMP. I. Evidence for alternative regulation of signaling through two different receptors linked to phosphatidylinositol hydrolysis in murine B cells. J. Immunol. 1991;147:2483–2492. [PubMed] [Google Scholar]
  • 165.Whisler RL, Beiqing L, Grants IS, Newhouse YG. Cyclic AMP modulation of human B cell proliferative responses: role of cAMP-dependent protein kinases in enhancing B cell responses to phorbol diesters and ionomycin. Cell. Immunol. 1992;142:398–415. doi: 10.1016/0008-8749(92)90300-e. [DOI] [PubMed] [Google Scholar]
  • 166.Holte H, Torjesen P, Blomhoff HK, Ruud E, Funderud S, Smeland EB. Cyclic AMP has the ability to influence multiple events during B cell stimulation. Eur. J. Immunol. 1988;18:1359–1366. doi: 10.1002/eji.1830180909. [DOI] [PubMed] [Google Scholar]
  • 167.Li YS, Kouassi E, Revillard JP. Cyclic AMP can enhance mouse B cell activation by regulating progression into the late G1/S phase. Eur. J. Immunol. 1989;19:1721–1725. doi: 10.1002/eji.1830190929. [DOI] [PubMed] [Google Scholar]
  • 168.Jetschmann JU, Benschop RJ, Jacobs R, Kemper A, Oberbeck R, Schmidt RE, Schedlowski M. Expression and in vivo modulation of alpha- and beta-adrenoceptors on human natural killer (CD16+) cells. J. Neuroimmunol. 1997;74:159–164. doi: 10.1016/s0165-5728(96)00221-4. [DOI] [PubMed] [Google Scholar]
  • 169.Benschop RJ, Oostveen FG, Heijnen CJ, Ballieux RE. Beta 2-adrenergic stimulation causes detachment of natural killer cells from cultured endothelium. Eur. J. Immunol. 1993;23:3242–3247. doi: 10.1002/eji.1830231230. [DOI] [PubMed] [Google Scholar]
  • 170.Maisel AS, Harris T, Rearden CA, Michel MC. Beta-adrenergic receptors in lymphocyte subsets after exercise. Alterations in normal individuals and patients with congestive heart failure. Circulation. 1990;82:2003–2010. doi: 10.1161/01.cir.82.6.2003. [DOI] [PubMed] [Google Scholar]
  • 171.Takamoto T, Hori Y, Koga Y, Toshima H, Hara A, Yokoyama MM. Norepinephrine inhibits human natural killer cell activity in vitro. Int. J. Neurosci. 1991;58:127–131. doi: 10.3109/00207459108987189. [DOI] [PubMed] [Google Scholar]
  • 172.Whalen MM, Bankhurst AD. Effects of beta-adrenergic receptor activation, cholera toxin and forskolin on human natural killer cell function. Biochem. J. 1990;272:327–331. doi: 10.1042/bj2720327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Murray DR, Irwin M, Rearden CA, Ziegler M, Motulsky H, Maisel AS. Sympathetic and immune interactions during dynamic exercise. Mediation via a beta 2-adrenergic-dependent mechanism. Circulation. 1992;86:203–213. doi: 10.1161/01.cir.86.1.203. [DOI] [PubMed] [Google Scholar]
  • 174.Schedlowski M, Hosch W, Oberbeck R, Benschop RJ, Jacobs R, Raab HR, Schmidt RE. Catecholamines modulate human NK cell circulation and function via spleen-independent beta 2-adrenergic mechanisms. J. Immunol. 1996;156:93–99. [PubMed] [Google Scholar]
  • 175.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. [PubMed] [Google Scholar]
  • 176.Abrass CK, O'Connor SW, Scarpace PJ, Abrass IB. Characterization of the beta-adrenergic receptor of the rat peritoneal macrophage. J. Immunol. 1985;135:1338–1341. [PubMed] [Google Scholar]
  • 177.Rosati C, Hannaert P, Dausse JP, Braquet P, Garay R. Stimulation of beta-adrenoceptors inhibits calcium-dependent potassium-channels in mouse macrophages. J. Cell. Physiol. 1986;129:310–314. doi: 10.1002/jcp.1041290307. [DOI] [PubMed] [Google Scholar]
  • 178.Hjemdahl P, Larsson K, Johansson MC, Zetterlund A, Eklund A. Beta-adrenoceptors in human alveolar macrophages isolated by elutriation. Br. J. Clin. Pharmacol. 1990;30:673–682. doi: 10.1111/j.1365-2125.1990.tb03835.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Liggett SB. Identification and characterization of a homogeneous population of beta 2-adrenergic receptors on human alveolar macrophages. Am. Rev. Respir. Dis. 1989;139:552–555. doi: 10.1164/ajrccm/139.2.552. [DOI] [PubMed] [Google Scholar]
  • 180.Henricks PA, Van Esch B, Van Oosterhout AJ, Nijkamp FP. Specific and non-specific effects of beta-adrenoceptor agonists on guinea pig alveolar macrophage function. Eur. J. Pharmacol. 1988;152:321–330. doi: 10.1016/0014-2999(88)90727-3. [DOI] [PubMed] [Google Scholar]
  • 181.van Esch B, Henricks PA, van Oosterhout AJ, Nijkamp FP. Guinea pig alveolar macrophages possess beta-adrenergic receptors. Agents Actions. 1989;26:123–124. doi: 10.1007/BF02126582. [DOI] [PubMed] [Google Scholar]
  • 182.Remold-O'Donnell E. Stimulation and desensitization of macrophage adenylate cyclase by prostaglandins and catecholamines. J. Biol. Chem. 1974;249:3615–3621. [PubMed] [Google Scholar]
  • 183.Higgins TJ, David JR. Effect of isoproterenol and aminiphylline on cyclic AMP levels of guinea pig macrophages. Cell. Immunol. 1976;27:1–10. doi: 10.1016/0008-8749(76)90147-7. [DOI] [PubMed] [Google Scholar]
  • 184.Ikegami K. Modulation of adenosine 3',5'-monophosphate contents of rat peritoneal macrophages mediated by beta2-adrenergic receptors. Biochem. Pharmacol. 1977;26:1813–1816. doi: 10.1016/0006-2952(77)90351-3. [DOI] [PubMed] [Google Scholar]
  • 185.Lavis VR, Strada SJ, Ross CP, Hersh EM, Thompson WJ. Comparison of the responses of freshly isolated and cultured human monocytes and P388D1 cells to agents affecting cyclic AMP metabolism. J. Lab. Clin. Med. 1980;96:551–561. [PubMed] [Google Scholar]
  • 186.Verghese MW, Snyderman R. Hormonal activation of adenylate cyclase in macrophage membranes is regulated by guanine nucleotides. J. Immunol. 1983;130:869–873. [PubMed] [Google Scholar]
  • 187.Welscher HD, Cruchaud A. The influence of various particles and 3', 5' cyclic adenosine monophosphate on release of lysosomal enzymes by mouse macrophages. J. Reticuloendothel. Soc. 1976;20:405–420. [PubMed] [Google Scholar]
  • 188.Hara N, Ichinose M, Sawada M, Maeno T. The activation of Ca(2+)-dependent K+ conductance by adrenaline in mouse peritoneal macrophages. Pflugers Arch. 1991;419:371–379. doi: 10.1007/BF00371119. [DOI] [PubMed] [Google Scholar]
  • 189.Young JD, Ko SS, Cohn ZA. The increase in intracellular free calcium associated with IgG gamma 2b/gamma 1 Fc receptor-ligand interactions: role in phagocytosis. Proc. Natl. Acad. Sci. U.S.A. 1984;81:5430–5434. doi: 10.1073/pnas.81.17.5430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Sanders PNI II chapter
  • 191.Bartik MM, Brooks WH, Roszman TL. Modulation of T cell proliferation by stimulation of the beta-adrenergic receptor: lack of correlation between inhibition of T cell proliferation and cAMP accumulation. Cell. Immunol. 1993;148:408–421. doi: 10.1006/cimm.1993.1122. [DOI] [PubMed] [Google Scholar]
  • 192.Carlson SL, Brooks WH, Roszman TL. Neurotransmitter-lymphocyte interactions: dual receptor modulation of lymphocyte proliferation and cAMP production. J. Neuroimmunol. 1989;24:155–162. doi: 10.1016/0165-5728(89)90109-4. [DOI] [PubMed] [Google Scholar]
  • 193.Carlson SL, Trauth K, Brooks WH, Roszman TL. Enhancement of beta-adrenergic-induced cAMP accumulation in activated T-cells. J. Cell. Physiol. 1994;161:39–48. doi: 10.1002/jcp.1041610106. [DOI] [PubMed] [Google Scholar]
  • 194.Hadden JW, Hadden EM, Middleton E., Jr Lymphocyte blast transformation. I. Demonstration of adrenergic receptors in human peripheral lymphocytes. Cell. Immunol. 1970;1:583–595. doi: 10.1016/0008-8749(70)90024-9. [DOI] [PubMed] [Google Scholar]
  • 195.Johnson DL, Ashmore RC, Gordon MA. Effects of beta-adrenergic agents on the murine lymphocyte response to mitogen stimulation. J. Immunopharmacol. 1981;3:205–219. doi: 10.3109/08923978109026427. [DOI] [PubMed] [Google Scholar]
  • 196.Johnson DL, Gordon MA. Effect of chronic beta-adrenergic therapy on the human lymphocyte response to concanavalin A. Res. Commun. Chem. Pathol. Pharmacol. 1981;32:377–380. [PubMed] [Google Scholar]
  • 197.Betz M, Fox BS. Prostaglandin E2 inhibits production of Th1 lymphokines but not of Th2 lymphokines. J. Immunol. 1991;146:108–113. [PubMed] [Google Scholar]
  • 198.Gajewski TF, Schell SR, Fitch FW. Evidence implicating utilization of different T cell receptor-associated signaling pathways by TH1 and TH2 clones. J. Immunol. 1990;144:4110–4120. [PubMed] [Google Scholar]
  • 199.Qiu Y, Peng Y, Wang J. Immunoregulatory role of neurotransmitters. Adv. Neuroimmunol. 1996;6:223–231. doi: 10.1016/s0960-5428(96)00018-6. [DOI] [PubMed] [Google Scholar]
  • 200.Feldman RD, Hunninghake GW, McArdle WL. Beta-adrenergic-receptor-mediated suppression of interleukin 2 receptors in human lymphocytes. J. Immunol. 1987;139:3355–3359. [PubMed] [Google Scholar]
  • 201.Krause DS, Deutsch C. Cyclic AMP directly inhibits IL-2 receptor expression in human T cells: expression of both p55 and p75 subunits is affected. J. Immunol. 1991;146:2285–2296. [PubMed] [Google Scholar]
  • 202.Mary D, Peyron JF, Auberger P, Aussel C, Fehlmann M. Modulation of T cell activation by differential regulation of the phosphorylation of two cytosolic proteins, Implication of both Ca2+ and cyclic AMP-dependent protein kinases. J. Biol. Chem. 1989;264:14498–14502. [PubMed] [Google Scholar]
  • 203.Novak TJ, Rothenberg EV. cAMP inhibits induction of interleukin 2 but not of interleukin 4 in T cells. Proc. Natl. Acad. Sci. U.S.A. 1990;87:9353–9357. doi: 10.1073/pnas.87.23.9353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Selliah N, Bartik MM, Carlson SL, Brooks WH, Roszman TL. cAMP accumulation in T-cells inhibits anti-CD3 monoclonal antibody-induced actin polymerization. J. Neuroimmunol. 1995;56:107–112. doi: 10.1016/0165-5728(94)00142-b. [DOI] [PubMed] [Google Scholar]
  • 205.Wacholtz MC, Minakuchi R, Lipsky PE. Characterization of the 3',5'-cyclic adenosine monophosphate-mediated regulation of IL2 production by T cells and Jurkat cells. Cell. Immunol. 1991;135:285–298. doi: 10.1016/0008-8749(91)90274-f. [DOI] [PubMed] [Google Scholar]
  • 206.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. [DOI] [PubMed] [Google Scholar]
  • 207.Katamura K, Shintaku N, Yamauchi Y, Fukui T, Ohshima Y, Mayumi M, Furusho K. Prostaglandin E2 at priming of naive CD4+ T cells inhibits acquisition of ability to produce IFN-gamma and IL-2, but not IL-4 and IL-5. J. Immunol. 1995;155:4604–4612. [PubMed] [Google Scholar]
  • 208.Paliogianni F, Boumpas DT. Prostaglandin E2 inhibits the nuclear transcription of the human interleukin 2, but not the IL-4, gene in human T cells by targeting transcription factors AP-1 and NF-AT. Cell. Immunol. 1996;171:95–101. doi: 10.1006/cimm.1996.0178. [DOI] [PubMed] [Google Scholar]
  • 209.Yoshimura T, Nagao T, Nakao T, Watanabe S, Usami E, Kobayashi J, Yamazaki F, Tanaka H, Inagaki N, Nagai H. Modulation of Th1- and Th2-like cytokine production from mitogen-stimulated human peripheral blood mononuclear cells by phosphodiesterase inhibitors. Gen. Pharmacol. 1998;30:175–180. doi: 10.1016/s0306-3623(97)00103-1. [DOI] [PubMed] [Google Scholar]
  • 210.Borger P, Kauffman HF, Postma DS, Vellenga E. Interleukin-4 gene expression in activated human T lymphocytes is regulated by the cyclic adenosine monophosphate-dependent signaling pathway. Blood. 1996;87:691–698. [PubMed] [Google Scholar]
  • 211.Crocker IC, Townley RG, Khan MM. Phosphodiesterase inhibitors suppress proliferation of peripheral blood mononuclear cells and interleukin-4 and -5 secretion by human T-helper type 2 cells. Immunopharmacology. 1996;31:223–235. doi: 10.1016/0162-3109(95)00053-4. [DOI] [PubMed] [Google Scholar]
  • 212.Lacour M, Arrighi JF, Muller KM, Carlberg C, Saurat JH, Hauser C. cAMP up-regulates IL-4 and IL-5 production from activated CD4+ T cells while decreasing IL-2 release and NF-AT induction. Int. Immunol. 1994;6:1333–1343. doi: 10.1093/intimm/6.9.1333. [DOI] [PubMed] [Google Scholar]
  • 213.Wirth S, Lacour M, Jaunin F, Hauser C. Cyclic adenosine monophosphate (cAMP) differentially regulates IL-4 in thymocyte subsets. Thymus. 1996;24:101–109. [PubMed] [Google Scholar]
  • 214.Madden KS, Moynihan JA, Brenner GJ, Felten SY, Felten DL, Livnat S. Sympathetic nervous system modulation of the immune system. III. Alterations in T and B cell proliferation and differentiation in vitro following chemical sympathectomy. J. Neuroimmunol. 1994;49:77–87. doi: 10.1016/0165-5728(94)90183-x. [DOI] [PubMed] [Google Scholar]
  • 215.Hatfield SM, Petersen BH, DiMicco JA. Beta adrenoceptor modulation of the generation of murine cytotoxic T lymphocytes in vitro. J. Pharmacol. Exp. Ther. 1986;239:460–466. [PubMed] [Google Scholar]
  • 216.Cook-Mills JM, Mokyr MB, Cohen RL, Perlman RL, Chambers DA. Neurotransmitter suppression of the in vitro generation of a cytotoxic T lymphocyte response against the syngeneic MOPC-315 plasmacytoma. Cancer Immunol. Immunother. 1995;40:79–87. doi: 10.1007/BF01520288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Strom TB, Carpenter CB, Garovoy MR, Austen KF, Merrill JP, Kaliner M. The modulating influence of cyclic nucleotides upon lymphocyte-mediated cytotoxicity. J. Exp. Med. 1973;138:381–393. doi: 10.1084/jem.138.2.381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Bender BS, Bell WE, Taylor S, Scarpace PJ. Decreased sensitivity to cAMP in the in vitro generation of memory splenic cytotoxic T-lymphocytes from aged mice: role of phosphodiesterase. J. Pharmacol. Exp. Ther. 1993;264:1381–1386. [PubMed] [Google Scholar]
  • 219.Takayama H, Trenn G, Sitkovsky MV. Locus of inhibitory action of cAMP-dependent protein kinase in the antigen receptor-triggered cytotoxic T lymphocyte activation pathway. J. Biol. Chem. 1988;263:2330–2336. [PubMed] [Google Scholar]
  • 220.Kouassi E, Li YS, Boukhris W, Millet I, Revillard JP. Opposite effects of the catecholamines dopamine and norepinephrine on murine polyclonal B-cell activation. Immunopharmacology. 1988;16:125–137. doi: 10.1016/0162-3109(88)90001-x. [DOI] [PubMed] [Google Scholar]
  • 221.Burchiel SW, Melmon KL. Augmentation of the in vitro humoral immune response by pharmacologic agents. I: An explanation for the differential enhancement of humoral immunity via agents that elevate cAMP. Immunopharmacology. 1979;1:137–150. doi: 10.1016/0162-3109(79)90050-x. [DOI] [PubMed] [Google Scholar]
  • 222.Gilbert KM, Hoffmann MK. cAMP is an essential signal in the induction of antibody production by B cells but inhibits helper function of T cells. J. Immunol. 1985;135:2084–2089. [PubMed] [Google Scholar]
  • 223.Melmon KL, Bourne HR, Weinstein Y, Shearer GM, Kram J, Bauminger S. Hemolytic plaque formation by leukocytes in vitro. Control by vasoactive hormones. J. Clin. Invest. 1974;53:13–21. doi: 10.1172/JCI107530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Patke CL, Orson FM, Shearer WT. Cyclic AMP-mediated modulation of immunoglobulin production in B cells by prostaglandin E1. Cell. Immunol. 1991;137:36–45. doi: 10.1016/0008-8749(91)90054-f. [DOI] [PubMed] [Google Scholar]
  • 225.Watson J, Epstein R, Cohn M. Cyclic nucleotides as intracellular mediators of the expression of antigen-sensitive cells. Nature. 1973;246:405–409. doi: 10.1038/246405a0. [DOI] [PubMed] [Google Scholar]
  • 226.Hall NR, McClure JE, Hu SK, Tare NS, Seals CM, Goldstein AL. Effects of 6-hydroxydopamine upon primary and secondary thymus dependent immune responses. Immunopharmacology. 1982;5:39–48. doi: 10.1016/0162-3109(82)90035-2. [DOI] [PubMed] [Google Scholar]
  • 227.Kasahara K, Tanaka S, Hamashima Y. Suppressed immune response to T-cell dependent antigen in chemically sympathectomized mice. Res. Commun. Chem. Pathol. Pharmacol. 1977;18:533–542. [PubMed] [Google Scholar]
  • 228.Livnat S, Felten SY, Carlson SL, Bellinger DL, Felten DL. Involvement of peripheral and central catecholamine systems in neural-immune interactions. J. Neuroimmunol. 1985;10:5–30. doi: 10.1016/0165-5728(85)90031-1. [DOI] [PubMed] [Google Scholar]
  • 229.Chelmicka-Schorr E, Checinski M, Arnason BG. Chemical sympathectomy augments the severity of experimental allergic encephalomyelitis. J. Neuroimmunol. 1988;17:347–350. doi: 10.1016/0165-5728(88)90125-7. [DOI] [PubMed] [Google Scholar]
  • 230.Miles K, Quintans J, Chelmicka-Schorr E, Arnason BG. The sympathetic nervous system modulates antibody response to thymus-independent antigens. J. Neuroimmunol. 1981;1:101–105. doi: 10.1016/0165-5728(81)90012-6. [DOI] [PubMed] [Google Scholar]
  • 231.Kruszewska B, Felten SY, Moynihan JA. Alterations in cytokine and antibody production following chemical sympathectomy in two strains of mice. J. Immunol. 1995;155:4613–4620. [PubMed] [Google Scholar]
  • 232.Kruszewska B, Felten DL, Stevens SY, Moynihan JA. Sympathectomy-induced immune changes are not abrogated by the glucocorticoid receptor blocker RU-486. Brain Behav. Immun. 1998;12:181–200. doi: 10.1006/brbi.1998.0527. [DOI] [PubMed] [Google Scholar]
  • 233.Esquifino AI, Cardinali DP. Local regulation of the immune response by the autonomic nervous system. Neuroimmunomodulation. 1994;1:265–273. doi: 10.1159/000097175. [DOI] [PubMed] [Google Scholar]
  • 234.Sanders VM, Munson AE. Kinetics of the enhancing effect produced by norepinephrine and terbutaline on the murine primary antibody response in vitro. J. Pharmacol. Exp. Ther. 1984;231:527–531. [PubMed] [Google Scholar]
  • 235.Sanders VM, Munson AE. Beta adrenoceptor mediation of the enhancing effect of norepinephrine on the murine primary antibody response in vitro. J. Pharmacol. Exp. Ther. 1984;230:183–192. [PubMed] [Google Scholar]
  • 236.Sanders VM, Powell-Oliver FE. Beta 2-adrenoceptor stimulation increases the number of antigen-specific precursor B lymphocytes that differentiate into IgM-secreting cells without affecting burst size. J. Immunol. 1992;148:1822–1828. [PubMed] [Google Scholar]
  • 237.Madden KS, Felten SY, Felten DL, Hardy CA, Livnat S. Sympathetic nervous system modulation of the immune system. II. Induction of lymphocyte proliferation and migration in vivo by chemical sympathectomy. J. Neuroimmunol. 1994;49:67–75. doi: 10.1016/0165-5728(94)90182-1. [DOI] [PubMed] [Google Scholar]
  • 238.Callahan TA, Moynihan JA. Contrasting pattern of cytokines in antigen- versus mitogen-stimulated splenocyte cultures from chemically denervated mice. Brain Behav. Immun. 2002;16:764–773. doi: 10.1016/s0889-1591(02)00029-6. [DOI] [PubMed] [Google Scholar]
  • 239.Felsner P, Hofer D, Rinner I, Porta S, Korsatko W, Schauenstein K. Adrenergic suppression of peripheral blood T cell reactivity in the rat is due to activation of peripheral alpha 2-receptors. J. Neuroimmunol. 1995;57:27–34. doi: 10.1016/0165-5728(94)00158-k. [DOI] [PubMed] [Google Scholar]
  • 240.Fuchs BA, McCall CO, Munson AE. Enhancement of the murine primary antibody response by phenylephrine in vitro. Drug Chem. Toxicol. 1991;14:67–82. doi: 10.3109/01480549109017869. [DOI] [PubMed] [Google Scholar]
  • 241.Heilig M, Irwin M, Grewal I, Sercarz E. Sympathetic regulation of T-helper cell function. Brain Behav. Immun. 1993;7:154–163. doi: 10.1006/brbi.1993.1017. [DOI] [PubMed] [Google Scholar]
  • 242.Brown R, Li Z, Vriend CY, Nirula R, Janz L, Falk J, Nance DM, Dyck DG, Greenberg AH. Suppression of splenic macrophage interleukin-1 secretion following intracerebroventricular injection of interleukin-1: Evidence for pituitary-adrenal and sympathetic control. Cell. Immunol. 1991;132:84–93. doi: 10.1016/0008-8749(91)90008-y. [DOI] [PubMed] [Google Scholar]
  • 243.Henricks PA, van Esch B, Nijkamp FP. Beta-agonists can depress oxidative metabolism of alveolar macrophages. Agents Actions. 1986;19:353–354. doi: 10.1007/BF01971251. [DOI] [PubMed] [Google Scholar]
  • 244.Henricks PA, Verhoef J, Nijkamp FP. Modulation of phagocytic cell function. Vet. Res. Commun. 1986;10:165–188. doi: 10.1007/BF02213979. [DOI] [PubMed] [Google Scholar]
  • 245.Schopf RE, Lemmel EM. Control of the production of oxygen intermediates of human polymorphonuclear leukocytes and monocytes by beta-adrenergic receptors. J. Immunopharmacol. 1983;5:203–216. doi: 10.3109/08923978309039106. [DOI] [PubMed] [Google Scholar]
  • 246.Fukushima T, Sekizawa K, Jin Y, Yamaya M, Sasaki H, Takishima T. Effects of beta-adrenergic receptor activation on alveolar macrophage cytoplasmic motility. Am. J. Physiol. 1993;265:L67–L72. doi: 10.1152/ajplung.1993.265.1.L67. [DOI] [PubMed] [Google Scholar]
  • 247.Conlon PD, Ogunbiyi PO, Black WD, Eyre P. Beta-adrenergic receptor function and oxygen radical production in bovine pulmonary alveolar macrophages. Can J Physiol Pharmacol. 1988;66:1538–1541. doi: 10.1139/y88-251. [DOI] [PubMed] [Google Scholar]
  • 248.Fuller RW, O'Malley G, Baker AJ, MacDermot J. Human alveolar macrophage activation: inhibition by forskolin but not beta-adrenoceptor stimulation or phosphodiesterase inhibition. Pulm. Pharmacol. 1988;1:101–106. doi: 10.1016/s0952-0600(88)80006-1. [DOI] [PubMed] [Google Scholar]
  • 249.Capelli A, Lusuardi M, Carli S, Zaccaria S, Trombetta N, Donner CF. In vitro effect of beta 2-agonists on bacterial killing and superoxide anion(O2−) release from alveolar macrophages of patients with chronic bronchitis. Chest. 1993;104:481–486. doi: 10.1378/chest.104.2.481. [DOI] [PubMed] [Google Scholar]
  • 250.Balter NJ, Schwartz SL. Accumulation of norepinephrine by macrophages and relationships to known uptake processes. J. Pharmacol. Exp. Ther. 1997;201:636–643. [PubMed] [Google Scholar]
  • 251.Issaad C, Ventura MA, Thomopoulos P. Biphasic regulation of macrophage attachment by activators of cyclic adenosine monophosphate-dependent kinase and protein kinase C. J. Cell. Physiol. 1989;140:317–322. doi: 10.1002/jcp.1041400218. [DOI] [PubMed] [Google Scholar]
  • 252.Petty HR, Berg KA. Combinative ligand-receptor interactions: epinephrine depresses RAW264 macrophage antibody-dependent phagocytosis in the absence and presence of met-enkephalin. J. Cell. Physiol. 1988;134:281–286. doi: 10.1002/jcp.1041340215. [DOI] [PubMed] [Google Scholar]
  • 253.Serio M, Potenza MA, Montagnani M, Mansi G, Mitolo-Chieppa D, Jirillo E. Beta-adrenoceptor responsiveness of splenic macrophages in normotensive and hypertensive rats. Immunopharmacol. Immunotoxicol. 1996;18:247–265. doi: 10.3109/08923979609052735. [DOI] [PubMed] [Google Scholar]
  • 254.Koff WC, Dunegan MA. Modulation of macrophage-mediated tumoricidal activity by neuropeptides and neurohormones. J. Immunol. 1985;135:350–354. [PubMed] [Google Scholar]
  • 255.Javierre MQ, Pinto LV, Lima AO, Sassine WA. Immunologic phagocytosis by macrophages: effect by stimulation of alpha adrenergic receptors. Rev. Bras. Pesqui. Med. Biol. 1975;8:271–274. [PubMed] [Google Scholar]
  • 256.Lappin D, Whaley K. Adrenergic receptors on monocytes modulate complement component synthesis. Clin. Exp. Immunol. 1982;47:606–612. [PMC free article] [PubMed] [Google Scholar]
  • 257.Spengler RN, Chensue SW, Giacherio DA, Blenk N, Kunkel SL. Endogenous norepinephrine regulates tumor necrosis factor-alpha production from macrophages in vitro. J. Immunol. 1994;152:3024–3031. [PubMed] [Google Scholar]
  • 258.Chelmicka-Schorr E, Kwasniewski MN, Czlonkowska A. Sympathetic nervous system modulates macrophage function. Int. J. Immunopharmacol. 1992;14:841–846. doi: 10.1016/0192-0561(92)90082-v. [DOI] [PubMed] [Google Scholar]
  • 259.Chelmicka-Schorr E, Kwasniewski MN, Czlonkowska A. Sympathetic nervous system and macrophage function. Ann. N.Y. Acad. Sci. 1992;650:40–45. doi: 10.1111/j.1749-6632.1992.tb49092.x. [DOI] [PubMed] [Google Scholar]
  • 260.Hellstrand K, Hermodsson S, Strannegard O. Evidence for a beta-adrenoceptor-mediated regulation of human natural killer cells. J. Immunol. 1985;134:4095–4099. [PubMed] [Google Scholar]
  • 261.Benschop RJ, Nijkamp FP, Ballieux RE, Heijnen CJ. The effects of beta-adrenoceptor stimulation on adhesion of human natural killer cells to cultured endothelium. Br. J. Pharmacol. 1994;113:1311–1316. doi: 10.1111/j.1476-5381.1994.tb17141.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Benschop RJ, Schedlowski M, Wienecke H, Jacobs R, Schmidt RE. Adrenergic control of natural killer cell circulation and adhesion. Brain Behav. Immun. 1997;11:321–332. doi: 10.1006/brbi.1997.0499. [DOI] [PubMed] [Google Scholar]
  • 263.Shakhar G, Ben-Eliyahu S. In vivo beta-adrenergic stimulation suppresses natural killer activity and compromises resistance to tumor metastasis in rats. J. Immunol. 1998;160:3251–3258. [PubMed] [Google Scholar]
  • 264.Bachen EA, Manuck SB, Cohen S, Muldoon MF, Raible R, Herbert TB, Rabin BS. Adrenergic blockade ameliorates cellular immune responses to mental stress in humans. Psychosom. Med. 1995;57:366–372. doi: 10.1097/00006842-199507000-00008. [DOI] [PubMed] [Google Scholar]
  • 265.Marsland AL, Manuck SB, Wood P, Rabin BS, Muldoon MF, Cohen S. Beta 2-adrenergic receptor density and cardiovascular response to mental stress. Physiol. Behav. 1995;57:1163–1167. doi: 10.1016/0031-9384(94)00378-i. [DOI] [PubMed] [Google Scholar]
  • 266.Shakhar G, Ben-Eliyahu S. In vivo beta-adrenergic stimulation suppresses natural killer activity and compromises resistance to tumor metastasis in rats. J. Immunol. 1998;160:3251–3258. [PubMed] [Google Scholar]
  • 267.Brenner GJ, Felten SY, Felten DL, Moynihan JA. Sympathetic nervous system modulation of tumor metastases and host defense mechanisms. J. Neuroimmunol. 1992;37:191–201. doi: 10.1016/0165-5728(92)90003-4. [DOI] [PubMed] [Google Scholar]
  • 268.Irwin M, Hauger RL, Brown M, Britton KT. CRF activates autonomic nervous system and reduces natural killer cytotoxicity. Am. J. Physiol. 1988;255:R744–R747. doi: 10.1152/ajpregu.1988.255.5.R744. [DOI] [PubMed] [Google Scholar]
  • 269.Irwin M, Hauger RL, Jones L, Provencio M, Britton KT. Sympathetic nervous system mediates central corticotropin-releasing factor induced suppression of natural killer cytotoxicity. J. Pharmacol. Exp. Ther. 1990;255:101–107. [PubMed] [Google Scholar]
  • 270.Mills PJ, Karnik RS, Dillon E. L-selectin expression affects T-cell circulation following isoproterenol infusion in humans. Brain Behav. Immun. 1997;11:333–342. doi: 10.1006/brbi.1997.0500. [DOI] [PubMed] [Google Scholar]
  • 271.Gonzalez-Ariki S, Husband AJ. The role of sympathetic innervation of the gut in regulating mucosal immune responses. Brain Behav. Immun. 1998;12:53–63. doi: 10.1006/brbi.1997.0509. [DOI] [PubMed] [Google Scholar]
  • 272.Carlson SL, Fox S, Abell KM. Catecholamine modulation of lymphocyte homing to lymphoid tissues. Brain Behav. Immun. 1997;11:307–320. doi: 10.1006/brbi.1997.0501. [DOI] [PubMed] [Google Scholar]
  • 273.Ernstrom U, Sandberg G. Effects of adrenergic alpha- and beta-receptor stimulation on the release of lymphocytes and granulocytes from the spleen. Scand. J. Haematol. 1973;11:275–286. doi: 10.1111/j.1600-0609.1973.tb00130.x. [DOI] [PubMed] [Google Scholar]
  • 274.Rogausch H, del Rey A, Oertel J, Besedovsky HO. Norepinephrine stimulates lymphoid cell mobilization from the perfused rat spleen via beta-adrenergic receptors. Am. J. Physiol. 1999;276:R724–R730. doi: 10.1152/ajpregu.1999.276.3.R724. [DOI] [PubMed] [Google Scholar]
  • 275.Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes. Nature (Lond) 1996;383:787–793. doi: 10.1038/383787a0. [DOI] [PubMed] [Google Scholar]
  • 276.Fearon DT, Locksley RM. The instructive role of innate immunity in the acquired immune response. Science (Wash DC) 1996;272:50–53. doi: 10.1126/science.272.5258.50. [DOI] [PubMed] [Google Scholar]
  • 277.Mosmann TR, Sad S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol. Today. 1996;17:138–146. doi: 10.1016/0167-5699(96)80606-2. [DOI] [PubMed] [Google Scholar]
  • 278.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. [PubMed] [Google Scholar]
  • 279.Lerner BH. Can stress cause disease? Revisiting the tuberculosis research of Thomas Holmes 1949–1961. Ann. Intern. Med. 1996;124:673–680. doi: 10.7326/0003-4819-124-7-199604010-00008. [DOI] [PubMed] [Google Scholar]
  • 280.Altare F, Durandy A, Lammas D, Emile JF, Lamhamedi S, Le Deist F, Drysdale P, Jouanguy E, Doffinger R, Bernaudin F, Jeppsson O, Gollob JA, Meinl E, Segal AW, Fischer A, Kumararatne D, Casanova JL. Impairment of mycobacterial immunity in human interleukin-12 receptor deficiency. Science (Wash DC) 1998;280:1432–1435. doi: 10.1126/science.280.5368.1432. [DOI] [PubMed] [Google Scholar]
  • 281.Elenkov IJ, Papanicolaou DA, Wilder RL, Chrousos GP. Modulatory effects of glucocorticoids and catecholamines on human interleukin-12 and interleukin-10 production: clinical implications. Proc. Assoc. Am. Physicians. 1996;108:374–381. [PubMed] [Google Scholar]
  • 282.Elenkov IJ, Chrousos GP. Stress hormones, Th1/Th2 patterns, pro/antiinflammatory cytokines and susceptibility to disease. Trends Endocrinol. Metab. 1999;10:359–368. doi: 10.1016/s1043-2760(99)00188-5. [DOI] [PubMed] [Google Scholar]
  • 283.Katoh K, Nomura M, Nakaya Y, Iga A, Nada T, Hiasa A, Ochi Y, Kawaguchi R, Uemura N, Honda H, Shimizu I, Ito S. Autonomic nervous activity before and after eradication of Helicobacter pylori in patients with chronic duodenal ulcer. Aliment Pharmacol. Ther. 2002;16(Suppl. 2):180–186. doi: 10.1046/j.1365-2036.16.s2.27.x. [DOI] [PubMed] [Google Scholar]
  • 284.Levenstein S. Stress and peptic ulcer: Life beyond Helicobacter. Br. Med. J. 1998;316:538–541. doi: 10.1136/bmj.316.7130.538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Levenstein S, Ackerman S, Kiecolt-Glaser JK, Dubois A. Stress and peptic ulcer disease. J.A.M.A. 1999;281:10–11. doi: 10.1001/jama.281.1.10. [DOI] [PubMed] [Google Scholar]
  • 286.Elenkov IJ, Webster E, Papanicolaou DA, Fleisher TA, Chrousos GP, Wilder RL. Histamine potently suppresses human IL-12 and stimulates IL-10 production via H2 receptors. J. Immunol. 1998;161:2586–2593. [PubMed] [Google Scholar]
  • 287.Haraguchi S, Good RA, Day NK. Immunosuppressive retroviral peptides: cAMP and cytokine patterns. Immunol. Today. 1995;16:595–603. doi: 10.1016/0167-5699(95)80083-2. [DOI] [PubMed] [Google Scholar]
  • 288.Cole SW, Korin YD, Fahey JL, Zack JA. Norepinephrine accelerates HIV replication via protein kinase A-dependent effects on cytokine production. J. Immunol. 1998;161:610–616. [PubMed] [Google Scholar]
  • 289.Haraguchi S, Good RA, James-Yarish M, Cianciolo GJ, Day NK. Differential modulation of Th1- and Th2-related cytokine mRNA expression by a synthetic peptide homologous to a conserved domain within retroviral envelope protein. Proc. Natl. Acad. Sci. U.S.A. 1995;92:3611–3615. doi: 10.1073/pnas.92.8.3611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Haraguchi S, Good RA, James-Yarish M, Cianciolo GJ, Day NK. Induction of intracellular cAMP by a synthetic retroviral envelope peptide: A possible mechanism of immunopathogenesis in retroviral infections. Proc. Natl. Acad. Sci. U.S.A. 1995;92:5568–5571. doi: 10.1073/pnas.92.12.5568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Kelley SP, Moynihan JA, Stevens SY, Grota LJ, Felten DL. Chemical sympathectomy has no effect on the severity of murine AIDS: murine AIDS alone depletes norepinephrine levels in infected spleen. Brain Behav. Immun. 2002;16:118–139. doi: 10.1006/brbi.2001.0627. [DOI] [PubMed] [Google Scholar]
  • 292.Kelley SP, Moynihan JA, Stevens SY, Grota LJ, Felten DL. Sympathetic nerve destruction in spleen in murine AIDS. Brain Behav. Immun. 2003;17:94–109. doi: 10.1016/s0889-1591(02)00101-0. [DOI] [PubMed] [Google Scholar]
  • 293.Pastores SM, Hasko G, Vizi ES, Kvetan V. Cytokine production and its manipulation by vasoactive drugs. New Horiz. 1996;4:252–264. [PubMed] [Google Scholar]
  • 294.Eigler A, Sinha B, Hartmann G, Endres S. Taming TNF: Strategies to restrain this proinflammatory cytokine. Immunol. Today. 1997;18:487–492. doi: 10.1016/s0167-5699(97)01118-3. [DOI] [PubMed] [Google Scholar]
  • 295.Palsson J, Ricksten SE, Delle M, Lundin S. Changes in renal sympathetic nerve activity during experimental septic and endotoxin shock in conscious rats. Circ. Shock. 1988;24:133–141. [PubMed] [Google Scholar]
  • 296.Jones SB, Kovarik MF, Romano FD. Cardiac and splenic norepinephrine turnover during septic peritonitis. Am. J. Physiol. 1986;250:R892–R897. doi: 10.1152/ajpregu.1986.250.5.R892. [DOI] [PubMed] [Google Scholar]
  • 297.Leinhardt DJ, Arnold J, Shipley KA, Mughal MM, Little RA, Irving MH. Plasma NE concentrations do not accurately reflect sympathetic nervous system activity in human sepsis. Am. J. Physiol. 1993;265:E284–E288. doi: 10.1152/ajpendo.1993.265.2.E284. [DOI] [PubMed] [Google Scholar]
  • 298.Zhou M, Hank Simms H, Wang P. Increased gut-derived norepinephrine release in sepsis: upregulation of intestinal tyrosine hydroxylase. Biochim. Biophys. Acta. 2004;1689:212–218. doi: 10.1016/j.bbadis.2004.03.008. [DOI] [PubMed] [Google Scholar]
  • 299.Miura T, Kudo T, Matsuki A, Sekikawa K, Tagawa Y, Iwakura Y, Nakane A. Effect of 6-hydroxydopamine on host resistance against Listeria monocytogenes infection. Infect. Immun. 2001;69:7234–7241. doi: 10.1128/IAI.69.12.7234-7241.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Straub RH, Linde HJ, Männel DN, Schölmerich J, Falk W. A bacteria-induced switch of sympathetic effector mechanisms augments local inhibition of TNF-α and IL-6 secretion in the spleen. F.A.S.E.B. J. 2000;14:1380–1388. doi: 10.1096/fj.14.10.1380. [DOI] [PubMed] [Google Scholar]
  • 301.Rice PA, Boehm GW, Moynihan JA, Bellinger DL, Stevens SY. Chemical sympathectomy increases the innate immune response and decreases the specific immune response in the spleen to infection with Listeria monocytogenes. J. Neuroimmunol. 2001;114:19–27. doi: 10.1016/s0165-5728(00)00421-5. [DOI] [PubMed] [Google Scholar]
  • 302.Straub RH, Pongratz G, Weidler C, Linde HJ, Kirschning CJ, Gluck 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. [DOI] [PubMed] [Google Scholar]
  • 303.Cao L, Hudson CA, Lawrence DA. Acute cold/restraint stress inhibits host resistance to Listeria monocytogenes via β1-adrenergic receptors. Brain Behav. Immun. 2003;17:121–133. doi: 10.1016/s0889-1591(03)00026-6. [DOI] [PubMed] [Google Scholar]
  • 304.Green BT, Lyte M, Chen C, Xie Y, Casey MA, Kulkarni-Narla A, Vulchanova L, Brown DR. Adrenergic modulation of Escherichia coli O157:H7 adherence to the colonic mucosa. Am. J. Physiol. Gastrointest. Liver Physiol. 2004;287:G1238–G1246. doi: 10.1152/ajpgi.00471.2003. [DOI] [PubMed] [Google Scholar]
  • 305.Takahashi H, Kobayashi M, Tsuda YDN, Herndon F, Suzuki Contribution of the sympathetic nervous system on the burn-associated impairment of CCL3 production. Cytokine. 2005;29:208–214. doi: 10.1016/j.cyto.2004.10.014. [DOI] [PubMed] [Google Scholar]
  • 306.Redwine L, Snow S, Mills P, Irwin M. Acute psychological stress: effects on chemotaxis and cellular adhesion molecule expression. Psychosom. Med. 2003;65:598–603. doi: 10.1097/01.psy.0000079377.86193.a8. [DOI] [PubMed] [Google Scholar]
  • 307.van der Poll T, Coyle SM, Barbosa K, Braxton CC, Lowry SF. Epinephrine inhibits tumor necrosis factor-alpha and potentiates interleukin 10 production during human endotoxemia. J. Clin. Invest. 1996;97:713–719. doi: 10.1172/JCI118469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Szabo C, Hasko G, Zingarelli B, Nemeth 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Hasko G, Nemeth ZH, Szabo C, Zsilla G, Salzman AL, Vizi ES. Isoproterenol inhibits IL-10, TNF-alpha, and nitric oxide production in RAW 264.7 macrophages. Brain Res. Bull. 1998;45:183–187. doi: 10.1016/s0361-9230(97)00337-7. [DOI] [PubMed] [Google Scholar]
  • 310.Elenkov IJ, Hasko G, Kovacs KJ, Vizi ES. Modulation of lipopolysaccharide-induced tumor necrosis factor-alpha production by selective alpha- and beta-adrenergic drugs in mice. J. Neuroimmunol. 1995;61:123–131. doi: 10.1016/0165-5728(95)00080-l. [DOI] [PubMed] [Google Scholar]
  • 311.Chou RC, Stinson MW, Noble BK, Spengler RN. Beta-adrenergic receptor regulation of macrophage-derived tumor necrosis factor-alpha production from rats with experimental arthritis. J. Neuroimmunol. 1996;67:7–16. doi: 10.1016/0165-5728(96)00023-9. [DOI] [PubMed] [Google Scholar]
  • 312.Hasko G, Szabo C, Nemeth ZH, Salzman AL, Vizi ES. Stimulation of beta-adrenoceptors inhibits endotoxin-induced IL-12 production in normal and IL-10 deficient mice. J. Neuroimmunol. 1998;88:57–61. doi: 10.1016/s0165-5728(98)00073-3. [DOI] [PubMed] [Google Scholar]
  • 313.Ignatowski TA, Spengler RN. Tumor necrosis factor-alpha: Presynaptic sensitivity is modified after antidepressant drug administration. Brain Res. 1994;665:293–299. doi: 10.1016/0006-8993(94)91350-1. [DOI] [PubMed] [Google Scholar]
  • 314.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. [PubMed] [Google Scholar]
  • 315.Gerard C, Bruyns C, Marchant A, Abramowicz D, Vandenabeele P, Delvaux A, Fiers W, Goldman M, Velu T. Interleukin 10 reduces the release of tumor necrosis factor and prevents lethality in experimental endotoxemia. J. Exp. Med. 1993;177:547–550. doi: 10.1084/jem.177.2.547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Suberville S, Bellocq A, Fouqueray B, Philippe C, Lantz O, Perez J, Baud L. Regulation of interleukin-10 production by beta-adrenergic agonists. Eur. J. Immunol. 1996;26:2601–2605. doi: 10.1002/eji.1830261110. [DOI] [PubMed] [Google Scholar]
  • 317.Panina-Bordignon P, Mazzeo D, Lucia PD, D'Ambrosio D, Lang R, Fabbri L, Self C, Sinigaglia F. Beta2-agonists prevent Th1 development by selective inhibition of interleukin 12. J. Clin. Invest. 1997;100:1513–1519. doi: 10.1172/JCI119674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Fessler HE, Otterbein L, Chung HS, Choi AM. Alpha-2 adrenoceptor blockade protects rats against lipopolysaccharide. Am. J. Respir. Crit. Care Med. 1996;154:1689–1693. doi: 10.1164/ajrccm.154.6.8970356. [DOI] [PubMed] [Google Scholar]
  • 319.Hayes MA, Timmins AC, Yau EH, Palazzo M, Hinds CJ, Watson D. Elevation of systemic oxygen delivery in the treatment of critically ill patients. N. Engl. J. Med. 1994;330:1717–1722. doi: 10.1056/NEJM199406163302404. [DOI] [PubMed] [Google Scholar]
  • 320.Hensler T, Heidecke C, Hecker H, Heeg K, Bartels H, Zantl N, Wagner H, Siewert J, Holzmann B. Increased susceptibility to postoperative sepsis in patients with impaired monocyte IL-12 production. J. Immunol. 1998;161:2655–2659. [PubMed] [Google Scholar]
  • 321.Lyte M. The role of catecholamines in gram-negative sepsis. Med. Hypotheses. 1992;37:255–258. doi: 10.1016/0306-9877(92)90197-k. [DOI] [PubMed] [Google Scholar]
  • 322.Chiolero R, Berger M. Endocrine response to brain injury. New Horiz. 1994;2:432–442. [PubMed] [Google Scholar]
  • 323.Jarek MJ, Legare EJ, McDermott MT, Merenich JA, Kollef MH. Endocrine profiles for outcome prediction from the intensive care unit. Crit. Care Med. 1993;21:543–550. doi: 10.1097/00003246-199304000-00015. [DOI] [PubMed] [Google Scholar]
  • 324.Rothwell PM, Lawler PG. Prediction of outcome in intensive care patients using endocrine parameters. Crit. Care Med. 1995;23:78–83. doi: 10.1097/00003246-199501000-00015. [DOI] [PubMed] [Google Scholar]
  • 325.Rothwell NJ, Luheshi G, Toulmond S. Cytokines and their receptors in the central nervous system: Physiology, pharmacology, and pathology. Pharmacol. Ther. 1996;69:85–95. doi: 10.1016/0163-7258(95)02033-0. [DOI] [PubMed] [Google Scholar]
  • 326.O'Sullivan ST, Lederer JA, Horgan AF, Chin DH, Mannick JA, Rodrick ML. Major injury leads to predominance of the T helper-2 lymphocyte phenotype and diminished interleukin-12 production associated with decreased resistance to infection. Ann. Surg. 1995;222:482–490. doi: 10.1097/00000658-199522240-00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Fabian TC, Croce MA, Fabian MJ, Trenthem LL, Yockey JM, Boscarino R, Proctor KG. Reduced tumor necrosis factor production in endotoxin-spiked whole blood after trauma: Experimental results and clinical correlation. Surgery. 1995;118:63–72. doi: 10.1016/s0039-6060(05)80011-x. [DOI] [PubMed] [Google Scholar]
  • 328.Brune IB, Wilke W, Hensler T, Holzmann B, Jr, Siewert Downregulation of T helper type 1 immune response and altered pro-inflammatory and anti-inflammatory T cell cytokine balance following conventional but not laparoscopic surgery. Am. J. Surg. 1999;177:55–60. doi: 10.1016/s0002-9610(98)00299-2. [DOI] [PubMed] [Google Scholar]
  • 329.Woiciechowsky C, Asadullah K, Nestler D, Eberhardt B, Platzer C, Schoning B, Glockner F, Lanksch WR, Volk HD, Docke WD. Sympathetic activation triggers systemic interleukin-10 release in immunodepression induced by brain injury. Nat. Med. 1998;4:808–813. doi: 10.1038/nm0798-808. [DOI] [PubMed] [Google Scholar]
  • 330.Prass K, Meisel C, Hoflich 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;98:725–736. doi: 10.1084/jem.20021098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Plata-Salaman CR. Brain injury and immunosuppression. Nat. Med. 1998;4:768–769. doi: 10.1038/nm0798-768. [DOI] [PubMed] [Google Scholar]
  • 332.Wilder RL. Neuroendocrine-immune system interactions and autoimmunity. Annu. Rev. Immunol. 1995;13:307–338. doi: 10.1146/annurev.iy.13.040195.001515. [DOI] [PubMed] [Google Scholar]
  • 333.Elenkov IJ, Hoffman J, Wilder RL. Does differential neuroendocrine control of cytokine production govern the expression of autoimmune diseases in pregnancy and the postpartum period? Mol. Med. Today. 1997;3:379–383. doi: 10.1016/s1357-4310(97)01089-7. [DOI] [PubMed] [Google Scholar]
  • 334.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. [DOI] [PubMed] [Google Scholar]
  • 335.Segal BM, Dwyer BK, Shevach EM. An interleukin (IL)-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease. J. Exp. Med. 1998;187:537–546. doi: 10.1084/jem.187.4.537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Maini RN, Elliott MJ, Charles PJ, Feldmann M. Immunological intervention reveals reciprocal roles for tumor necrosis factor-alpha and interleukin-10 in rheumatoid arthritis and systemic lupus erythematosus. Springer Semin. Immunopathol. 1994;16:327–336. doi: 10.1007/BF00197526. [DOI] [PubMed] [Google Scholar]
  • 337.Horwitz DA, Gray JD, Behrendsen SC, Kubin M, Rengaraju M, Ohtsuka K, Trinchieri G. Decreased production of interleukin-12 and other Th1-type cytokines in patients with recent-onset systemic lupus erythematosus. Arthritis Rheum. 1998;41:838–844. doi: 10.1002/1529-0131(199805)41:5<838::AID-ART10>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
  • 338.Arnason BG, Noronha AB, Reder AT. Immunoregulation in rapidly progressive multiple sclerosis. Ann. N.Y. Acad. Sci. 1988;540:4–12. doi: 10.1111/j.1749-6632.1988.tb27046.x. [DOI] [PubMed] [Google Scholar]
  • 339.Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N. Engl. J. Med. 1995;332:1351–1362. doi: 10.1056/NEJM199505183322008. [DOI] [PubMed] [Google Scholar]
  • 340.Rogers MP, Fozdar M. Psychoneuroimmunology of autoimmune disorders. Adv. Neuroimmunol. 1996;6:169–177. doi: 10.1016/0960-5428(96)00015-0. [DOI] [PubMed] [Google Scholar]
  • 341.Cohen WR, Galen LH, Vega-Rich M, Young JB. Cardiac sympathetic activity during rat pregnancy. Metabolism. 1988;37:771–777. doi: 10.1016/0026-0495(88)90013-3. [DOI] [PubMed] [Google Scholar]
  • 342.Magiakou MA, Mastorakos G, Rabin D, Margioris AN, Dubbert B, Calogero AE, Tsigos C, Munson PJ, Chrousos GP. The maternal hypothalamic-pituitary-adrenal axis in the third trimester of human pregnancy. Clin. Endocrinol. (Oxf) 1996;44:419–428. doi: 10.1046/j.1365-2265.1996.683505.x. [DOI] [PubMed] [Google Scholar]
  • 343.Lin H, Mosmann TR, Guilbert L, Tuntipopipat S, Wegmann TG. Synthesis of T helper 2-type cytokines at the maternal-fetal interface. J. Immunol. 1993;151:4562–4573. [PubMed] [Google Scholar]
  • 344.Sacks GP, Studena K, Sargent IL, Redman CWG. Normal pregnancy and preeclampsia both produce inflammatory changes in peripheral blood lekocytes akin to those of sepsis. Am. J. Obstet. Gynecol. 1998;179:80–86. doi: 10.1016/s0002-9378(98)70254-6. [DOI] [PubMed] [Google Scholar]
  • 345.Sacks G, Sargent I, Redman C. An innate view of human pregnancy. Immunol. Today. 1999;20:114–118. doi: 10.1016/s0167-5699(98)01393-0. [DOI] [PubMed] [Google Scholar]
  • 346.Karaszewski JW, Reder AT, Maselli R, Brown M, Arnason BG. Sympathetic skin responses are decreased and lymphocyte beta-adrenergic receptors are increased in progressive multiple sclerosis. Ann. Neurol. 1990;27:366–372. doi: 10.1002/ana.410270404. [DOI] [PubMed] [Google Scholar]
  • 347.Karaszewski JW, Reder AT, Anlar B, Arnason GW. Increased high affinity beta-adrenergic receptor densities and cyclic AMP responses of CD8 cells in multiple sclerosis. J. Neuroimmunol. 1993;43:1–7. doi: 10.1016/0165-5728(93)90068-a. [DOI] [PubMed] [Google Scholar]
  • 348.Mackenzie FJ, Leonard JP, Cuzner ML. Changes in lymphocyte beta-adrenergic receptor density and noradrenaline content of the spleen are early indicators of immune reactivity in acute experimental allergic encephalomyelitis in the Lewis rat. J. Neuroimmunol. 1989;23:93–100. doi: 10.1016/0165-5728(89)90027-1. [DOI] [PubMed] [Google Scholar]
  • 349.Zoukos Y, Leonard JP, Thomaides T, Thompson AJ, Cuzner ML. beta-Adrenergic receptor density and function of peripheral blood mononuclear cells are increased in multiple sclerosis: A regulatory role for cortisol and interleukin-1. Ann. Neurol. 1992;31:657–662. doi: 10.1002/ana.410310614. [DOI] [PubMed] [Google Scholar]
  • 350.Leonard JP, Mackenzie FJ, Patel HA, Cuzner ML. Hypothalamic noradrenergic pathways exert an influence on neuroendocrine and clinical status in experimental autoimmune encephalomyelitis. Brain Behav. Immun. 1991;5:328–338. doi: 10.1016/0889-1591(91)90028-9. [DOI] [PubMed] [Google Scholar]
  • 351.Chelmicka-Schorr E, Kwasniewski MN, Thomas BE, Arnason BG. The beta-adrenergic agonist isoproterenol suppresses experimental allergic encephalomyelitis in Lewis rats. J. Neuroimmunol. 1989;25:203–207. doi: 10.1016/0165-5728(89)90138-0. [DOI] [PubMed] [Google Scholar]
  • 352.Wiegmann K, Muthyala S, Kim DH, Arnason BG, Chelmicka-Schorr E. Beta-adrenergic agonists suppress chronic/relapsing experimental allergic encephalomyelitis (CREAE) in Lewis rats. J. Neuroimmunol. 1995;56:201–206. doi: 10.1016/0165-5728(94)00153-f. [DOI] [PubMed] [Google Scholar]
  • 353.Kuis W, de Jong-de Vos van Steenwijk C, Sinnema G, Kavelaars A, Prakken B, Helders PJ, Heijnen CJ. The autonomic nervous system and the immune system in juvenile rheumatoid arthritis. Brain Behav. Immun. 1996;10:387–398. doi: 10.1006/brbi.1996.0034. [DOI] [PubMed] [Google Scholar]
  • 354.Leden I, Eriksson A, Lilja B, Sturfelt G, Sundkvist G. Autonomic nerve function in rheumatoid arthritis of varying severity. Scand J. Rheumatol. 1983;12:166–170. doi: 10.3109/03009748309102905. [DOI] [PubMed] [Google Scholar]
  • 355.Perry F, Heller PH, Kamiya J, Levine JD. Altered autonomic function in patients with arthritis or with chronic myofascial pain. Pain. 1989;39:77–84. doi: 10.1016/0304-3959(89)90177-2. [DOI] [PubMed] [Google Scholar]
  • 356.Kalliomaki JL, Saarimaa HA, Toivanen P. Axon reflex sweating in rheumatoid arthritis. Ann. Rheum. Dis. 1963;22:46–49. doi: 10.1136/ard.22.1.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Levine JD, Collier DH, Basbaum AI, Moskowitz MA, Helms CA. Hypothesis: the nervous system may contribute to the pathophysiology of rheumatoid arthritis. J. Rheumatol. 1985;12:406–411. [PubMed] [Google Scholar]
  • 358.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. doi: 10.1523/JNEUROSCI.06-12-03423.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.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. [PubMed] [Google Scholar]
  • 360.Levine JD, Goetzl E, Basbaum A. Contribution of the nervous system to the pathophysiology of rheumatoid arthritis and other polyarthritides. Rheum. Dis. Clin. North. Am. 1987;13:369–383. [PubMed] [Google Scholar]
  • 361.Colpaert FC, van den Hoogen RH. Time course of the ventilatory response to adjuvant arthritis in the rat. Life Sci. 1983;33:1065–1073. doi: 10.1016/0024-3205(83)90662-8. [DOI] [PubMed] [Google Scholar]
  • 362.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. [DOI] [PubMed] [Google Scholar]
  • 363.Levine JD, Coderre TJ, Helms C, Basbaum AI. Beta 2-adrenergic mechanisms in experimental arthritis. Proc. Natl. Acad. Sci. U.S.A. 1988;85:4553–4556. doi: 10.1073/pnas.85.12.4553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Lorton D, Bellinger DL, Duclos M, Felten SY, Felten DL. Application of 6-hydroxydopamine into the fatpads surrounding the draining lymph nodes exacerbates adjuvant-induced arthritis. J. Neuroimmunol. 1996;64:103–113. doi: 10.1016/0165-5728(95)00150-6. [DOI] [PubMed] [Google Scholar]
  • 365.Lubahn CL, Schaller JA, Bellinger DL, Sweeney S, Lorton D. The importance of timing of adrenergic drug delivery in relation to the induction and onset of adjuvant-induced arthritis. Brain Behav. Immun. 2004;18:563–571. doi: 10.1016/j.bbi.2004.02.004. [DOI] [PubMed] [Google Scholar]
  • 366.Coderre TJ, Basbaum AI, Helms C, Levine JD. High-dose epinephrine acts at alpha 2-adrenoceptors to suppress experimental arthritis. Brain Res. 1991;544:325–328. doi: 10.1016/0006-8993(91)90073-5. [DOI] [PubMed] [Google Scholar]
  • 367.Malfait A-M, Malik AS, Marinova-Mutafchieva L, Butler DM, Maini RN, Feldmann M. The β2-adrenergic agonist salbutamol is a potent suppressor of established collagen-induced arthritis: Mechanisms of action. J. Immunol. 1999;162:6278–6283. [PubMed] [Google Scholar]
  • 368.Baerwald C, Graefe C, Muhl C, Von Wichert P, Krause A. Beta 2-adrenergic receptors on peripheral blood mononuclear cells in patients with rheumatic diseases. Eur. J. Clin. Invest. 1992;22(Suppl 1):42–46. [PubMed] [Google Scholar]
  • 369.Baerwald C, Graefe C, von Wichert P, Krause A. Decreased density of beta-adrenergic receptors on peripheral blood mononuclear cells in patients with rheumatoid arthritis. J. Rheumatol. 1992;19:204–210. [PubMed] [Google Scholar]
  • 370.Baerwald CG, Laufenberg M, Specht T, von Wichert P, Burmester GR, Krause A. Impaired sympathetic influence on the immune response in patients with rheumatoid arthritis due to lymphocyte subset-specific modulation of beta 2-adrenergic receptors. Br. J. Rheumatol. 1997;36:1262–1269. doi: 10.1093/rheumatology/36.12.1262. [DOI] [PubMed] [Google Scholar]
  • 371.Lombardi MS, Kavelaars A, Schedlowski M, Bijlsma JWJ, Okihara KO, van der Pol M, Ochsmann S, Pawlak C, Schmidt RE, Heijnen CJ. Decreased expression and activity of G-protein-coupled receptors in peripheral blood mononuclear cells of patients with rheumatoid arthritis. F.A.S.E.B. J. 1999;13:715–725. doi: 10.1096/fasebj.13.6.715. [DOI] [PubMed] [Google Scholar]
  • 372.Kuis W, Heijnen CJ. Rheumatoid arthritis and juvenile chronic arthritis: the role of the neuroendocrine system. Clin. Exp. Rheumatol. 1994;10:S29–S34. 12 Suppl. [PubMed] [Google Scholar]
  • 373.Heijnen CJ, Rouppe van der Voort C, Wulffraat N, van der Net J, Kuis W, Kavelaars A. Functional alpha 1-adrenergic receptors on leukocytes of patients with polyarticular juvenile rheumatoid arthritis. J. Neuroimmunol. 1996;71:223–226. doi: 10.1016/s0165-5728(96)00125-7. [DOI] [PubMed] [Google Scholar]
  • 374.Lubahn C, Schaller J, Lorton D. Chronic treatment with adrenergic agents attenuates the inflammation and bone destruction associated with adjuvant arthritis in the Lewis rat. Brain Behav. Immun. 2001;15:167. [Google Scholar]
  • 375.Rouppe van der Voort C, Kavelaars A, van de Pol M, Heijnen CJ. Neuroendocrine mediators up-regulate alpha1b- and alpha1d-adrenergic receptor subtypes in human monocytes. J. Neuroimmunol. 1999;95:165–173. doi: 10.1016/s0165-5728(99)00011-9. [DOI] [PubMed] [Google Scholar]
  • 376.van der Poll TJ, Jansen E, Endert HP, Sauerwein HP, van Deventer SJH. Noradrenaline inhibits lipopolysaccharide-induced tumor necrosis factor and interleukin 6 production in human whole blood. Infect. Immun. 1994;62:2946–2050. doi: 10.1128/iai.62.5.2046-2050.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Monastra G, Secchi EF. Beta-adrenergic receptors mediate in vivo the adrenaline inhibition of lipopolysaccharide-induced tumor necrosis factor release. Immunol. Lett. 1993;38:127–130. doi: 10.1016/0165-2478(93)90177-4. [DOI] [PubMed] [Google Scholar]
  • 378.Ignatowski TA, Spengler RN. Regulation of macrophage-derived tumor necrosis factor production by modification of adrenergic receptor sensitivity. J. Neuroimmunol. 1995;61:61–70. doi: 10.1016/0165-5728(95)00074-c. [DOI] [PubMed] [Google Scholar]
  • 379.Lorton D, Lubahn C, Lindquist CA, Schaller J, Washington C, Bellinger DL. Changes in the density and distribution of sympathetic nerves in spleens from Lewis rats with adjuvant-induced arthritis suggest that an injury and sprouting response occurs. J. Comp. Neurol. 2005;489:260–273. doi: 10.1002/cne.20640. [DOI] [PubMed] [Google Scholar]
  • 380.Breneman SM, Moynihan JA, Grota LJ, Felten DL, Felten SY. Splenic norepinephrine is decreased in MRL-lpr/lpr mice. Brain Behav. Immun. 1993;7:135–143. doi: 10.1006/brbi.1993.1015. [DOI] [PubMed] [Google Scholar]
  • 381.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.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. F.A.S.E.B. J. 2000;14:2097–2107. doi: 10.1096/fj.99-1082com. [DOI] [PubMed] [Google Scholar]
  • 383.Harle P, Straub RH. Neuroendocrine-immune aspects of accelerated aging in rheumatoid arthritis. Curr. Rheumatol. Rep. 2005;7:389–394. doi: 10.1007/s11926-005-0027-6. [DOI] [PubMed] [Google Scholar]
  • 384.Green PG, Miao FJ, Strausbaugh H, Heller P, Janig W, Levine JD. Endocrine and vagal controls of sympathetically dependent neurogenic inflammation. Ann. N.Y. Acad. Sci. 1998;840:282–288. doi: 10.1111/j.1749-6632.1998.tb09568.x. [DOI] [PubMed] [Google Scholar]
  • 385.Miao FJ, Janig W, Levine JD. Role of sympathetic postganglionic neurons in synovial plasma extravasation induced by bradykinin. J. Neurophysiol. 1996;75:715–724. doi: 10.1152/jn.1996.75.2.715. [DOI] [PubMed] [Google Scholar]
  • 386.Clark JA, Inui TS, Silliman RA, Bokhour BG, Krasnow SH, Robinson RA, Spaulding M, Talcott JA. Patient’s perception of quality of life after treatment for early prostate cancer. J. Clin. Oncol. 2003;21:3777–3784. doi: 10.1200/JCO.2003.02.115. [DOI] [PubMed] [Google Scholar]
  • 387.Visser A, van Andel G, Willems P, Voogt E, Dijkstra A, Rovers P, Goodkin K, Kurth KH. Changes in health-related quality of life of men with prostate cancer 3 months after diagnosis: the role of psychosocial factors and comparison with benign prostate hyperplasia patients. Patient Educ. Couns. 2003;49:225–232. doi: 10.1016/s0738-3991(02)00221-5. [DOI] [PubMed] [Google Scholar]
  • 388.Cohen S, Rabin BS. Psychologic stress, immunity, and cancer: editorial; comment. J. Natl. Cancer Inst. 1998;90:3–4. doi: 10.1093/jnci/90.1.3. [DOI] [PubMed] [Google Scholar]
  • 389.Turner-Cobb JM, Sephton SE, Spiegel D. Psychosocial effects on immune function and disease progression in cancer: Human studies. In: Ader R, Cohen N, Felten DL, editors. Psychoneuroimmunology. 3rd edition. San Diego: Academic Press; 2001. pp. 565–582. [Google Scholar]
  • 390.Herberman R. Natural killer cells: Characteristic and possible role in resistance against tumor growth. In: Reif AE, Mitchel MS, editors. Immunity to cancer. San Diego: Academic Press; 1985. [Google Scholar]
  • 391.Trinchieri G. Biology of natural killer cells. Adv. Immunol. 1989;47:187–376. doi: 10.1016/S0065-2776(08)60664-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Whiteside TL, Herberman RB. Role of human natural killer cells in immune surveillance of cancer. Curr. Opin. Immunol. 1994;7:704–710. doi: 10.1016/0952-7915(95)80080-8. [DOI] [PubMed] [Google Scholar]
  • 393.Karre K, Ljunggren HG, Piontek G, Kiesslinger R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defense strategy. Nature. 1986;51:675–678. doi: 10.1038/319675a0. [DOI] [PubMed] [Google Scholar]
  • 394.Ljunggren HG, Karre K. In search of the “missing self”: MHC molecules and NK cell recognition. Immunol. Today. 1990;11:237–244. doi: 10.1016/0167-5699(90)90097-s. [DOI] [PubMed] [Google Scholar]
  • 395.Raulet DH, Correa I, Corral L, Dorfman J, Wu MF. Inhibitory effects of class I molecules on murine NK cells: Speculations on function, specificity and self-tolerance. Sem. Immunol. 1995;7:103–107. doi: 10.1006/smim.1995.0014. [DOI] [PubMed] [Google Scholar]
  • 396.Sanda MG, Restifo NP, Walsh JC, Kawakami Y, Nelson WG, Pardoll DM, Simons JW. Molecular characterization of defective antigen processing in human prostate cancer. J Natl. Cancer Inst. 1995;87:280–285. doi: 10.1093/jnci/87.4.280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Nasu Y, Bangma CH, Hull GW, Lee HM, Hu J, Wang J, McCurdy MA, Shimura S, Yang G, Timme TL, Thompson TC. Adenovirus-mediated interleukin-12 gene therapy for prostate cancer: Suppression of orthotopic tumor growth and pre-established lung metastases in an orthotopic model. Gene Ther. 1999;6:338–349. doi: 10.1038/sj.gt.3300834. [DOI] [PubMed] [Google Scholar]
  • 398.Lee H-M, Timme TL, Thompson TC. Resistance to lysis by cytotoxic T cells: A dominant effect in metastatic mouse prostate cancer cells. Cancer Res. 2000;60:1927–1933. [PubMed] [Google Scholar]
  • 399.Levy SM, Herberman RB, Lippman M, D’Angelo T, Lee J. Immunological and psychosocial predictors of disease recurrence in patients with early-stage breast cancer. Behav. Med. 1991;17:67–75. doi: 10.1080/08964289.1991.9935161. [DOI] [PubMed] [Google Scholar]
  • 400.Mantovani A, Bottazzi B, Colotta F, Sozanni S, Ruco L. The origin and function of tumor-associated macrophages. Immunol. Today. 1992;13:265–270. doi: 10.1016/0167-5699(92)90008-U. [DOI] [PubMed] [Google Scholar]
  • 401.King R. Cancer Biology. Harlow, England: Addison Wesley Longman, Edinburgh Gate; 1996. [Google Scholar]
  • 402.Shu S, Plautz G, Krauss J, Chang A. Tumor immunology. J. Am. Med. Assoc. 1997;278:1972–1981. [PubMed] [Google Scholar]
  • 403.Andersen BL, Farrar WB, Golden-Kreutz D, Kutz D, Kutz LA, MacCallum R, Courtney ME, Glaser R. Stress and immune responses after surgical treatment for regional breast cancer. J. Natl. Cancer. Inst. 1998;90:30–36. doi: 10.1093/jnci/90.1.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404.Baron RS, Cutrona CE, Hicklin D, Russell DW, Lubaroof DM. Social support and immune function among spouses of cancer patients. J. Pers. Soc. Psychol. 1990;59:344–352. doi: 10.1037//0022-3514.59.2.344. [DOI] [PubMed] [Google Scholar]
  • 405.Glaser R, Kiecolt-Glaser JK, Bonneau RH, Malarkey W, Kennedy S, Hughes J. Stress-induced modulation of the immune response to recombinant hepatitis B vaccine. Psychosom. Med. 1992;54:22–29. doi: 10.1097/00006842-199201000-00005. [DOI] [PubMed] [Google Scholar]
  • 406.Linn BS, Linn MW, Kilmas NG. Effects of Psychosocial stress on surgical outcomes. Psychosom. Med. 1988;50:230–244. doi: 10.1097/00006842-198805000-00002. [DOI] [PubMed] [Google Scholar]
  • 407.Snyder BK, Roghmann KJ, Sigal LH. Stress and psychosocial factors: Effects on primary cellular immune response. J. Behav. Med. 1993;16:143–161. doi: 10.1007/BF00844890. [DOI] [PubMed] [Google Scholar]
  • 408.Theorell T, Orth-Gomer K, Eneroth P. Slow-reacting immunoglobulin in relation to social support and changes in job strain: A preliminary note. Psychosom. Med. 1990;52:511–516. doi: 10.1097/00006842-199009000-00003. [DOI] [PubMed] [Google Scholar]
  • 409.Thomas PD, Goodwin JM, Goodwin JS. Effect of social support on stress-related changes in cholesterol level, uric acid level, and immune function in an elderly sample. Am. J. Psychiatry. 1985;142:735–737. doi: 10.1176/ajp.142.6.735. [DOI] [PubMed] [Google Scholar]
  • 410.Kiecolt-Glaser JK, Dura JR, Speicher CE, Trask OJ, Glaser R. Spousal caregivers of dementia victims: longitudinal changes in immunity and health. Psychosom. Med. 1991;53:345–362. doi: 10.1097/00006842-199107000-00001. [DOI] [PubMed] [Google Scholar]
  • 411.Kiecolt-Glaser JK, Glaser R. Psychoneuroimmunology: Can psychological interventions modulate immunity: Special issue: Behavioral medicine: an update for the 1990’s. J. Consult. Clin. Psychol. 1992;60:569–575. doi: 10.1037//0022-006x.60.4.569. [DOI] [PubMed] [Google Scholar]
  • 412.Van Rood YR, Bogaards M, Goulmy E, Houwelingen HC. The effects of stress and relaxation on the in vitro immune response in man: A meta-analytic study. J. Behav. Med. 1993;16:163–181. doi: 10.1007/BF00844891. [DOI] [PubMed] [Google Scholar]
  • 413.Bergsma J. Illness, the mind, and the body: Cancer and immunology, an introduction. Theor. Med. 1994;15:337–347. doi: 10.1007/BF00993792. [DOI] [PubMed] [Google Scholar]
  • 414.Landmann RM, Muller FB, Perini C, Wesp M, Erne P, Buhler FR. Changes of immunoregulatory cells induced by psychological and physical stress: relationship to plasma catecholamines. Clin. Exp. Immunol. 1984;58:127–135. [PMC free article] [PubMed] [Google Scholar]
  • 415.Naliboff BD, Benton D, Solomon GF, Morley JE, Fahey JL, Bloom ET, Makinodan T, Gilmore SL. Immunological changes in young and old adults during brief laboratory stress. Psychosom. Med. 1991;53:121–132. doi: 10.1097/00006842-199103000-00002. [DOI] [PubMed] [Google Scholar]
  • 416.Schedlowski M, Jacobs R, Stratmann G, Richter S, Hadicke A, Tewes U, Wagner TO, Schmidt RE. Changes of natural killer cells during acute psychological stress. J. Clin. Immunol. 1993;13:119–126. doi: 10.1007/BF00919268. [DOI] [PubMed] [Google Scholar]
  • 417.Esterling BA, Kiecolt-Glaser JK, Bodnar JC, Glaser R. Chronic stress, social support and persistent alterations in the natural killer cell response to cytokines in older adults. Health Psychol. 1994;13:291–298. doi: 10.1037//0278-6133.13.4.291. [DOI] [PubMed] [Google Scholar]
  • 418.Kiecolt-Glaser JK, Ricker D, George J, Messick G, Speicher CE, Garner W, Glaser R. Urinary cortisol levels, cellular immunocompetency, and loneliness in psychiatric inpatients. Psychosom. Med. 1984;46(1):5–23. doi: 10.1097/00006842-198401000-00004. [DOI] [PubMed] [Google Scholar]
  • 419.Kiecolt-Glaser JK, Fisher LD, Ogrocki P, Stout JC, Speicher CE, Glaser R. Marital quality, marital disruption, and immune function. Psychosom. Med. 1987;49:13–34. doi: 10.1097/00006842-198701000-00002. [DOI] [PubMed] [Google Scholar]
  • 420.Kiecolt-Glaser JK, Glaser R, Shuttleworth EC, Dyer CS, Ogrocki P, Speicher CE. Chronic stress and immunity in family caregivers of Alzheimer’s disease victims. Psychosom. Med. 1987;49:523–535. doi: 10.1097/00006842-198709000-00008. [DOI] [PubMed] [Google Scholar]
  • 421.Kemeny ME, Cohen F, Zegans LS, Conant MA. Psychological and immunological predictors of genital herpes recurrence. Psychosom. Med. 1989;52:195–208. doi: 10.1097/00006842-198903000-00008. [DOI] [PubMed] [Google Scholar]
  • 422.Stein M, Miller AH, Trestman RL. Depression, the immune system, and health and illness. Findings in search of meaning. Arch. Gen. Psychiatry. 1991;48:171–177. doi: 10.1001/archpsyc.1991.01810260079012. [DOI] [PubMed] [Google Scholar]
  • 423.Ironson G, Lapierre A, Antoni M, O’Hearn P, Schneiderman N, Klimas N, Fletcher MA. Changes in immune and psychological measures as a function of anticipation and reaction to news of HIV-1 antibody status. Psychosom. Med. 1990;52:247–270. doi: 10.1097/00006842-199005000-00001. [DOI] [PubMed] [Google Scholar]
  • 424.Lechin F, van der Dijs B, Vitelli-Florez G, Lechin-Baez S, Azocar J, Cabrera A, Lechin A, Jara H, Lechin M, Gomez F. Psychoneuroendocrinological and immunological parameters in cancer patients: Involvement of stress and depression. Psychoneuroendocrinology. 1990;15:435–451. doi: 10.1016/0306-4530(90)90067-j. [DOI] [PubMed] [Google Scholar]
  • 425.Levy SM. Perceived social support and tumor estrogen/progesterone receptor status as predictors of natural killer cell activity in breast cancer patients. Psychosom. Med. 1990;52:73–85. doi: 10.1097/00006842-199001000-00006. [DOI] [PubMed] [Google Scholar]
  • 426.Temoshok L, Heller BW, Sagebiel RW, Blois MS, Sweet DM, DiClemente RJ, Gold ML. The relationship of psychosocial factors to prognostic indicators in cutaneous malignant melanoma. J. Psychosom. Res. 1985;29:139–153. doi: 10.1016/0022-3999(85)90035-2. [DOI] [PubMed] [Google Scholar]
  • 427.Shavit Y, Terman GW, Martin FC, Lewis JW, Liebeskind JC, Gale RP. Stress, opioid peptides, the immune system, and cancer. J. Immunol. 1985;135:834s–837s. [PubMed] [Google Scholar]
  • 428.Ben-Eliyahu S, Yirmiya R, Liebeskind JC, Taylor AN, Gale RP. Stress increases metastatic spread of a mammary tumor in rats: Evidence for mediation by the immune system. Brain Behav. Immun. 1991;5:193–205. doi: 10.1016/0889-1591(91)90016-4. [DOI] [PubMed] [Google Scholar]
  • 429.Hellstrand K, Hermodsson S. An immunopharmacological analysis of adrenaline-induced suppression of human natural killer cell cytotoxicity. Int. Arch. Allergy Appl. Immunol. 1989;89:334–341. doi: 10.1159/000234972. [DOI] [PubMed] [Google Scholar]
  • 430.Masera R, Gatti G, Sartori ML, Carignola R, Salvadori A, Magro E, Aneli A. Involvement of Ca2+-dependent pathways in the inhibition of human natural killer (NK) cell activity by cortisol. Immunopharmacology. 1989;18:11–22. doi: 10.1016/0162-3109(89)90026-x. [DOI] [PubMed] [Google Scholar]
  • 431.Benschop RJ, Nijkamp FP, Ballieux RE, Heijnen CJ. The effects of beta-adrenoceptor stimulation on adhesion of human natural killer cells to cultured endothelium. Br. J. Pharmacol. 1994;113:1311–1316. doi: 10.1111/j.1476-5381.1994.tb17141.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Benschop RJ, Rodriguez-Feuerhahn M, Schedlowski M. Catecholamine-induced leukocytosis: early observations, current research, and future directions. Brain Behav. Immun. 1996;10:77–91. doi: 10.1006/brbi.1996.0009. [DOI] [PubMed] [Google Scholar]
  • 433.Schedlowski M, Hosch W, Oberbeck R, Benschop RJ, Jacob R, Raab HR, Schmidt RE. Catecholamines modulate human NK cell circulation and function via spleen-independent beta 2-adrenergic mechanisms. J. Immunol. 1996;156:93–99. [PubMed] [Google Scholar]
  • 434.Dhabhar FS, Miller AH, McEwen BS, Spencer RL. Effects of stress on immune cell distribution. Dynamics and hormonal mechanisms. J. Immunol. 1995;154:5511–5527. [PubMed] [Google Scholar]
  • 435.Romeo HE, Colombo LL, Esquifino AI, Rosenstein RE, Chuluyan HE, Cardinali DP. Slower growth of tumours in sympathetically denervated murine skin. J. Auton. Nerv. Syst. 1991;32:159–164. doi: 10.1016/0165-1838(91)90066-c. [DOI] [PubMed] [Google Scholar]
  • 436.ThyagaRajan S, Madden KS, Kalvass JC, Dimitrova SS, Felten SY, Felten DL. L-deprenyl-induced increase in IL-2 and NK cell activity accompanies restoration of noradrenergic nerve fibers in the spleens of old F344 rats. J. Neuroimmunol. 1998;92:9–21. doi: 10.1016/s0165-5728(98)00039-3. [DOI] [PubMed] [Google Scholar]
  • 437.ThyagaRajan S, Felten SY, Felten DL. Restoration of sympathetic noradrenergic nerve fibers in the spleen by low doses of L-deprenyl treatment in young sympathectomized and old Fischer 344 rats. J. Neuroimmunol. 1998;81:144–157. doi: 10.1016/s0165-5728(97)00169-0. [DOI] [PubMed] [Google Scholar]
  • 438.Thyagarajan S, Meites J, Quadri SK. Deprenyl reinitiates estrous cycles, reduces serum prolactin, and decreases the incidence of mammary and pituitary tumors in old acyclic rats. Endocrinology. 1995;136:1103–1110. doi: 10.1210/endo.136.3.7867565. [DOI] [PubMed] [Google Scholar]
  • 439.Brenner GJ, Felten SY, Felten DL, Moynihan JA. Sympathetic nervous system modulation of tumor matastases and host defense mechanisms. J. Neuroimmunol. 1992;37:191–201. doi: 10.1016/0165-5728(92)90003-4. [DOI] [PubMed] [Google Scholar]
  • 440.Ben-Eliyahu S. Stress, natural killer cell activity, and tumor metastasis: The role of catecholamines and corticosteroids. In: Levi A, Grauer E, Ben-Nathan D, De-Koet ER, editors. New Frontiers in Stress Research: Modulation in Brain Function. Amsterdam: Harwood Academic; 1998. pp. 203–215. [Google Scholar]
  • 441.Stefanski V, Ben-Eliyahu S. Social confrontation and tumor metastasis in rats: Defeat and beta-adrenergic mechanism. Physiol. Behav. 1996;60:277–282. doi: 10.1016/0031-9384(96)00014-5. [DOI] [PubMed] [Google Scholar]
  • 442.Shakhar G, Ben-Eliyahu S. In vivo beta-adrenergic stimulation suppresses natural killer activity and compromises resistance to tumor metastasis in rats. J. Immunol. 1998;160:3251–3258. [PubMed] [Google Scholar]
  • 443.Ben-Eliyahu S, Shakhar G. The impact of stress, catecholamines, and the menstrual cycle on NK activity and tumor development: from in vitro studies to biological significance. In: Ader R, Cohen N, Felten DL, editors. Psychoneuroimmunology. 3rd edition. v. 2. San Diego: Academic Press; 2001. pp. 545–563. [Google Scholar]
  • 444.Hasegawa H, Saiki I. Psychosocial stress augments tumor development through beta-adrenergic activation in mice. Jpn. J. Cancer Res. 2002;93:729–735. doi: 10.1111/j.1349-7006.2002.tb01313.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Kyprianou N, Benning CM. Suppression of human prostate cancer cell growth by alpha1-adrenoceptor antagonists doxazosin and terazosin via induction of apoptosis. Cancer Res. 2000;60:4550–4555. [PubMed] [Google Scholar]
  • 446.Yang G, Timme TL, Park SH, Wu X, Wyllie MG, Thompson TC. Transforming growth factor beta 1 transduced mouse prostate reconstitutions: II. Induction of apoptosis by doxazosin. Prostate. 1997;33:157–163. doi: 10.1002/(sici)1097-0045(19971101)33:3<157::aid-pros2>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
  • 447.Benning CM, Kyprianou N. Quinazoline-derived alpha1-adrenoceptor antagonists induce prostate cancer cell apoptosis via an alpha1-adrenoceptor-independent action. Cancer Res. 2002;62:597–602. [PubMed] [Google Scholar]
  • 448.Carmena MJ, Clemente C, Carrero I, Solano RM, Prieto JC. G Proteins and β-adrenergic stimulation of adenylate cyclase activity in the diabetic rat prostate. Prostate. 1997;33:46–54. doi: 10.1002/(sici)1097-0045(19970915)33:1<46::aid-pros8>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
  • 449.Juarranz MG, Guijarro LG, Bajo AM, Carmena MJ, Prieto JC. Ontogeny of vasoactive intestinal peptide receptors in rat ventral prostate. Gen. Pharmacol. 1994;5:509–514. doi: 10.1016/0306-3623(94)90207-0. [DOI] [PubMed] [Google Scholar]
  • 450.Cox ME, Deeble PD, Lakhani S, Parsons SJ. Acquisition of neuroendocrine characteristics by prostate tumor cells is reversible: implications for prostate cancer progression. Cancer Res. 1999;59:3821–3830. [PubMed] [Google Scholar]
  • 451.Cox ME, Deeble PD, Bissonette EA, Parsons SJ. Activated 3’,5’-cyclic AMP dependent protein kinase is sufficient to induce neuroendocrine like differentiation of the LNCaP prostate tumor cell line. J. Biol. Chem. 2000;275:13812–13818. doi: 10.1074/jbc.275.18.13812. [DOI] [PubMed] [Google Scholar]
  • 452.Vizi ES, Toth IE, Orsó E, Szalay KSZ, Szabó D, Baranyi M, Vinson GP. Dopamine is taken up from the circulation by, and released from, local noradrenergic varicose axon terminals in zona glomerulosa of the rat: A neurochemical and immunocytochemical study. J. Endocrinol. 1993;139:213–226. doi: 10.1677/joe.0.1390213. [DOI] [PubMed] [Google Scholar]

RESOURCES