+ All Categories
Home > Documents > Autonomic regulation of cellular immune function

Autonomic regulation of cellular immune function

Date post: 23-Dec-2016
Category:
Upload: dianne
View: 212 times
Download: 0 times
Share this document with a friend
86
Autonomic Regulation of Cellular Immune Function Denise L. Bellinger, Dianne Lorton PII: S1566-0702(14)00008-3 DOI: doi: 10.1016/j.autneu.2014.01.006 Reference: AUTNEU 1625 To appear in: Autonomic Neuroscience: Basic and Clinical Received date: 12 December 2013 Accepted date: 17 January 2014 Please cite this article as: Bellinger, Denise L., Lorton, Dianne, Autonomic Regulation of Cellular Immune Function, Autonomic Neuroscience: Basic and Clinical (2014), doi: 10.1016/j.autneu.2014.01.006 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 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.
Transcript
Page 1: Autonomic regulation of cellular immune function

�������� ����� ��

Autonomic Regulation of Cellular Immune Function

Denise L. Bellinger, Dianne Lorton

PII: S1566-0702(14)00008-3DOI: doi: 10.1016/j.autneu.2014.01.006Reference: AUTNEU 1625

To appear in: Autonomic Neuroscience: Basic and Clinical

Received date: 12 December 2013Accepted date: 17 January 2014

Please cite this article as: Bellinger, Denise L., Lorton, Dianne, Autonomic Regulationof Cellular Immune Function, Autonomic Neuroscience: Basic and Clinical (2014), doi:10.1016/j.autneu.2014.01.006

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 proofbefore it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

Page 2: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

1

Autonomic Regulation of Cellular Immune Function

Denise L. Bellingera, Dianne Lortonb

Department of Human Anatomy and Pathology

aDepartment of Human Anatomy and Pathology, Loma Linda University, School of Medicine,

Loma Linda, California, 92350, USA;

bCollege of Arts and Sciences, Kent State University and the Kent Summa Initiative for Clinical

and Translational Research, Summa Health System, Akron, OH 44304,USA.

*Corresponding Author and Contact Information:

Denise L. Bellinger, Ph.D.

Departments of Pathology and Human Anatomy

Loma Linda University School of Medicine

11021 Campus Street, AH 325

Loma Linda, CA 92350

Tel.: 909-558-7069

Fax: 909-558-0432

Email: [email protected]

Page 3: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

2

Abstract

The nervous system and the immune system (IS) are two integrative systems that work

together to detect threats and provide host defense, and to maintain/restore homeostasis.

Cross-talk between the nervous system and the IS is vital for health and well-being. One of the

major neural pathways responsible for regulating host defense against injury and foreign

antigens and pathogens is the sympathetic nervous system (SNS). Stimulation of adrenergic

receptors (ARs) on immune cells regulates immune cell development, survival, and proliferative

capacity, circulation and trafficking of immune cells for immune surveillance and for recruitment,

and directs the cell surface expression of molecules and cytokine production important for cell-

to-cell interactions and “positioning” immune cells for a coordinated immune response (IR).

Finally, AR activation of effector immune cells regulates the activational state of immune cells

and modulates the functional capacity. This review focuses on our current understanding of the

role of the SNS in regulating the IS important for host defense and immune homeostasis. SNS

regulation of IS functioning is a critical link to the development and exacerbation of chronic

immune-mediated diseases. There are many diverse mechanisms that need to be further

unraveled in order to develop sound treatment strategies that act on neural-immune interaction

to resolve or prevent chronic inflammatory diseases, and to improve health and quality of life.

Page 4: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

3

Keywords: sympathetic nervous system; innate and adaptive immunity; bone marrow; spleen,thymus;

lymph nodes; mucosal associated lymphoid tissue; hematopoeis.

Page 5: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

4

Introduction

Altered immune function can be achieved using classical behavioral conditioning

paradigms whereby an immune stimulus is repeatedly paired with a non-immune type of

stimulus (associative training), and subsequently results in the non-immune stimulus producing

a similar effect on immune function as the immune stimulus (reviewed in Cohen et al., 1994;

Riether et al., 2008). Learning and memory are required for behavioral conditioning, indicating

mediation of conditioning effects via the central nervous system (CNS). Moreover, behavioral

conditioning can alter inflammation and disease outcome (Bovbjerg et al., 1987; Ader and

Cohen, 1982; Giang et al., 1996) indicating an important role for CNS-to-immune system (IS)

regulation in health and illness. Although, all neuroendocrine hormones can affect immunity,

there are two major pathways that mediate CNS-to-IS signaling, the sympathetic nervous

system (SNS) and the hypothalamic-pituitary-adrenocortical (HPA) axis. Both pathways are

regulated centrally by limbic and autonomic circuits known to mediate the effects of stressor on

body functions. Here, we review the central autonomic circuits that influence SNS-to-IS

signaling, and how they affect immune function. A large body of research indicate that

sympathetic activation either pharmacologically or through physical or psychosocial stressors

can significantly alter immune function (Kemeny and Schedlowski, 2007), with consequences

for mental and physical health. Lesioning or stimulating or blocking the activity of, specific

autonomic/limbic brain regions/nuclei known to regulate autonomic/neuroendocrine outflow also

alters measures of immune function (reviewed Felten et al., 1991). Finally, experimental

manipulation of the autonomic outflow from the CNS to immune organs, can affect such

functions as delayed-type sensitivity reactions, natural killer (NK) cytotoxicity, cytokine

secretion, lymphocyte proliferation, and antibody production (reviewed in Bellinger et al., 2008b;

Elenkov et al., 2001). The effects of these manipulations are generally reproducible, dose-

dependent when adrenergic ligands are used and reversible by blocking the sympathetic

manipulative actions of the treatment. It is clear from these findings that the ANS provides

“hard-wiring” to and from the CNS to lymphoid tissues and SNS.

The autonomic nervous system (ANS) plays a critical role in physiological regulation under

basal conditions and in response to acute and chronic stressors. The ANS, particularly the SNS

regulates the homeostatic and host defense functions of the IS. Foreign substances termed

antigens or tissue damage are perceived by the brain as stressors, and therefore activate the

SNS, which, in turn, regulates host defense mechanisms. Psychological and physical stressor,

via activation of the HPA and SNS modulate homeostasis of the IS, can induce inflammation,

and affect the IS’s ability to heal wounds and fight infections. In this paper, we review the

regulation of the IS by the SNS.

Page 6: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

5

Both arms of the ANS, the PaSNS and SNS are classically “two-neuron chain circuits”

consisting of a pre- to postganglionic neuron connection. However, central autonomic circuits

between the forebrain, limbic system, and certain hypothalamic regions/nuclei and brainstem

nuclei make up the brain-ANS-immune axis. Postganglionic sympathetic neurons from specific

sympathetic ganglia innervate immune organs and tissues (Fig. 1); however, evidence is

lacking for parasympathetic innervation of immune organs (discussed in Bellinger et al., 2013).

Vagal afferents, as well as other visceral afferents, convey immune signals to regions of the

brain that regulate sympathetic outflow to immune organs (reviewed in Bellinger et al., 2013).

Additionally, preganglionic neurons supply the adrenal medulla, which releases the

catecholamines, norepinephrine and epinephrine into the circulation. Therefore the SNS can

have both localized and systemic effects on immunity. These pathways provide the anatomical

substrate for SNS-immune cross-talk. Afferent visceral fibers, including those in the vagus

nerves, are an important route for conveying information concerning tissue damage or antigen

exposure and subsequent immune activation to the central nervous system (CNS). Additionally,

as a slower route of communication circulating cytokines, particularly proinflammatory cytokines

can interact with the brain at circumventricular organs (CVOs), which lack a blood-brain barrier

or can be actively transported into certain regions of the brain (reviewed in Dantzer, 2001).

Immune activation also induces the production of proinflammatory cytokines centrally (Rothwell,

1991; Watkins and Maier, 2000; Neumann, 2001; Felderhoff-Mueser et al., 2005).

“Figure 1 here”

While multiple neuromediators are released from sympathetic nerves, norepinephrine is

the major neurotransmitter that regulates cells of the IS, and most research has focused on

noradrenergic regulation. Cells of the IS respond to noradrenergic signaling via the cell surface

expression of α- and β-adrenergic receptors (ARs) (Fig. 1). Research is beginning to unravel

the mechanisms through which the intracellular signaling pathways used by α- and β-ARs

communicate with the signaling pathways used to regulate immune functions.

Here, we discuss our current understanding of the brain-SNS-immune axis, sympathetic

regulation of immune cell development and immune function. An overview of recent findings

indicating that activation of β2-AR signaling pathways suppresses T-helper (Th)1 responses and

cellular immunity, and drives Th2 responses important for humoral immunity, and adrenergic

regulation of more recently characterized Th17- and Treg-cells are presented. Under chronic

conditions where the IS fails to eliminate the threat and/or to restore immune homeostasis, the

SNS can promote inflammation and boost cellular immunity, the goal of which is presumed to

localize the inflammatory response and provide protection systemically from the detrimental

effects of inflammation. SNS regulation of immune cell development in the bone marrow (BM)

and thymus is complex and is not well understood. Most limited is our understanding of the

Page 7: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

6

effects of sympathetic outflow on mucosal immunity, and effects on effectors cells of the innate

IS. Research reviewed here underscores the need for better understanding central mechanism

that influence sympathetic outflow, and α1- and β-AR-mediated regulation of immune function in

order to develop early-intervention therapeutics aimed at recovery of sympathetic-immune

dysregulatory mechanisms under certain physiological conditions and disease states.

Central Regulation of Autonomic Outflow to Immune Organs

Central descending sympathetic pathways responsible for regulating sympathetic nerve

activity (SNA) in lymphoid organs (LOs) and tissues are best defined for the spleen. The spleen

is innervated by neurons in the superior mesenteric –celiac ganglion (SMCG), and subsequently

sympathetic preganglionic neuron (SPNs) (Bellinger et al., 1993; Wan et al., 1993). Supraspinal

sympathetic circuits regulate resting tonic and bursting patterns of SNA in the spleen, as well

as, the relationship between splenic SNA and other target organs that must work together to

achieve an integrative sympathetic response during homeostasis and to acute or chronic stress

(Kenney et al., 2003a,b; Osborn et al., 1987; Taylor and Schramm, 1987; Loewy, 1990;

Dampney, 1994; Sun, 1995). Although limited, available data about central sympathetic

regulatory circuits derive from transsynaptic retrograde tracing, electrophysiological recording

combined with brain lesions and/or microinjection of pharmacological agents into discrete brain

regions, and brain lesioning studies. Brain lesioning and stimulation studies are consistent with

the transsynaptic tracing findings. Cano and colleagues (2000, 2001) have mapped the central

location/density of pseudorabies virus (PRV) across time after its injection into the rat spleen.

They report that postganglionic SMCG neurons innervating the spleen receive input from

intermediolateral cell column SPNs from T3 to T12 (Cano et al., 2001), consistent with region-

specific SPN-evoked splenic SNA activity (Taylor and Weaver, 1992).

Between 72 and 110 h post-inoculation there is progressively increased PRV-positive

neurons in central autonomic regulatory nuclei/regions. Their findings are similar to PRV tracing

studies in other sympathetically-targeted organs/tissues (i.e., adrenal medulla, pineal gland,

heart, urinary bladder, rat tail skin, and adipose tissue), supports a basic hierarchy of central

autonomic regions involved in regulating sympathetic outflow, but does not delineate the organ-

specific microcircuitry that regulate sympathetic outflow (Janig and McLachlan, 2013). For

example, the parvocellular region of the paraventricular nucleus (PVN), A5 region, ventral,

medial and lateral medulla, and caudal raphe project to SPNs (Strack et al., 1989a,b; Jansen et

al., 1993; Schramm et al., 1993). Nuclei labeled after longer intervals post-PRV treatment agree

with reports of indirect projects to the SPNs to influence sympathetic activity (Loewy, 1990).

Cano and colleagues corroborate the absence of efferent parasympathetic innervation of the

Page 8: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

7

spleen (Bellinger et al., 1993; Wan et al., 1993). Labeled neurons in the A7, NTS (medial part)

and C3 cell group are unique (Cano et al., 2001) compared with other transneural tracing

studies (Strack et al., 1989a,b; Jansen et al., 1993; Schramm et al., 1993). Importantly, their

findings generally agree with the general view of the CNS circuitry that regulates autonomic

function and the stress response (Loewy, 1990; Saper, 1995).

As for the exceptions, the lateral parabrachial nucleus and autonomic-regulating thalamic

nuclei are not infected in this study. Secondly, Barrington’s nucleus (BN), subcoeruleus and EW

labeled after PRV injection (Cano et al., 2000, 2001) are generally not included in central

sympathetic circuitry. The BN and EW regulate parasympathetic outflow, but more complex

broader roles have been suggested (Jansen et al., 1997; Farkas et al., 1998; Cano et al., 2000;

Pardini et al., 1989), such as integrating supraspinal inputs for coordinating central sympathetic

and parasympathetic activity. The BN receives inputs from diverse brain regions and connects

with systems mediating stress responses (Chen and Herbert, 1995; Imaki et al., 1991, 1993;

Palkovits et al., 1997; discussed in Sved et al., 2002). Studies that examine autonomic circuits

to the spleen and other visceral organs using different strategies (i.e., brain lesioning,

electrophysiology, and CRH microinjection) are consistent with these anatomical findings.

Paraventricular Nucleus (PVN). As a major SNS regulator, the PVN integrates

information from the brainstem, forebrain, neuroendocrine and limbic regions (Swanson and

Sawchenko, 1983; Nunn et al., 2011), providing a neural substrate for generating complex

output patterns to sympathetic nerves supplying visceral organs, like the spleen. Its projections

to the RVLM, raphe nuclei, parabrachial nucleus, BN, and intermediolateral cell column

(Swanson and Sawchenko, 1983; Cano et al., 2001) form the anatomical substrate for

influencing sympathetic outflow (Swanson and Sawchenko, 1983; Luitten et al., 1985; Krukoff et

al., 1994; Badoer, 2001). Moreover, functional studies support PVN regulation of sympathetic

outflow to the spleen. Microinjecting glutamate into the PVN (Katafuchi et al., 1993a,b)

increases splenic nerve activity, and electrolytic lesions of PVN reduces splenocyte mitogenic

responsiveness (Brooks et al., 1982). Similarly, hypothalamic lesions that include the PVN

attenuate stress-mediated suppression of splenocyte proliferation (Pezzone et al., 1994).

Multiple neurotransmitters/neuromodulators and their receptors interact in the PVN in

complex ways, including -aminobutyric acid (GABA) via GABAA receptors, nitric oxide,

glutamate and angiotensin II (Kenney et al., 2001b, 2003a,b; Herman et al., 2000).

Electrophysiological studies after neurotransmitter microinjection into the PVN has increased

our understanding of its regulation of splenic sympathetic outflow. In conscious or anesthetized

rats, glutamate or other excitatory amino acids injected into the PVN generally increases plasma

catecholamine content (Martin and Haywood, 1992) and SNA in spleen (Katafuchi et al.,

1993a,b), brown fat, and other visceral organs (Kannan et al., 1989; Katafuchi et al., 1988;

Page 9: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

8

Deering and Coote, 2000; Lu et al., 1991; Yoshimatsu et al., 1993). However, amino acids

transmitters can suppress renal SNA activity (Katafuchi et al., 1988; Deering and Coote, 2000;

Lu et al., 1991) or exert mixed effects in cardiac sympathetic nerves (Deering & Coote, 2000).

Directionally opposing responses in SNA in many targets are reported for many experimental

manipulations in the PVN (Morrison, 2001). Additionally, selectivity in frequency-domain

characteristics of sympathetic nerve discharge bursts in different targets occurs after various

manipulating in the PVN (Kenney, 1994; Claassen et al., 1998; Kenney et al., 1999). Kenney et

al. (2003a,b) report that the PVN can elicit multiple response profiles in different target organs.

For example, D,L-homocysteic acid (excitatory amino acid) microinjection induces non-uniform

discharge patterns in sympathetic nerves from different targets (including the spleen), whereas

bicuculline (GABAA blocker) administration promotes uniform frequency characteristics in SNA.

Similarly, central IL-1 can differentially regulate tonic splenic and renal SNA in intact rats (Lu et

al., 2003). These findings suggest anatomically-distinct functional circuits in the PVN regulate

SNS outflow in an organ-specific manner.

Medial Preoptic Area (MPO). Bilaterally lesioning the MPO or microinjecting glutamate

suppresses splenic SNA (Katafuchi et al., 1993a), whereas interferon (IFN)-α microinjection

augments splenic SNA, blockable by prior splenic denervation (Take et al., 1993). Electrolytic

lesioning also increases splenic macrophage activity (Roszman et al., 1982) and splenocytes

number (Cross et al., 1982), but reduces splenic NK activity (Cross et al., 1984) and mitogen-

induce lymphocyte proliferation (Brooks et al., 1982; Roszman et al., 1982). The posterior and

lateral hypothalamus also may be involved in regulating splenic immune activity. Cano et al.

(2001) consistently found PRV-infected neurons in the lateral hypothalamus at early survival

times, suggesting lateral hypothalamic innervation of splenic SPNs or of spinal interneurons that

innervate these SPNs. Electrically stimulating the posterior or lateral hypothalamus suppresses

(Weber and Pert, 1990) or increases (Wenner et al., 1996) splenic NK activity, respectively.

Locus Coeruleus (LC). Cano et al. (2001) consistently found PRV-infected neurons in the

LC (ventral region) and lateral hypothalamus at survival times at the second earliest time point,

suggesting sparse LC innervation of splenic SPNs or of spinal interneurons that innervate these

SPNs. Another report (Sawchenko and Swanson, 1982) showing the LC ventral region

projecting to the spinal cord is consistent with this view. The LC responds to a variety, but not all

types, of stressors. The LC can modulate splenic immune function. Ablating brainstem

noradrenergic neurons, including the LC, by i.p. treatment with N-(2-chloroethyl)-N-ethyl-2-

bromobenzylamine (DSP-4) suppresses splenic immune functions, like LPS-induced splenocyte

proinflammatory cytokine production and the percentage of NK-cells (Engler et al., 2010). This

finding supports the importance of central noradrenergic tone in maintaining normal immune

function. The LC also participates in enhancing behaviorally conditioned recall-mediated splenic

Page 10: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

9

neutrophil activity (Chao et al., 2005) and the induction phase of a graft-vs.-host reaction in rats

(Furukawa et al., 2004). Finally, LC stimulation by local CRH administration suppresses in vitro

splenic mitogen-induced proliferation and antibody response to T-cell receptor antigens

(Rassnick et al., 1994; Caroleo et al., 1993). Therefore, data supports the LC involvement in

CNS circuitry regulating stress-induced immunosuppression in the spleen.

Edinger-Westphal Nucleus (EW). The EW projects to the periaqueductal gray and some

other brainstem areas, where sympathetic premotor neurons reside (Luppi et al., 1988; Klooster

et al., 1993), indirectly supporting EW regulation of the SNS. EW is a major central source of

urocortin (Vaughan et al., 1995). In the EW, urocortin activation of CRH receptors alters the

stress response (Weninger et al., 2000) and intracerebroventricular (icv) administration of

urocortin induces a sympathetically-driven immunosuppression in the spleen (Okamoto et al.,

1998). The stress-responsive LC and PVN project to the EW, consistent with a role regulatory

role for the EW in autonomic functioning during stress (Weninger et al., 2000).

Dorsal Vagal Complex (DVC). Cano et al. (2001) show anatomical linkage between the

NTS/AP and splenic innervation, since post-PRV inoculation of the spleen, the NTS and the C3

cell group were infected at intermediate intervals, followed by PRV-infected neurons in the AP

and dorsal motor nucleus of the vagus (DMV) boundaries later on. Peripheral nerves convey

immune signals to the NTS, a component of the DVC (Watkins et al., 1995). Similarly, immune

stimuli activate the brain at CVOs like the AP, which express interleukin (IL)-1 receptor-1

(Ericsson et al., 1995) and projects to the NTS (Cunningham et al., 1994), which in turn projects

to PVN and RVLM (Sawchenko and Swanson, 1981). Intravenous injection of

lipopolysaccharide (LPS) (Elmquist and Saper, 1996) or IL-1 (Ericsson et al., 1994) induces Fos

expression in the NTS and AP (with higher doses needed for expression in the latter). Similarly,

intraperitoneal treatment with IL-1 increases c-fos mRNA in the rat NTS and AP (Brady et al.,

1994). However, AP lesions do not alter IL-1-induced Fos expression in NTS or PVN (Ericsson

et al., 1997). Still IL-1β (285 ng/kg, intravenous) in an anesthetized rat increases splenic SNA,

an effect abrogated by high cervical spinal or midbrain transection, indicating that an intact

forebrain is necessary for splenic (and lumbar) sympathoexcitatory responses to IL-1β (Kenney

et al., 2002). In senescent rats, the responsiveness of sympathetic circuits to either hypo- or

hyperthermia are attenuated (Kenney and Fels, 2002; Kenney and Musch, 2004; Helwig et al.,

2006). IL-1β, with or without mild hypothermia or applied in different sequences, produces non-

uniform changes in SNA and SNA-coupling between renal and IBAT nerves or the splenic and

lumbar nerves (Kenney et al., 2002), supporting functional plasticity in the central sympathetic

circuits through complex relationships between immune mediators and central sympathetic

circuits. Collectively, these studies support both the NTS and AP as part of the circuitry involved

in sympathetic control of splenic immune activity, and the transduction of peripheral immune

Page 11: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

10

signals conveyed by cytokines like IL-1. Injection of IL-1 icv, but not IL-6, enhances splenic

norepinephrine turnover, as well as other organs (e.g., lung, pancreas, but not kidney, liver or

heart) (Terao et al., 1994), suggesting that central IL-1 can act on specific sympathetic circuit to

effect SNA to some organs.

Rostral Ventrolateral Medulla (RVLM). The RVLM is a key CNS region for maintaining

basal SNA and mediating sympathetic nerve responses evoked from supraspinal sites. The

RVLM receives somatic and visceral inputs, including the splenic nerve, suggesting the RVLM is

an integrative center of allostatic responses to internal and external environmental changes

(Ermirio et al., 1993). Sympathetic input is not topographically organized in the RVLM, but the

RVLM still regulates sympathetic excitatory or inhibitory drive (Beluli and Weaver, 1991b).

RVLM neurons non-uniformly regulate the resting discharge in splenic and renal sympathetic

nerves (Hayes and Weaver, 1990; Beluli and Weaver 1991a,b) under the influence of GABA.

GABAA receptor activation suppresses basal and discharge rates in splenic and other visceral

nerves (e.g., cardiac and renal) as well as bursts after activation by a variety of stimuli (e.g.,

hyperthermia, icv IL-1 or IL-6) (Hosking et al., 2009). Applying excitatory amino acids (i.e.,

glycine) or GABAA receptor agonists into the RVLM has a greater effect on renal, gastric,

hepatic, adrenal or lumbar than splenic or intestinal SNA (Meckler and Weaver, 1985, 1988a,b;

Qu et al., 1988; Stein & Weaver, 1988; Yardley et al., 1989; Stein et al., 1989; Beluli and

Weaver 1991; Hayes and Weaver, 1990), effects reducible by spinal cord transection in the

renal, but not the splenic nerve. Similarly, amino acid-activated pontine reticular neurons

tonically and actively drive splenic sympathetic nerves by distinctly different populations of

reticular neurons (Hayes and Weaver, 1992).

The identity of central sites of autonomic regulation provides potential targets for future

pharmacological and physiological studies that increase our understanding of brain-immune

interactions under normal and pathological conditions. Anatomical and functional differences

exist between sympathetic nerves supplying the spleen and non-immune organs. The electrical

activity of both the splenic and renal nerves relate to cardiovascular functions, but the number of

fibers showing cardiac-related firing pattern and the magnitude of the responses of the nerve

activity to the changes in arterial blood pressure are less in the splenic nerve than in the renal

nerve (Meckler and Weaver, 1988a,b). Additionally, the SPNs that regulate the splenic SNA

distribute more uniformly in the lateral spinal gray horn than those activating the renal nerve

(Taylor and Weaver, 1992). Finally, the splenic sympathetic nerve elicits a greater response,

both in terms of latency and frequency, to systemic administration of LPS than the renal nerve

(MacNeil et al., 1996, 1997). Moreover, functional studies like those described for the RVLM

suggest that tonic influences from supraspinal sympathetic regulatory sites distributed in a non-

uniform pattern and differentially affect the outflow sympathetic nerves under basal and

Page 12: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

11

allostatic load conditions. SNA to the kidney is more dependent upon excitatory drive from the

RVLM than SNA directed to the spleen and intestine. They also suggest discrete circuits that

regulate the outflow to specific sympathetic nerves with complex mechanisms for cross-talk

between these sympathetic circuits at multiple levels of the neuroaxis in order to coordinate an

appropriate stress-mediated sympathetic response across target organs.

Evidence for Parasympathetic Innervation of Lymphoid Organs is Lacking

No creditable neuroanatomical data are available to indicate parasympathetic efferent

nerves in immune organs (Nance et al., 1987; Nance and Sanders, 2007). Definitive markers of

parasympathetic fibers and their terminals have not been demonstrated in any immune organs

(Schäfer et al., 1998; Nance and Sanders, 2007). However, a role for afferent fibers in the

vagus, glossopharyngeal, and other sensory nerves in immune regulation has been

demonstrated (Antonica et al., 1991; Maier et al., 1998; Rosas-Ballina and Tracey, 2009), which

indirectly mediates immune functions after the information these nerves convey to the CNS.

Tracy and colleagues (Rosas-Ballina and Tracey, 2009; Rosas-Ballina et al., 2008; Tracey,

2007) has coined the term “the cholinergic anti-inflammatory reflex” in reference to the pathway

mediating IRs resulting from efferent vagal stimulation. This term, however, is a misnomer on

two accounts. First, they have not determined whether the pathway that mediates the vagal

response meets the criteria for a reflex. Anatomically, the cellular target, and afferent and

efferent limbs of this so-called reflex have not been defined. Similarly, the specific stimulus

initiating the response and the immunological response that result from stimulation also has not

been defined. Secondly, since the major neurotransmitter of postganglionic parasympathetic

neurons is acetylcholine, the term “cholinergic” infers that parasympathetic system functions as

the efferent limb, which is not consistent with neuroanatomical findings. Therefore, the proposal

by Tracey and colleagues that the efferent vagus nerves plays an important role in regulating

local and systemic inflammation by blocking splenic macrophage TNF-α production is clearly an

over interpretation of their data and not supported by the existing literature. They report that

electrically stimulating the cut efferent vagus nerve inhibits endotoxin-induced sepsis, TNF-α

production, and localized inflammation in a dermal air pouch, all dependent on the activation of

cholinergic α7 nicotinic receptors (Wang et al., 2003). No studies have been carried out by this

group to rule out the possibility that their effects could be mediated by the SNS, even though

Nance, Greenberg and colleagues (Brown et al., 1991; Vriend et al., 1993) and others (Yoon et

al., 2006) have reported similar suppression of local and systemic inflammation and splenic

macrophages TNF-α production that are sympathetically-mediated.

More recently, Tracey (2009) has acknowledged the absence of efferent vagal innervation

Page 13: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

12

of the spleen, proposing instead that the vagal efferents innervate prevertebral sympathetic

postganglionic neurons that distribute to the spleen. However, carefully performed transsynaptic

(Cano et al., 2001) and anterograde (Nance and Burns, 1989) studies are not consistent with

this hypothesis, as they failed to demonstrate a vagal-sympathetic-spleen connection.

Moreover, a recent systematic electrophysiological study (Bratton et al., 2012) has further

substantiated that direct vagal-splenic nerve connection do not exist in rats. Stimulating the

intact vagus nerve in which sensory fibers are present produces an electrophysiological

response in the splenic nerve characterized by inhibition and then excitation. Crushing the

central branch of the vagus nerve (comprised of afferent fibers) ameliorates this response

indicating that the link between the splenic nerve and the vagal stimulation occurs via vagal

afferent input to the CNS, consistent with the lack of efferent vagal innervation of the spleen.

Splanchnic nerve stimulation immediately increases the firing rate in postganglionic cell bodies

that specifically target the spleen, but vagal stimulation has no effect on their firing rates

(Bratton et al., 2012). Finally, they find no contacts between vagal efferent terminals and splenic

sympathetic motor neurons after combined retrograde and anterograde tracing from the spleen

and DMV, respectively (Bratton et al., 2012), consistent with their electrophysiological findings

and previous neuroanatomical studies (Nance and Burns, 1989; Cano et al., 2001). Vida et al.

(2011a) show that vagally-mediated splenic macrophage TNF-α production after LPS treatment

remains intact in α7 knockout mice even after cutting the efferent end of the vagus nerve,

discounting the requirement for α7 nicotinic receptor involvement or efferent vagal motor

neurons. Altered immune regulation reported in α7 knockout mice may reflect defects in the

SNS documented in these mice (i.e., diminished norepinephrine release and adrenergic ligand

sensitivity of target organs), whereas the PaSNS remains normal (Franceschini et al., 2000).

Moreover, they report that direct stimulation of the splenic sympathetic nerve in α7 knockout

mice dramatically suppresses LPS-induced TNF-α to a similar extent as that produced by

afferent vagal stimulation, an effect that is not present in β2-AR knockout mice (Vida et al.,

2011b). Finally, β-AR agonists protect mice from septic shock, long after vagal stimulation and

nicotinic agonists failed to do so (Vita et al., 2011b). Collectively, these findings underscore the

importance of vagal afferents and splenic sympathetic efferents in the regulation of immune

function. Since afferent sensory nerves in other body regions can active splenic SNA to

suppress innate immunity (Sato, 1997), the afferent vagal response is not unique, but rather

shares this function with many other afferent nerves.

Anti-inflammatory effects mediated via efferent vagal stimulation require the spleen, the

splenic nerve and β2-ARs (Vida et al., 2011a, Vida et al., 2011b). Since vagal efferent fibers do

not synapse on splenic postganglionic sympathetic neurons vagal efferent-mediated anti-

inflammatory effects must also be mediated by an indirect pathway with a non-neural

Page 14: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

13

intermediary. At this time, the non-neural intermediary and the mechanism for splenic nerve

discharges remain unknown. In another animal model system, non-neural intermediary release

of hormones from the gut is mediated by the efferent vagus nerve (Uvnäs-Moberg et al., 1992).

As an example, skeletal (electrical) or cutaneous (thermal or vibratory) stimulation increases gut

release of cholecystokinin (CCK) and gastrin into the circulation via a vagal cholinergically-

mediated mechanism that can be blocked by co-stimulation of the vagal and splanchnic nerves

(the adrenal medullary, gut and splenic sympathetic nerves are present in the splanchnic nerve)

(Uvnäs-Moberg et al., 1992). The vagus nerve-mediated CCK production suppresses

inflammatory cytokine release due to a high-fat meal (Luyer et al., 2005), during endotoxic

shock (Ling et al., 2001) and in the spleen post-LPS treatment (Meng et al., 2002) – all

comparable to that mediated by efferent vagal stimulation in the studies by Tracey group.

Circulating CCK activates intact vagal afferent fibers (Lubbers et al., 2010), as well as, central

autonomic regulatory sites known to be activated by immune stimuli, like LPS (Verbalis et al.,

1991). Additionally, churning of the gut induced by chronic efferent vagus stimulation would

activate alimentary tract and abdominal visceral sensory afferent fibers as well. Sensory

feedback would, in turn, activate descending sympathetic central and peripheral neural

pathways producing β2-AR-mediated anti-inflammatory actions, including blockade of LPS-

induced splenic TNF-α production, anti-septic shock, and TNF-α production in the inflamed air

pouch model (Aoki et al., 2005; Nance et al., 2005; Kwon et al., 2003). Thus, efferent vagal

stimulation can elicit the release of many signaling molecules from gut mucosa and/or gut IS

that activate central sympathetic pathways via the afferent vagus nerves or circulation in the

blood may explain these findings, but remains to be determined. It is clear that the anti-

inflammatory response that results from vagal stimulation is NOT mediated by a direct neural

link with the SNS or the spleen (see Martelli, McKinley & McAllen 2014, Cervi, Lukewich and

Lomax 2014 in this issue of Autonomic Neuroscience).

Sympathetic Nerves Innervate Lymphoid Organs (LOs) and Tissues

Norepinephrine, the major neurotransmitter in sympathetic nerves, fulfills the criteria for

neurotransmission with immunocytes in primary and secondary LOs (SLOs) (reviewed in

Madden et al., 1995), confirmed by a large literature using a variety of in vivo, ex vivo, and in

vitro approaches. In primary and SLOs, noradrenergic sympathetic nerves innervate the

vasculature and parenchymal fields of lymphocytes and associated cells in all mammalian

species examined. These organs include bone marrow, thymus, spleen, lymph nodes (LNs),

and mucosa-associated lymphoid tissues (MALT) – gut (GALT: the appendix, sacculus

rotundus, and Peyer’s patch) and bronchus (BALT: tonsils).

Page 15: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

14

Noradrenergic varicosities closely appose many immune cell types – lymphocytes,

macrophages, dendritic cells (DCs), eosinophils, mast cells (MCs), thymocytes, thymic epithelial

cells, and BM hematopoietic stem cells (HSCs) and others. In fact, electron microscopic studies

in the splenic white pulp have demonstrated that sympathetic nerve terminals in direct

apposition to T-cells and adjacent to both DCs and B-cells, with the neuroimmune junction being

approximately 6 nm wide (Felten et al., 1985; Felten and Olschowka, 1987) in contrast to a

typical CNS synapse that is approximately 20 nm wide or the 205 nm wide junctions in other

SNS-effector target. In this paper, we refer to these non-synaptic junctions as neuroeffector

junctions, meaning that they are sites where sympathetic motor neurons release

neurotransmitter, predominantly norepinephrine, to affect non-neuronal immune target cells.

Characteristic of these junctions are varicose nerves that release transmitters ‘en passage’ to

act on transmitter-specific receptors expressed on target immune cells with varying distances

from where the transmitter is released. Finally, there are no post-junctional membrane

specializations, but the target cells express abundant receptors for norepinephrine and for other

neurotransmitters released by sympathetic nerves.

The presence of “classical” or the atypical close contact to cells is probably not

prerequisite for neurotransmission in lymphoid tissue. Since there are no physical barriers,

norepinephrine released from noradrenergic nerve terminals diffuses through the parenchyma

establishing a high-to-low gradient from the site of release. This type of release and subsequent

diffuse to target cells is called paracrine release (Lever et al., 1965; Su and Bevan, 1970). This

route of signaling is particularly likely in the spleen where high concentrations of norepinephrine

reach the venous drainage (Brown, 1964). Noradrenergic innervation is regional and specific,

generally distributing to T-cell and plasma cell (PC) compartments, but avoiding nodular regions

and zones where B-cells develop or mature. Neuropeptides, like neuropeptide Y, colocalize

with norepinephrine in sympathetic nerve terminals, can be differentially released based on the

firing patterns and activity of the nerve, and can modulate the functional effects of released

norepinephrine (Felten et al., 1985; Bellinger et al., 2013). Paracrine release suggests that the

juxtaposition of the sympathetic terminal to immunocytes in geographically specific patterns may

be considerably important functionally.

After its release, norepinephrine binds to α- and/or β-ARs on the cell surface of

immunocytes, including antigen-presenting cells (macrophages, DCs), T- and B-cells, HSCs,

thymocytes, granulocytes, and other cells of the IS (Felten et al., 1985; Ricci et al., 1999;

Bellinger et al., 2013). The effects of catecholamines on immune measures may be determined

in part by the available concentration of norepinephrine in an immune cells milieu, for example

excitatory at low and inhibitory at higher norepinephrine, the concentration, the AR subtype,

density, activational state, and the type of G protein coupled to the AR. Generally, β2-ARs are

Page 16: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

15

predominantly expressed in cells of the IS, and in physiological concentrations their activation

limits the magnitude of acute and chronic inflammation, promotes Th2-driven antibody response

and suppresses cellular IRs (e.g., cytotoxic T-cell, NK-cell, neutrophil, and macrophage activity).

Sympathetic nerve function can modulate both the initiation and effector phases of an IR

(summarized later). Differential effects on Th1-driven antibody responses have been reported

depending on Th1-cell activational state, and their mechanism of activation. Chemical

sympathectomy by selective neurotoxins can have variable effects on antibody response,

depending on where nerve loss occurs after birth (neonatal) or in adult rodents. The drive

towards a Th2 antibody response results from a shift in the balance of cytokine production

towards a Th2 phenotype – that is an increase in IL-4 and IL-10, and a decrease in production

of IFN-γ and IL-12. Catecholamines inhibit TNF-α and IL-1, major proinflammatory cytokines

produced by antigen-presenting cells, and in concert with IL-12 play a central pathogenic role in

Th1-driven autoimmune reactions. In contrast to β2-AR mediated effects, α-agonist treatment is

generally proinflammatory, promoting an increase in TNF-α, IL-1, and a decrease in IL-10 and

IL-6. Therefore, the balance in the activation of α- vs. β-ARs may explain differential effects of

sympathetic activation on immune functions, and mechanistically, a way to either ramp up or

resolve an IR.

Bone Marrow (BM). The BM is the main site of hematopoietic stem cells (HSCs) from

which all blood cells derive. The BM also is integral for inflammation, infection or injury by

increasing production and release of platelets, leukocytes and HSCs, and promoting the acute

phase response. The SNS regulates hematopoietic cell function via direct AR-mediated effects

on hematopoietic progenitor stem cells (HPSCs) and HSCs and indirect actions on stromal and

endosteal preosteoblastic cells (Muthu et al., 2007). Intercellular interactions occur within HSC

niches innervated by sympathetic nerves with the vasculature and BM cells as neuroeffector

targets.

The SNS has important roles in regulating (1) HSC hibernation, self-renewal rate and fate;

(2) HSC pool size; (3) blood cell differentiation and functional changes; and (4) HSC and mature

blood cell ingress/egress (reviewed in Bellinger et al., 2008a). BM cells differentially express

specific α- and β-AR subclasses. Beta2-AR stimulation promotes hematopoiesis for all types of

blood cells (Fig. 2), and enhances the acute phase response after infection or tissue damage,

including neutrophil mobilization. Beta2-AR blockade (β2B) decreases the number of proliferating

HSCs. Chronic SNS activation from systemic infection or traumatic injury reduces HSC/HPSC

numbers and consequently causes anemia (Elhassan et al., 2011; Fonseca et al., 2005),

promoting multiple organ system failure. The mechanisms mediating this response vary

depending on the type of injury/infection (Baranski et al., 2011). Beta2-AR-mediated anti-

proliferative effects are evident in neutrophil, monocyte, and erythrocyte precursors (Dresch et

Page 17: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

16

al., 1981; Maestroni et al., 1998).

“Figure 2 here”

Alpha-AR activation also regulates hematopoiesis (Fig. 2). In normal mice, or after

syngeneic BM transplantation, α-AR stimulation greatly suppresses granulocyte-monocyte

colony-forming units and platelet production, but increases lymphopoiesis (Maestroni, 1995;

Maestroni and Conti, 1994a,b), effects mediated in part by transforming growth factor-β (TGF-β)

(Maestroni, 1995). Prazosin, an α1-AR antagonist, reduces thymocyte and splenic T- and B-cells

numbers after BM transplantation without affecting the relative proportions of thymocyte subsets

(Maestroni and Conti, 1994a,b). In α1B-AR knockout mice, hematopoietic recovery after

irradiation is impaired; pre-B-cell and erythrocyte regeneration is faster than in wildtype mice

(Maestroni, 2000a). Sympathetic activation can rescue hematopoiesis from sublethal X-ray

irradiation or chemotherapeutic drugs, increasing survival in mice (Dresch et al., 1984; Togni

and Maestroni, 1996). Surviving mice have greater leukocyte and platelets counts and

granulocyte/monocyte colony-forming units in the BM. These effects are mediated by α1- (Togni

and Maestroni, 1996) and β2- (Dresch et al., 1984) ARs. Lipopolysaccharide (LPS)-induced

sympathetic activation induces CD14 expression in macrophages, necessary for efficient LPS

signaling via toll-like receptors (TLRs) (Ebong et al., 2001; Wright et al., 1990) and subsequently

tumor necrosis factor (TNF)-α production (Muthu et al., 2005).

The SNS controls the circadian rhythm of hematopoiesis (Fig. 3). BM norepinephrine and

dopamine concentrations and their metabolites display diurnal rhythms (Maestroni et al., 1998)

with higher SNS activity at night, when cell division and HSC release into the blood is greatest.

Selective sympathetic denervation of the tibia ameliorates circadian progenitor egress pattern

without affecting cell viability (Méndez-Ferrer et al., 2008). Although the mechanisms for

circadian HSC/HPSC egress are not entirely understood, the SNS inhibits nocturnal CXCL12

up-regulation, reducing binds to CXCR4, which inhibit circadian HSC egress (Fig. 3) (Méndez-

Ferrer et al., 2008; Katayama et al., 2006). Stromal cell β3-ARs and osteoblast β2-ARs

participate in this response by exerting opposing actions on CXCL12 expression during the day

and night, respectively (Fig. 3) (Méndez-Ferrer et al., 2008). Beta-AR activation on osteoclast

also stimulates matrix metalloproteinase-9 release, which degrades adhesion molecules that

retain HSCs/HPSCs in the BM, and receptor activator of nuclear factor kappa-B ligand to reduce

CXCL12 expression (Kollet et al., 2006).

“Figure 3 here”

Granulocyte colony-stimulating factor (G-CSF) is required for CXCL12 and CXCR4-

mediated progenitor mobilization (Petit et al., 2002; Tesio et al., 2011). Physical/emotional

distress or injury/infection increases SNA, which induces G-CSF-mediated HSC mobilization,

reduces CXCL12 expression, and triggers specific protease-mediated degradation of adhesion

Page 18: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

17

molecules (Katayama et al., 2006; Dar et al., 2011). Disrupting sympathetic neurotransmission

in mice (i.e., chemical sympathectomy or β-adrenoceptor block [βB]) markedly impairs G-CSF-

induced HSC mobilization. Additionally, TGF-β release from Schwann cells that surround

sympathetic nerves also retains HSC via TGF-β type II receptor/Smad signal transduction

(Yamazaki et al., 2011). Collectively, these findings have clinical implications. Augmenting SNA

may be beneficial in potentiating G-CSF-induced HSC mobilization for transplantation, attracting

HSCs to HSC niches after transplantation, and developing myeloablative-protective strategies

for patients receiving chemo- and/or radiotherapies. Sympathetically-driven HSC

expansion/mobilization may improve health outcomes for wound healing and preventing anemia

during sepsis and certain cancer treatments. The SNS-mediated-hemopoietic regulation may

provide new target for re-establish homeostatic maintenance of hematopoiesis under a number

of conditions where, βB-induced suppression of HPSC mobilization improves survival, like

traumatic brain injury, burns, noncardiac surgery and trauma – conditions which strongly

activates the SNS (Beiermeister et al., 2010).

Thymus. Sympathetically-mediated effects on thymocytes and thymic non-lymphoid cells

(i.e., thymic epithelial, smooth muscle, and mast cells) occur predominantly by catecholamine

binding predominantly to β2- and α1-ARs (Loveland et al., 1981; Marchetti et al., 1994;

Kavelaars, 2002; Pešić et al., 2009). Additionally, catecholamines can translocate to the nucleus

where they influence transcription processes of steroid receptors and nuclear factor kappa-light-

chain-enhancer of activated B-cells (NF-B) to modulate apoptosis in thymocytes (Bergquist et

al., 1997). Sympathetic nerves are “positioned” in the thymic cortex for regulating key

mechanisms responsible for the (1) ingress of HPSC; (2) thymocyte proliferation and

maturation; (3) T-cell receptor (TCR) gene rearrangement; and (4) positive and negative

selection (reviewed in Leposavić et al., 2006; Bellinger et al., 2008a). Sympathetic nerves

distribute to vascular and cellular compartments in the thymus including thymocytes, with

particularly high density of fibers at the corticomedullary junction (Fig. 4A). Additionally,

noradrenergic nerves course along the thymic vasculature, where they may regulate HPSC

ingress and thymocyte egress. These nerves also closely appose MCs, macrophages,

eosinophils, fibroblast and other accessory cells (Novotny et al., 1990; Vizi et al., 1995). The

functional role of the SNS for these cell populations in the thymus is not clear. It is presumed

that norepinephrine ligation of α- and/or β-ARs expressed on multiple cell types in the thymus

participates in regulating T-cell development, with the AR subclasses exerting differential

influences on T-cell development. “Figure 4A-C here”

Remodeling of the thymus and its nerve supply occur under certain physiological

conditions (i.e., stress, immune activation, infection, pregnancy, and aging) causing thymic

involution largely due to increased activation of the HPA axis and the SNS (Leposavić and

Page 19: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

18

Perisić, 2008; Jondal et al., 2004). Age-related decline in the secretion of hormones that

promote growth is also associated with thymic involution (Patel and Taub, 2009). With thymic

involution, the density of noradrenergic nerves increase as thymic volume shrinks without

altering the normal nerve distribution, nerve volume, or norepinephrine content (Bellinger et al.,

1988; ThyagaRajan et al., 2011). Normal or increased SNA in a smaller thymic volume will

increase norepinephrine availability for target cells with consequences for AR regulation in

target cells. Similarly, antigen challenge causes greater thymocyte apoptosis and/or egress to

SLOs, which consequently increases nerve density (Novotny and Hsu, 1993). Maintained

sympathetic nerve integrity indicates that neuroprotective support is not compromised under all

of these conditions. Interestingly, sympathetic innervation of the rat thymus declines

concomitant with reduce thymic cortical mass as pregnancy proceeds, but recovers to pre-

pregnancy conditions at postpartum (Kendall et al., 1994).

The SNS is not required for development of normal T cells, and most studies support a

suppressive role of the SNS in this process. Mice deficient in dopamine-β-hydroxylase (DBH),

which converts dopamine to norepinephrine, have no intrinsic developmental or functional

defects in immunity (Alaniz et al., 1999). Moreover, sympathectomy permits thymocyte

development in fetal nude mice, and in fetal nonlymphoid thymic rudiments transplanted into a

sympathetically denervated eye (Singh, 1985a,b). Still, housing DBH-deficient mice in normal

vivarium conditions compared with a pathogen-free environment greatly increases mortality.

The increased mortality is associated with marked thymic involution, reduced frequency of

immature CD4+CD8+ (double positive, DP) thymocytes (Alaniz et al., 1999), a cell population

most affected by stress and rapidly turns over (Kroemer, 1995). After primary infection with

Listeria monocytogenes, DBH―/― mice housed under pathogen-free conditions have greater

liver and splenic bacterial load and higher mortality 5-8 days post-challenge than control mice

(Alaniz et al., 1999). Re-challenge with bacteria 3 weeks later leads to higher bacterial burden in

DBH―/― mice, without affecting mortality. Innate immunity limits bacterial growth the first 3 days

post-infection. Then, antigen-specific, T-cell-mediated immunity eliminates active infection and

protects against secondary infection, in part by producing cytokines, like IFN-γ and TNF-α.

Therefore, resistance to bacterial infection is reduced in DBH―/― mice, suggesting defective T-

cell responses to infection. Consistently, ex vivo proliferation and cytokine production is reduced

in anti-CD3-stimulated splenocytes from mice with primary and secondary infections.

Suppressed Th1-specific immune responses in DBH―/― mice exposed to Mycobacterium

tuberculosis or TNP-keyhole limpet hemocyanin are consistent with these findings; further

supporting impaired T-cell responses.

The SNS has a regulatory role in thymic seeding of CD4―CD8― (double negative, DN)

HPSCs (Singh, 1985b), and maintaining thymic cellularity by modulating the balance between

Page 20: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

19

apoptosis, proliferation and egress (Singh 1985a,b; Alaniz et al., 1999; Leposavić et al., 2006,

2010). Support for sympathetic modulation of thymocyte development is mostly from ex vivo

studies and flow cytometric staining for cell surface markers of T-cell differentiation after

manipulations that prevent AR ligand binding, either by in vivo treatment with α- or β-blockers or

chemical or surgical sympathectomy in rodents (Leposavić et al., 2000, 2006; Madden and

Felten, 2001; Rauski et al., 2003; Pešić et al., 2007). The α1-blocker used for some of these

studies also has central effects on sympathetic outflow, so future work needs to verify a1-

mediated effects on thymocyte development.

Before discussing SNS effects further, a brief review of thymocyte development is

warranted. After entering the thymic medulla, BM-derived HPSCs migrate to the outer cortex

seeding the thymus with thymocytes that subsequently migrate toward the medulla as they

mature. Most die by apoptosis, but those that survive acquire TCRαβ, CD4, CD8 and other cell

surface markers; these processes are tightly regulated by the local cortical milieu. The TCRαβ

allows T-cells to recognize short peptides presented by antigen-presenting cells (APCs),

whereas CD4 and CD8 define T-cells as Th- or cytotoxic T (Tc)-cells, respectively, and serve

distinct functions in these T-cell populations. In the cortex, the β chain of the TCR is rearranged

by gene recombination allowing for β chain selection (Fig. 5A). The TCR must retain structural

properties that permit cell surface expression with pre-TCRα or the thymocyte is eliminated by

apoptosis. TCRβ chain rearrangement produces a T-cell repertoire that recognizes an

impressively diverse array of peptides, which is critical for host defense.

“Figure 5A-B here”

After β chain rearrangement/selection, thymocytes increase CD4 and CD8 co-expression

(double-positive cells, DP) (Fig. 5A), and then undergo TCRα chain rearrangement at or near the

corticomedullary junction. CD4 and CD8 are co-receptors needed for TCR binding to major

histocompatibility complex (MHC) in the next major developmental stage, positive selection. DP

TCRαβ+ thymocytes must highly express TCRs that are capable of binding to MHC to pass

positive selection and survive. Next, surviving cells mature into CD8+ or CD4+ (single-positive

(SP) thymocytes by down-regulating both co-receptors and then up-regulating the appropriate

co-receptors for the MHC class that bind its TCR (i.e., CD8 or CD4 for MHC class I or II,

respectively). Finally, autoreactive thymocytes (those with high affinity TCR binding to self-

peptides presented by MHC I or II) are eliminated in the medulla by negative selection (Fig. 5A).

Medullary epithelial cells express multiple peripheral self-antigens, aiding in negative selection.

At this stage, some thymocytes, usually those with intermediate affinity for self-peptides are

selected to become Treg-cells (thymocytes expressing CD25+RT6.1+ with CD4 or CD8). The

SNS affects thymocyte develop and differentiation in part by modulating the expression of these

cell surface proteins on thymocytes.

Page 21: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

20

Figure 5A illustrates the effects of α- and β-AR stimulation (upper and lower panels,

respectively) on thymocyte differentiation. In the cortex, α-ARs exert their greatest influence on

TCRβ rearrangement, increasing the frequency of DP TCRαβ cells without affecting DN TCR―

cell frequency (Fig. 5A, upper panel, cortex) (Rauski, et al., 2003; Pešić et al., 2009;

Leposavić et al., 2010). In the medulla, α-AR activation increases CD8+ thymocyte and reduces

CD4+ and Treg thymocyte frequencies (Fig. 5A, upper panel, medulla). Irrespective of

thymocyte phenotype, α-AR activation generally suppresses thymocyte proliferation and

reduces thymocyte yield (Plećaš-Solarović et al., 2005; Pešić et al., 2009; Leposavić et al.,

2010). Interestingly, α1-AR expression is reduced with thymocyte maturation, supporting early

effects on thymocyte development. In contrast, β2-AR stimulation (Fig. 5A; lower panel)

negatively influences positive and negative selection (Leposavić et al., 2006; Pešić et al., 2007;

Madden and Felten, 2001; Pešić et al., 2007). Activation of the β2-AR-cAMP-PKA pathway (Fig.

5B) suppresses the expression of thymus leukemia and Thy-1, antigens that are associated

with thymocyte differentiation (Wajeman-Chao et al., 1998; LaJevic et al., 2010). This inhibitory

effect is mediated by reduced Thy-1 mRNA stability (LaJevic et al., 2010). The down-regulation

of Thy-1 is involved in TCR-dependent selection (Wajeman-Chao et al., 1998; Hueber et al.,

1997; Killeen, 1997). βB can raise Thy-1 expression in DP TCRαβlow/high thymocytes

approximately 2-fold (Leposavić et al., 2006; Pešić et al., 2007), consistent with norepinephrine

or cAMP reducing Thy-1 mRNA in S49 murine thymoma cells (Wajeman-Chao et al., 1998;

LaJevic et al., 2010;). Thymocytes from Thy-1―/― mice exhibit greater TCR signaling,

exaggerated negative selection, and lower de novo production of mature SP T-cells (Hueber et

al., 1997). Finally, some β-AR-mediated effects on thymocyte development may be indirect as

β-AR-ligand binding in LPS-activated thymic epithelial cells increases their cytokine production

(i.e., IL-6 and TNF-α production) (von Patay et al., 1998). Since these cytokines, particularly IL-

6, are important regulators of thymocyte development, this may be an important indirect route

for “fine tuning” CD4+ and CD8+ T-cell maturation under homeostatic conditions and with

immune activation.

Collectively, these data indicate that α- and β-ARs generally induce tonic inhibition of

thymocyte development through differential mechanisms (Plećaš-Solarović et al., 2004, 2005;

Leposavić et al., 2006; Pešić et al., 2009). Activation of β2-AR reduces thymocyte proliferation,

the efficiency of positive selection, and negative selection in part by their influence on Thy-1

expression. Importantly, α1B-ARs stimulation enhances DP TCRαβlow/high frequency and skews

thymocyte lineage commitment towards greater CD8+ and lower CD4+ thymocyte frequency.

Secondary LOs (SLO) and Tissues. All SLOs (spleen (Fig. 4B), LNs (Fig. 4C)) and

MALT) receive sympathetic innervation (Stevens-Felten and Bellinger, 1997; Bellinger et al.,

1992b; Felten et al., 1984). Sympathetic neural relationships with specific types of cellular

Page 22: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

21

compartments are similar across LOs and tissues. In these immune sites, noradrenergic nerves

travel with the local vasculature and associated connective tissue. From these neurovascular

plexuses, noradrenergic nerves enter into the surrounding lymphoid parenchyma where they

form neuroeffector junctions with cells of the IS. Species/strain differences exist in sympathetic

innervation of SLOs, and innervation can be affected by changes in certain physiological or

pathological conditions. For example, in the spleen, the extent of parenchymal nerves in the red

pulp, and the density of noradrenergic nerves in the white pulp can change under certain

physiological and pathological conditions. With advancing age, noradrenergic nerves in the

white pulp are lost (Bellinger et al., 1987), and increased in the red pulp in autoimmune arthritis

(Lorton et al., 2005, 2009). Similar to the spleen, nerve density in T-cells zones is influenced by

aging (Bellinger et al., 1992a; Bellinger et al., 1992b), LN location (Li and Novotny, 2001),

available neurotrophic support (Carlson et al., 1995), the activational state of the IS (Novotny et

al., 1994), and by diseases, particularly autoimmune disorders (del Rey et al., 2006; Lorton et

al., 1997, 2005, 2009).

GALT and BALT are innervated by sympathetic nerves (Felten, et al., 1981; Felten et al.,

1985; Straub et al., 2008; Elenkov et al., 2000; Bellinger et al., 2008a; Nohr and Weihe, 1991).

Like SLOs, sympathetic nerves extend from vascular beds in the gut and bronchial walls into

lymphoid compartments that make up GALT and BALT, respectively. In GALT, these nerves

enter into T-cell-dependent zones, course among enterochromaffin cells, and interdomally-

located plasma cells. Sympathetic nerves also supply the appendix, Peyer’s patches and

lymphoid tissue in the distal ileal “sac” that transitions into the colon (Felten et al., 1981; Felten

et al., 1985). In both GALT and BALT, sympathetic nerves avoid the germinal center of lymph

nodules, where B-cells reside. In BALT, noradrenergic nerves predominate around the lymphoid

epithelium associated with smooth muscle and the vasculature, and commonly extend into the

periphery of the follicle (Nohr and Weihe, 1991; Russo et al., 2009; Ueyama et al., 1990;

Sirot'áková et al., 2002). Innervation of the tonsil is similar to BALT (Felten et al., 1985).

Noradrenergic nerves reside near MCs and ED1+-macrophages, supporting their regulation of

these cell types. Generally, noradrenergic nerves are assumed to be sympathetic, but their

origin is not known; however, in the palatine tonsils they arise from cranial cervical and

cervicothoracic ganglia.

Tertiary Lymphoid Tissues (TLT)

The development of tertiary lymphoid tissues (TLTs) has recently been reviewed by Neyt

and colleagues (2012). With chronic inflammation, TLT or TLOs can be induced in inflamed

tissue or organs. TLT has a similar organization and structural features to lymphoid follicles in

Page 23: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

22

SLOs, including postcapillary high endothelial venules (Neyt et al., 2012), which are specialized

by cuboidal endothelial cells that permit circulating lymphocyte to move into the lymph nodes for

immune surveillance. Infiltrated cells organize into structured lesions or distinct T- and B-cell-

rich aggregates associated with follicular reticular cells or follicular dendritic cells (FDCs),

respectively (Link et al., 2011; Fletcher et al., 2010). TLO induction can occur by LT-inducer

(LTi) cells, Th-cells, or Th17-cells. In both SLO and TLO genesis, retinoic acid signals LTi or

immune cells via the retinoic-acid receptor-related orphan receptor γT (ROR)γT (Eberl et al.,

2004; Moyron-Quiroz et al., 2004; Rangel-Moreno et al., 2011; Lochner et al., 2011), which is

critical for their interaction with stromal LT organizer (LTo) cells and lymphotoxin α1β2 signaling

via lymphotoxin-β receptors. This interaction induces LTo cells to express several adhesion

molecules (vascular endothelial growth factor-1, intercellular adhesion molecule-1 (ICAM-1),

mucosal addressin cell adhesion molecule (MADCAM1) and homeostatic CCL19, CCL21, and

CXCL13. CCL19 and CCL21 acting via CCR7 attract and organize T-cells and DCs, whereas

CXCL13 attracts and organize B cells in a FDC network, so these chemokines drive the

infiltration of lymphocytes (Rangel-Moreno et al., 2007; Wengner et al., 2007; Moyron-Quiroz et

al., 2004; McDonald et al., 2005). An increase in lymphangiogenic growth factors, vascular

endothelial growth factor (VEGF)-C, VEGF-D, fibroblast growth factor-2 and hepatocyte growth

factor induce the formation of lymphatic vessels (Vondenhoff et al., 2009; Peduto et al., 2009).

LTo cells develop into FDCs and follicular reticular cells, which provide the framework on which

T- and B-cells migrate and interact with each other. Fibroblasts production of CXCL13 attracts

LTi cells causing their aggregation. Signals derived from the neurons are proposed to provide

the initial signal for LN development (van de Pavert et al., 2009; Veiga-Fernandes et al., 2007),

and possibly TLT (Rangel-Moreno et al., 2007; Wengner et al., 2007; Moyron-Quiroz et al.,

2004; McDonald et al., 2005). Specifically, both SLO and TLO appear to be controlled by

retinoic acid-mediated expression of CXCL13 and peripheral nerves may be the source of

retinoic acid (RA). Neurons contain an enzyme, retinaldehyde dehydrogenase 2, necessary for

retinoic acid synthesis, and Schwann cells produce RA. RA production increases with peripheral

nerve injury, which occurs with chronic inflammation. Despite these findings no studies have

examined the distribution of sympathetic nerves in TLTs or the role of the SNS in their

development. However, since much of the BALT and GALT are formed after birth in response to

external triggers, many investigators consider them to be at least in part TLT (Pabst and

Tschernig, 2010). Thus, the sympathetic nerve supply to BALT described above supports the

likelihood that TLT receives sympathetic nerve supply with a compartmentation similar to that

described for SLOs.

SNS Regulation of Resident and Infiltrating Inflammatory Innate Immune Cells

Page 24: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

23

Generally, cells of the innate IS express both α- and β-ARs. Alpha-AR expression of in

monocytes/macrophages may be induced after homing to tissues, since it is reported that

peripheral blood mononuclear cells (PBMCs) are α-AR-negative (Casale and Kaliner, 1984).

Dibutyryl cAMP up-regulates α1B-ARs in PBMCs by activating protein kinase A (PKA) (Rouppe

van der Voort et al., 1999, 2000b). Similarly, TNF-α or IL-1β can induce α1A-AR expression in

PBMCs (Heijnen et al., 2002). Catecholamines primarily increase circulating NK-cell and

granulocyte numbers in response to stressors, leaving T- and B-cell numbers remain relatively

unaffected (Ottaway and Husband, 1992, 1994; Benschop et al., 1996).

Stimulation of α1-AR exert effector cell in innate immune cells that are usually directionally

opposite to that seen with β2-AR stimulation, the former augmenting and the latter suppressing

effector functions. The effects of α- or β-AR stimulation on targeted immune cells are

summarized below and in Figure 6. Under homeostatic conditions, β-AR-mediated signals

usually predominates; however, when homeostatic conditions cannot be maintained an adaptive

or maladaptive response can occur whereby there is a shift towards a predominant α-AR

response. Under these conditions, β-AR may uncouple to Gs-cAMP-PKA pathway and can

promiscuously switch coupling to other G-protein signaling cascades (Daaka et al., 1997)

The roles of β2-AR in regulating resident and infiltrating inflammatory cells have been

largely studied for their role in the pathophysiology of respiratory diseases, particularly chronic

and exercise-induced asthma, for which β2-AR drugs are used therapeutically. Beta2-AR

agonists, both short (~ 4 h, albuterol) and, particularly, long acting agonists (8 and more than 20

h, formoterol and salmeterol, respectively), exert both direct and indirect inhibitory effects on

inflammatory cell activation, release of pro-inflammatory mediators, and cell survival, expansion

and recruitment. In many cases, inflammatory cell responsiveness/sensitivity to β2-agonists is

determined by β2-AR density and efficiency of coupling to Gs proteins.

“Figure 6 here”

Granulocytes. Granulocytes or polymorphonuclear leukocytes are white blood cells with

varying nuclear shapes that are loaded with secretory granules, including neutrophils,

eosinophils and basophils. Basophils have many similarities with MCs, which reside in tissues,

and will be discussed below with MCs. Cell components of innate immunity, neutrophils provide

first line host defense against invading bacteria by phagocytosis, release of cytotoxic mediators

and products that activate monocytes/macrophages promote phagocytosis and the formation of

oxidative free radicals for intracellular killing.

Granulocytes mobilize rapidly to sites of injury or infection. The limited available studies

indicates an inhibitory role of β2-ARs stimulation on neutrophil interactions with endothelial cells

(ECs) (i.e., rolling, adhesion, trans-endothelial migration) (Sato, 2004) and neutrophil function

Page 25: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

24

(Fig. 6). Activation of β2-ARs (in the nanomolar range for catecholamines) suppresses

neutrophil chemotaxis to number of chemotactic factors (i.e., C5a, leukotriene B4, and formyl-

methionyl-leucyl-phenylalanine) (Harvath et al., 1991), and suppresses phagocytosis (at higher

catecholamine concentrations ~10-5 M) (Zurier et al., 1974), lysosomal enzymes release (Zurier

et al., 1974), and respiratory bursts that is linked to degranulation (Nielson, 1987; Weiss et al.,

1996; Barnett et al., 1997). Catecholamines lower the rate of superoxide formation and shorten

the time of release (Gibson-Berry et al., 1993). Similar effects of β-agonists on eosinophils are

reported (Koyama et al., 1999). Intracellular cAMP (cAMPi) regulation of neutrophil function is

complex, involving cAMP extrusion and rate of cAMP metabolism, and its compartmentalization,

timing and magnitude of rise in concentration (Harvath et al., 1991). Data is limited for α1-AR-

mediated actions in neutrophils. Alpha-AR agonists induce granulocytes release (particularly

neutrophils) into the circulation from the marginal pool, lungs and BM (Fig. 6) (Benschop et al.,

1996). Pre-treatment with an α-AR antagonist or reserpine to deplete catecholamines reduces

LPS-induced neutrophilia, suggesting an α1-AR-mediated effect (Altenburg et al., 1997), which

may be either direct and/or indirect.

Salbutamol (β2-AR agonists) inhibits chemotactic responses of human eosinophils (Fig. 6),

without affect their synthesis of platelet-activating factor or leukotriene C4 after activation (Tool

et al., 1996). Consistent with this study, β-AR stimulation reduces chemotaxis and arylsufatase

release in guinea pig eosinophil after ingestion of Candida albicans (C. albicans) and marked it

for degradation, a process called opsonization (Masuyama & Ishikawa 1985). Arylsulfatase is

an enzyme that breaks down leukotrienes, a family of eicosanoid inflammatory mediators,

supporting an anti-inflammatory role for β2-AR activation. In contrast, α-agonist treatment

suppresses opsonized C. albicans phagocytosis and O2‒ production (Fig. 6). These findings

suggest that specific subtypes of ARs differentially regulate eosinophil functions.

MCs. MCs and basophils are major players in inflammation, particularly allergy and

anaphylaxis, but also serve protective functions against pathogens and in wound healing. MCs

and basophils are loaded with granules that contain histamine- and heparin, the main mediators

of allergic and anaphylactic responses, as well as other bioactive mediators that prevent blood

coagulation and promote inflammation. MC and basophils degranulation occurs after IgE that

has bound antigen binds to their cell surface.

Mast cells and basophils express β2-ARs, and their activation may be effective in treating

allergen-induced late-phase reaction or inhibit immediate-phase reaction (Yamaguchi et al.,

1995). Procaterol, a selective β2-agonist dose-dependently suppresses IL-8 and complement-

induced basophil migration, with 10-7 M reducing migration by these factors by 30%. In vitro

studies demonstrate that β2-agonists can completely inhibit histamine release from activated

MCs (formoterol>salmeterol>isoproterenol>albuterol) (Fig. 6) (Butchers et al., 1991), as well as

Page 26: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

25

other inflammatory mediators. Similarly, salmeterol, a long-acting β2-agonist, prevents IgE-

mediated histamine release from human basophils (Kleine-Tebbe et al., 1994), although β2-ARs

are not as effective in blocking this response as it is in MCs (Morita and Miyamoto, 1987;

Chazan et al., 1991). Agonist inhibition of MC mediator release is prevented by propranolol, a β-

AR antagonist, supporting β2-AR-mediated agonist effects (Johnson, 2002). Important for

clinical use, β2-agonists completely inhibit MC degranulation in the same concentration range as

that for relaxing respiratory smooth muscle (Butchers et al., 1991). Less consistent results

regarding β2-AR regulation of MC histamine release is reported from in vivo studies (Howarth et

al., 1982; Boulet et al., 1997; Proud et al., 1998). This is largely because MCs are more

susceptible to β2-AR desensitization (histamine>prostaglandin D2 (PGD2) or leukotrienes) than

bronchial smooth muscle (Chong et al., 1995, 2003; Chong and Peachell, 1999; Tsuji et al.,

2004); mediator-dependent differences in desensitization reflect differential β-AR-Gs-protein

uncoupling (Tsuji et al., 2004; McGraw and Liggett, 1997).

Beta2-AR-mediated MC degranulation occurs by the cAMP-PKA pathway, and to a less

extent β2-AR-mediated closing of Ca2+-activated K+ channels (iKCa1) (Duffy et al., 2005). No

mRNA or radioligand bindings data exists for α-ARs on MCs, and functional studies for α-ARs in

MCs are scarce. In a murine MC line, phentolamine, an α1-antagonist augments histamine

release that is blockable by phentolamine preincubation, suggesting α-AR down-regulation (Fig.

6) (Moroni et al., 1977). Additionally, prazosin (α1-antagonist) given during heart ischemia-

reperfusion injury prevents the decrease in MC peroxidase induced by norepinephrine-

preconditioning in a rat model that affords cardioprotection (Parikh and Singh, 1999).

Natural Killer (NK)-Cells. NK-cells are cytotoxic lymphocytes critical for both innate and

adaptive immunity. As part of the innate response, they are first responders to attack virally-

infected, stressed (apoptotic), and tumor cells. In adaptive immunity, NK-cells play a role in

developing antigen-specific immunological memory important for a rapid response to a

secondary infection with the same antigen. NK-cells are among the most responsive to the

suppressive effect of stress and catecholamines (Fig. 6) (Elenkov et al., 2000; Irwin, 1994).

Catecholamines have bimodal effects on NK-cells that are time-dependent relative to cell

activation. Infusion of norepinephrine or epinephrine, acute psychological stress or physical

exercise transiently increases circulating NK-cells by 400-600% (Benschop et al., 1996;

Schedlowski et al., 1996). NK-cells are mobilized predominantly from the spleen, but also the

marginating pool that binds to blood vessels (see Fig. 7) (Schedlowski et al., 1996). Acutely,

catecholamines reduce NK-EC interactions, an effect mediated by β-AR on NK-cells, and

independent of soluble ICAM-1, soluble E-selectin, or soluble vascular cell adhesion molecule-1

expression, but dependent on Ca2+/Mg2+ (Benschop et al., 1997), although dynamic exercise

can cause β-AR-mediated shedding of ICAM-1 (Rehman et al., 1997).

Page 27: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

26

“Figure 7 here”

In contrast, chronic stress or one-week treatment with β2-AR agonists suppresses

circulating NK-cells (as well as Tc-cells). Catecholamines or β2-agonists via β2-ARs potently

inhibit NK-cell activity (Shakhar and Ben-Eliyahu, 1998), Beta2-AR stimulation also act indirectly

on NK-cell function by inhibiting IL-12, IFN-γ or IFN-α production, cytokines essential for NK-cell

activity (Helstrand and Hermodsson, 1989; Elenkov et al., 1996; Hilbert et al., 2013). In mice,

two-hour restraint stress reduces NK-cell numbers in the lungs and blood, but not the spleen,

effects reversed by βB (Kanemi et al., 2005).

Directly stimulating the rat splenic nerve reduces NK activity, as well as Tc-cells and

activated macrophages, effects inhibited by βB (Katafuchi et al., 1993). The reduced NK activity

results from suppressed production of perforin, granzyme B and IFN- (Dokur et al., 2004).

Central administration of proinflammatory cytokines (i.e., IFN-, IFN-α, or IL-1β) or LPS

stimulate splenic SNA to suppress NK activity (Sundar et al., 1989; Katafuchi et al., 1993).

Catecholamines, β-AR ligands, and psychosocial stressors, like social disruption (SDR), inhibit

the effector functions of NK-cells against NK-sensitive tumor cells (Hellstrand and Hermodsson,

1989; Shakhar and Ben-Eliyahu, 1998; Whalen and Bankhurst, 1990). In C57Bl/6 mice, SDR

without immune stimulation changes cell surface expression on lung and splenic NK-cells

(reduced NKG2a and Ly49a, and increased CD16 and CD69) (Tarr et al., 2012) support SDR-

induced NK-cell activation. Moreover, greater CD107a expression on, and increased killing

activity and IFN-γ secretion by IgG/IL-2-costimuated splenic NK-cells suggests that SDR primes

NK-cells. SDR-induced priming of NK-cells is prevented by βB, supporting SNS-mediated

protection against non-specific cell-mediated and anti-viral/tumor immunity. NK-cells also

express A1- and A2-AR mRNAs; while less studied, their stimulation increases NK-cell

cytotoxicity by signaling via PLC- and PKA-mediated pathways, respectively (Xiao et al., 2010).

Antigen-Presenting Cells (APCs). Dendritic cells (DCs), macrophages, and B-cells can

present antigens to T-cells. DCs and macrophages phagocytize pathogens and cellular debris,

secrete immune cytokine/chemokines to orchestrate an appropriate IR, and initiate an adaptive

response by antigens presentation to T-cells via “immunological synapses” (Fig. 8. The

“immunological synapse occurs via MHC-antigen complex binding to T-cell receptor (TCR)

(signal 1) (Fig. 8A) combined with binding of CD28 on antigen-specific T-cell clones to B7

(CD80 or CD86) expressed on macrophages (or DCs) (signal 2) (Fig. 8B). Both signals are

required for T-cell activation. Antigen presentation in the absence of signal 2 (Fig. 8C) results in

T-cell anergy or apoptosis, or cytotoxic T-lymphocyte antigen 4 or (CTLA-4; also called CD154)

binding with CD40 “turns-off” the response. B cells also present antigen bound to cell surface

immunoglobulins via an “immunological synapse”, but without prior antigens processing

Page 28: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

27

(discussed below). Two subtypes of DCs have been described based on differences in the

secretion of cytokines. The predominant myeloid DCs express TLR2 and TLR4 and secrete IL-

12, whereas plasmacytoid DCs express TLR7 and TLR9 and secrete IFN-α, suggesting

differential functions in their IR to bacteria, and viruses or stressed cells, respectively. B-cells

can also present antigen to T-cells , which will be discussed below with the other lymphocyte

populations. Here, we review the available data for sympathetic regulation of

monocytes/macrophages and DCs via both α- and β-ARs.

““Figure 8 here”

Monocyte/Macrophages. Beta2-AR stimulation suppresses LPS-stimulated PBMN

expression of surface markers involved in regulating Th-cell differentiation and Th-cell balance,

including I-CAM1, CD40, and CD14 (Kuroki et al., 2004). Similarly, a1-AR antagonists can

induce IL-18 production, ICAM and CD40 in human monocytes (Takahashi et al., 2005).

Catecholamines and β-agonists also suppress LPS-induced TNF- production in monocytes,

macrophages and astrocytes (Fig. 6) (Hetier et al., 1991; Severn et al., 1992; van der Poll et al.,

1994; Nakamura et al., 1998). Similarly, IgE-activated MC TNF- secretion is blocked by β-

agonists (Bissonnette and Befus, 1997), whereas IL-10 production is enhanced (van der Poll

and Lowry, 1997). Since TNF- stimulates IL-1β production, catecholamines indirectly

suppress IL-1β (Koff et al., 1986). Catecholamines strongly suppress IL-12 production in

monocytes, LPS-activated macrophages, and DCs via β2-AR stimulation (Elenkov et al., 1996;

Panina-Bordignon et al., 1997; Haskó et al., 1998). IL-12 inhibition is critically involved in the

differentiation of CD4+ Th-cells (Panina-Bordignon et al., 1997) (see section on Th-cells below).

Catecholamines and β2-agonists also up-regulate IL-6 secretion by thymic epithelial-cells, brown

adipocytes, astrocytes and microglia (von Patay et al., 1998; Burýsek and Houstek, 1997;

Maimone et al., 1993; Norris and Benveniste, 1993). All of these effects are mediated via β2-AR

activation of the cAMP-PKA signaling pathway (Elenkov et al., 1996; van der Poll et al., 1996;

Suberville et al., 1996; Siegmund et al., 1998), except in brown adipocytes where cAMP-PKA

signaling is activated via β3-ARs.

Catecholamines suppress IFN-γ-mediated macrophage killing of melanoma cells or herpes

simplex virus-infected cells (Koff and Dunegan, 1985; 1986). Conversely, catecholamines

augment the anti-mycobacterial activity, an effect mediated via α2-ARs (Miles et al., 1996).

Stimulation by an α-agonist also increases phagocytosis by murine peritoneal macrophages in

vitro (Javierre et al., 1975). Similarly, α2-AR-stimulated peritoneal macrophages induce TNF-α

secretion in mice (Spengler et al., 1990, 1994; Szelényi et al., 2000). However, another study

reports that treating human-derived monocytes with epinephrine inhibits Mycobacterium avium

killing (Bermudez et al., 1990). With β2-AR stimulation or increased cAMPi, macrophage TNF-α,

IL-1, and IL-10 production is suppressed (Koff et al., 1986; Chou et al., 1996; Suberville et al.,

Page 29: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

28

1996; Haskó et al., 1998; Németh et al., 1997). Several mechanism account for discrepancies in

catecholamines effects on macrophages. Beta2-ARs inhibit, and α1-ARs potentiate, macrophage

effector functions. Beta2-ARs on naïve monocytes, down-regulate with continued stimulation

and/or coupling to Gi protein signaling, all of which mitigates β-AR-mediated inhibition.

Moreover, monocytes acquire α-ARs after entry from the blood into tissues, and expression

increases with activation, and causes an increase in α1B- and α1D-AR, and extracellular signal-

regulated kinase (ERK)2 expression (Herman et al., 1990; Rouppe van der Voort et al., 1999;

2000a,b, Kavelaars, 2002).

Dendritic Cells (DC). DCs express both α- and β-ARs that mediate SNS effects on DC

functions and their interactions with T-cells, which are critical for shaping the adaptive response

after antigen challenge (Maestroni, 2005, 2006; Manni and Maestroni, 2008; Kim and Jones,

2010; Yanagawa et al. 2010; Manni et al., 2011; Hilbert et al., 2013) (Fig. 6). Seiffert et al.

(2002) report that catecholamines inhibit the capacity of epidermal DCs to present antigen in

vitro. Consistent with this finding, intradermally injection of a catecholamine inhibits contact

hypersensitivity responses induced by epicutaneously administered haptens. Although

sympathetic nerves do not innervate the epidermis, there is no barrier to prevent norepinephrine

diffusion into the epidermis. Immature (iDCs), but not mature DCs (mDCs), express α1B-AR

mRNA, which contributes to their differential responses to adrenergic ligands. Available studies

generally use contact sensitivity models for in vivo studies, and rodent BM-derived DCs

(BMDCs) or human peripheral blood cells stimulated to drive DC development/maturation and

enriched for DCs. Collectively, these studies support a role for β2-ARs in DCs driving Th2- and

Th17- and suppressing Th1-type IRs by cytokine production. Beta-AR-cAMP signaling in CD40-

stimulated DCs suppresses IL-12 secretion, leading to suppressed Th1- and augmented Th2-

cell differentiation (Panina-Bordignon et al., 1997). Similarly, Goyarts et al. (2008) reports that

β2-agonists inhibit TNF-α, IL-6, IL-12p40, and IL-23 production in human DCs stimulated with

LPS. In TLR9- and TLR4-activated human plasmacytoid DCs, β2-agonists inhibit the secretion

of IFN-α and TNF secretion, respectively (Hilbert et al., 2013). Short-term exposure of LPS- or

keyhole limpet hemocyanin-activated BMDCs to norepinephrine at the start of culture attenuates

IL-12 and elevates IL-10 production, a response dependent on α2- and β-ARs and CD40 ligation

(Maestroni, 2002). Treating mice with norepinephrine-exposed DCs polarizes the IR toward a

Th2 profile. Consistent with these in vitro studies, Maestroni (2005, 2006) find a similar cytokine

response profile from skin DCs in a mouse contact sensitivity model. Similarly, β2-AR

stimulation reduces LPS-induced TNF-α, IL-1, and IL-6 secretion and reduced inflammation in

an allergy-challenge mouse model (Hu et al., 2012). Beta2-AR-mediated effects reduced

signaling through NF-B, MAPK and ERK pathways. Therefore, both in vitro and in vivo studies

consistently report β2-AR-cAMP signaling induced a Th2-type response bias by DCs activated

Page 30: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

29

by a number of different immune stimuli.

Beta2-AR agonists can also drive TLR2 and nucleotide-binding oligomerization domain-

containing protein 2 (NOD2)-co-activated BMDCs towards a Th17-type IR based on ex vivo

cytokine profiles (high IL-6 and IL-23 with IL-12 inhibition) and increased Th17/Th1-cell ratio

(Manni et al., 2011). Similarly, LPS-stimulated murine BMDCs express greater IL-33 levels in

the presence of catecholamines or cAMP, an effect reversed by β2B or a PKA inhibitor

(Yanagawa et al., 2011). Epinephrine pretreatment of BMDCs co-cultured with LPS- or

CD3/CD28-activated splenic CD4+ T-cells enhances DC MHCII, CD80, and CD86 expression,

and promotes a cytokine profile that drives Th2/Th17-type IRs in the presence of a pathogenic

stimulus (Kim and Jones, 2010) (Fig. 6). These findings may explain stress-associated diseases

that are mediated by dysregulated Th2- or Th17-type IRs.

Exposing mature DCs to a β2-agonist also reduces cross-presentation of protein antigens

while retaining exogenous peptide presentation capability, an effect mediated through Gαi/0-

protein (Hervé et al., 2013). Antigen uptake and costimulatory molecule expression are not

altered, but phagosomal Ag degradation is impaired. Cross-talk occurs between TLR4 and β2-

AR transduction pathways at the NF-κB level. Collectively, these findings support that in vivo β2-

AR agonist treatment in mice inhibits Ag protein cross-presentation to CD8+ T-cells, but

preserves their exogenous MHC class I peptide presentation capability.

Using a DC-based cancer vaccine in the murine E.G7-ovalbumin (OVA) model (Botta and

Maestroni, 2008), β2-AR expression at the site of DC inoculation can influence anti-tumor

efficacy by increasing local innate cytokine responses in tumor-bearing mice. In tumor-bearing

mice, adoptive transfer of immature OVA-loaded DCs after skin preconditioning increases the

antitumor response and diminishes tumor growth, but transfer of mature DCs treated the same

way has an opposite effect. Response differences were attributed to differences in DC migration

to draining LNs and subsequent recruitment of endogenous DCs that regulated the OVA-

specific Tc-lymphocyte response via a distinct cytokine profile and reduced indoleamine 2,3-

dioxygenase in the draining LNs.

In the presence of β1/β2B, intradermal injection of a TLR2, but not a TLR4, agonist and a

soluble protein shifts the recall memory response toward a Th1-type response (Manni and

Maestroni, 2008), mediated via increased local expression of CXCR3 ligands, IFN-β, IFN-γ, IL-

12 and IL-23 during the innate immune phase. More antigen-positive plasmacytoid DCs

migrated to the draining LNs. This is the first report of a β-AR-mediated drive in Th1 IRs, and

suggests a role for the SNS in Th1-sustained inflammatory diseases, such as psoriasis and

rheumatoid arthritis.

Stimulating α-ARs on DCs affects antigen detection/processing and DCs maturation and

migration (Fig. 6) (Maestroni, 2000b; Yanagawa et al., 2010). After norepinephrine treatment

Page 31: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

30

antigen, (OVA) capture, uptake and endocytosis by DCs increases, an effect blockable by an

α2-AR antagonist, and mediated by PI3K-ERK1/2 signaling (Yanagawa et al., 2010). These

findings support a role for SNS in immune enhancement early after antigen exposure.

Norepinephrine acting on α1-ARs enhances spontaneous skin DC emigration Systemic α2B-AR

mobilizes skin DCs, but local α1B-ARs inhibit DC migration (Maestroni, 2000b). Local α1B-AR at

the time of sensitization inhibits contact hypersensitivity on day 6 post-immunization.

Norepinephrine promotes the migration of immature BMDCs, but not mature CD40-activated

BMDCs via α1-AR stimulation, and is chemotactic for immature DC (Maestroni, 2000a,b),

monocytic cells and MC (Straub et al., 2000), likely contributing to the close proximity of these

cell populations to sympathetic nerves. Therefore, α-ARs direct DC migration from inflammatory

sites to regional LNs.

β2-AR, Lymphocytes and Adaptive Immunity

Lymphocytes are white blood cells that include large granular NK T-cells and smaller T-

and B-cells that mediate the adaptive IR. These immune cells are defined by specific cell

surface markers. NK T-cells lyse virally-infected and tumor cells. Similarly, Tc-cells kill virally-

infected cells, tumor cells and allografts. T-helper (Th)-cells regulate other immune cells through

the release of growth factors and cytokines. B-cells differentiate into antibody-secreting plasma

cells. T- and B-cell clones are activated by specific antigens, and after migration into sites where

antigens are present, become effector T- and plasma cells. The differentiation,

trafficking/migration, and effector functions of all of these lymphocyte subpopulations are

regulated by sympathetic nerve release of norepinephrine acting on ARs.

Lymphocyte Trafficking. The mechanisms that modulate catecholamine-regulated

lymphocyte distribution are not well established. The SNS innervates vascular smooth muscle to

regulate regional blood flow, and deliver lymphocytes to tissues (Benschop et al., 1996).

Lymphocytes traffic to and from tissues by passing through postcapillary venules. The SNS also

regulates the flow of lymph through tissues, which affects lymphocyte output. For example, α1-

agonists infused into the afferent lymphatic vessels of PLNs from sheep increase the

lymphocyte output of efferent lymph (Ottaway and Husband, 1992, 1994), indicating egress of

lymphocytes from the PLNs. Lymphocyte subsets differ in their density and activity of β-AR

expression which may influence catecholamine-mediated lymphocyte distribution. Lymphocytes

expressing more β-AR and/or generating more β-AR-stimulated cAMP (Tc-cells) are more

readily mobilized by catecholamine administration than those with fewer receptors and/or

weaker second messenger signaling (i.e., B-cells, Th2-cells). Like NK-cells, β2-AR-induced

mobilization of lymphocyte subsets from the marginal pool occurs via the activation of immune

Page 32: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

31

cells, not the ECs, and results from the break in adhesion between CD11a expressed on

leukocytes and CXC3R1 expressed on the EC (Fig. 7) (Dimitrov et al., 2010). In healthy human

subjects, infusion of isoproterenol mobilizes memory/activated CD8+CD29high T-cells 30 min

later, but not CD8+ CD45RA+ CD62L+ naïve T-cells (Mills et al., 2000). These effects are

prevented by βB.

Beta2-AR Expression and Th0-Cell Differentiation. Upon appropriate activation, naïve

or Th-0-cells can differentiate into a number of different types of Th-cells, based on the up-

regulation of specific intracellular transcription factors and cell surface markers (i.e., up-

regulation of the transcription factors T-bet, GATA-3, RORt, and FoxP3 in Th1-, Th2-, Th17-,

and Treg-cells, respectively; Fig. 9). Naïve T-cells, and CD4+ Th1- and Treg-cells predominantly

express β2-AR (Fig. 9A-D), while Th2-cells do not express β2-ARs (Fig. 9B) (Sanders et al.,

1997, 2001; Guereschi et al., 2013). Beta2-AR expression in other CD4+ Th subsets, i.e., Th3-

or Th17-cells, remains to be demonstrated, but functional studies support their expression in

Th17-cells (Ebbinghaus et al., 2012; Kim and Jones, 2010).

“Figure 9 here”

Figure 9 summarizes the effects of AR stimulation in naïve Th-cells (Th0-cells) on

differentiation, their dependence on the local presence of specific cytokines (black above and

below horizontal arrows) from other cellular sources. For Th1-cell differentiation, β 2-AR

stimulation induces greater IFN- secreted from activated naïve CD4+ T-cells cultured in the

presence of IL-12 (Fig. 9A) (Swanson et al., 2001; Sanders, 2012). Greater IL-12

concentrations drive greater IFN- secretion per Th1-cell and cell surface IL-12R expression,

and induce expression of the transcription factor, T-bet. In contrast, adding a β2-agonist to

activated effector Th1-cells causes suppression of IFN- secretion per cell. Beta2-AR effects on

Th1-cell differentiation are time dependent, such that β2-AR stimulation before, at the time of, or

after T-cell activation, causes no change, an increase, or a decrease, respectively, (black

vertical arrows, Fig. 9A) in IFN- production than with T-cells activated in the absence of a β2-

AR agonist (Ramer-Quinn et al., 1997, 2000; Sanders, 2012).

In the presence of IL-4 and IL-10, β2-AR stimulation on naïve T-cells drives Th2-cell

differentiation, increases IL-4 secretion per cell, and induces nuclear expression of the

transcription factor, GATA-3 (Fig. 9B). However, when activated naïve T-cells are cultured with

low or high concentrations of IL-4 and a β2-agonist, Th2-cells that develop produce greater or

low-to-normal IL-4 levels, respectively. However, when effector Th2-cells are activated with a

β2-agonist, IL-4 production does not differ from Th2-cells cultured without the β2-agonist. These

findings are consistent with the lack of β2-AR expression in Th2-cells; whereas other agents that

raise cAMP augment IL-4 production. These studies demonstrate the complexity of SNS-

immune cell interactions and indicate that a better understanding of the intracellular

Page 33: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

32

mechanisms responsible for these differences is needed before they can be translated to the

clinic in any meaningful way. Disease processes, particularly chronic inflammatory diseases,

can change β2-AR expression, intracellular signaling and/or functioning in naïve or effector

CD4+ T-cells, which contributes to altered cellular function and the development and/or disease

progression. An in depth understanding of disease-mediated changes in β2-AR expression and

functioning over the disease course is also necessary to provide rational neural-immune

approaches for restoring sympathetic regulation and for disease prevention.

Th1- and Th2-cells. In vivo studies indicate added complexities compared with in vitro

studies. Despite no β2-AR expression on Th2-cells, systemically applied catecholamines drive

Th2-type humoral IRs (Fig. 9B). Beta2-AR-driven humoral immunity is indirectly orchestrated by

APC TLR activation after they bind to intracellular microbial or viral components (Fig. 9B). TLR

activation induces DC maturation (APC in Fig. 9B) and their production of TNF- and IL-12

particularly with α1-AR co-stimulation, which drive Th0-cell differentiation towards cell-mediated

immunity (Th1-mediated) in the context of APC co-stimulation (Fig. 9A). NE binding to β2-ARs

in Th0- and differentiating Th0-cells increases IFN- secretion of per cell and together with APC

cytokine production (augmented by 1-AR) strongly drives Th0-cell differentiation towards T-

bet+Th1-effector-cells. Once Th0 cells are fully differentiated to Th1 effector cells, β2-AR

agonists either have no effect, or suppress IFN-, depending on pathogen clearance (right side,

Fig. 9A). Thus, β2-ARs exert bimodal effects on IFN- secretion, depending of the time of

stimulation relative to challenge, pathogen clearance and the state of Th-cell differentiation

(Ramer-Quinn et al., 1997, 2000). Therefore, β2-AR signaling via cAMP-PKA suppresses IFN-

production by Th1-effector cells (Saxena et al., 1999), and provides an important negative

feedback mechanism for restoring immune homeostasis after pathogen clearance. The

importance of feedback inhibition in cell-mediated autoimmunity is underscored by our recent

findings that this inhibitory feedback mechanism is not functional in lymph nodes that drain the

arthritic hind limbs in adjuvant-induced arthritis (Lorton et al., 2013). Administration of a β2-

agonist increases T-cell-derived IFN- production by altering the phosphorylation of β2-ARs-

which shifts β2-AR signaling from a cAMP-PKA to an ERK1/2 pathway. Reducing SNS tone

(unpublished findings) or co-treatment with an -and β2-AR antagonist and agonist,

respectively, restores this feedback inhibition and ameliorates the disease (Lubahn et al., 2004).

On the other hand, APC presentation of parasitic components or allergens (Fig. 9B)

suppresses DC TNF-α and IL-12 production particularly with β2-AR co-stimulation, which drives

IL-4 secretion by differentiating GATA-3+Th2 cells, and IL-6 and 10 production by the APC. This

cytokine profile suppresses Th1 differentiation and promotes Th2 differentiation and a Th2

cytokine profile (i.e., IL-4, IL-5, and antigen-depending on the antigen IL-13) (Fig. 9B; red lines

between Fig. 9A and B). APC production of TNF- and IL-12 is suppressed and IL-10 and IL-

Page 34: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

33

1β-induced IL-6 production enhanced (Koff et al., 1986; Hetier et al., 1991; Elenkov et al., 1995;

Nakamura et al., 1998; Severn et al., 1992; van der Poll and Lowry, 1997). IL-12, the main

driver of Th1 responses, reduces T-cell secretion of IFN- and increases IL-4 synthesis (Elenkov

et al., 1996; Blotta et al., 1997; Haskó et al., 1998). Additionally, β2-AR stimulation of Th0-cells

inhibits IFN- production to suppress Th1-cell and indirectly drive Th2-cell differentiation, without

actually affecting IL-4 production by Th2-cells (Borger et al., 1998; Sanders et al., 1997). In this

manner, β2-AR activation drives Th2-cell differentiation, Th2 cytokine production and humoral

(antibody) responses (Maimone et al., 1993; Norris and Benveniste, 1993; Elenkov et al., 1996;

Elenkov and Chrousos, 1999; van der Poll et al., 1996). Negative, cytokine-mediated counter-

regulation between Th1- and Th2-cell differentiation (red lines between Fig. 9A and B) in the

appropriate context ensures that the appropriate Th-cell response to a specific antigen

challenge is mounted.

Th17- and Treg-cells. Information regarding SNS regulation of other Th-cell subsets,

such as Treg- and Th17-cells, is limited, in part, because these are relatively newly-discovered

T-cell subsets. Th17-cells are a subset of Th-cells that secrete the proinflammatory cytokines,

IL-17, IL-21, IL-22, and IL-23 that perpetuate inflammation and autoimmune diseases, including

multiple sclerosis, psoriasis, autoimmune uveitis, and rheumatoid arthritis. In Th0 cells, β2-AR

stimulation also promotes Th17-cell differentiation and secretion of IL-17A and IL-23 (Fig. 9C)

(Kim and Jones, 2010). The differentiation of Th17-cells requires expression of cytokines, IL-6

and TGF-β. Kim and Jones (2010) provide data to support a β2-AR-mediated bias towards

Th17-cell activation, since β2-AR stimulation enhanced TGF-β in the presence of IL-6, which

promotes IL-17A production during Th0-cell differentiation (Fig. 9C). In contrast, β2-AR agonists

suppress the production of IL-17A in effector Th17 cells (Fig. 9C), suggesting that after

pathogen clearance, the SNS suppress Th17 effector-cells, restoring homeostasis.

Treg-cells are essential for sustaining self-tolerance and preventing the development of

autoimmune diseases, since some thymocytes with specificity to self-antigens can escape

deletion in the thymus. Elimination of self-antigens as part of normal tissue repair occurs by

processing and presentation to Th0 cells by APCs (i.e., immature DCs (iDC)) without inducing

an immune response. Presentation of a self-antigen to Th0-cells induces retinoic acids, which

drives their differentiation to Treg-cells (Fig. 9D). In this context, stimulation of β2-ARs on Th0-

cells increases TGF-β production, which facilitates retinoic acid-mediated Th0-cell differentiation

to FoxP3+Treg-cells (Fig. 9D) (Guereschi et al., 2013). Through secretion of TGF-β and IL-10,

and by direct interaction, Treg cells suppress Th0 differentiation into effector Th-cells to

maintain self-tolerance. Beta2-AR stimulation enhances Treg-cell suppression by inhibiting IL-2

and IL-2R expression in activated Th0 cells after self-antigen presentation (red lines, Fig. 9D),

since IL-2 and its receptor binding is required for proliferation of Th0-cells (referred to as clonal

Page 35: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

34

expansion). Also, β-agonists promote the production of TGF-β and IL-10, however the cell types

that produce these cytokines are not clear. In vivo manipulations of sympathetic functioning in

several studies support these in vitro findings. Sympathectomy increases Treg-cells numbers in

spleen and LNs, effects dependent on TGF-β and retinoic acid. Moreover, sympathectomy

inhibits the induction of experimental autoimmune encephalitis (Bhowmick et al., 2009).

Consistently, β2-AR activation augments OVA-induced oral tolerance in a rodent model of RA

(Cobelens et al. 2002). Therefore, both in vivo and in vitro studies support that noradrenergic

nerves promote Treg-cell differentiation and functions, however the mechanisms are unclear.

One possibility is that Treg cells require cAMP for their effector function to prevent

development of autoreactive T cells, however, they do not make cAMP. Treg cells obtain cAMP

from Th0 cells via gap junctions. Activation of β2-AR would increase cAMP in these cells,

providing a greater source of cAMP for Treg cells. Additionally, β2-AR agonists and c-AMP-

elevating agents increase Tc-lymphocyte antigen-4 (CTLA-4) expression in Treg-cells, an

important mediator of cell contact-dependent Treg-cell suppression (not shown in Fig. 9D)

(Guereschi et al., 2013; Vendetti et al., 2002). These β2-AR-mediated effects are PKA-

dependent and inhibited by β2B. Additionally, the SNS may play a role in the trafficking of Treg-

cells, as acute psychological stress reduces circulating CD4+Foxp3+ Treg- and Treg-cells

expressing CTLA-4, and up-regulates β1-AR expression on Treg-cells (Freier et al., 2010).

Together, these findings suggest that the SNS plays a role in tolerance and autoimmunity via its

regulation of Treg- and Th17-cell differentiation, and is consistent with severe stressors

preceding autoimmune onset and sympathetic dysregulation in many chronic inflammatory

autoimmune disease, including rheumatoid arthritis, multiple sclerosis, and lupus

erythematosus.

Cytotoxic T (Tc)-Cells. Available data for SNS effects on Tc-cells, while sparse, generally

support catecholamine-enhanced Tc-cell functions when they are present during initial Tc-cell

activation (Livnat et al., 1987), but subsequently inhibit effector functions (reviewed in Elenkov

et al., 2000). Inhibition of Tc-cell effector functions occurs in part by down-regulating TNF-α

production (Kalinichenko et al., 1999) and delaying the generation of effector Tc-cells (Livnat et

al., 1987). Cook-Mills et al. (1995) have shown that β-agonists inhibit the generation of anti-

MOPC-315 tumor cytotoxicity by murine splenic Tc-cells ex vivo. Similarly, β-AR agonist or

increased cAMPi suppresses the development of murine memory Tc activity against an

influenza virus (Bender et al., 1993). Catecholamines or cAMP analogs available at the time of

sensitization, augments the murine Tc-mediated cytotoxicity in a mixed lymphocyte reaction

(Hatfield et al., 1986).

B-Cells. B-cells are part of the adaptive arm of immunity, responsible for making

antibodies against antigens (i.e., humoral immunity) after they undergo differentiation into

Page 36: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

35

plasma cells. B-cells recognize antigens via the presence of B-cell receptors (BCR), a cell-

bound antibody on their cell surface. Th-cells support their maturation and function by cytokine

release and upon antigen-specific activation involving the TCR and MHC II presentation by an

antigen-presenting B-cell through an “immunological synapse” (Fig. 10). Most antigens are T-

cell-dependent and therefore require both BCR-antigen binding (1. in Fig. 10) and the co-

stimulation by antigen-specific Th-cells (2. in Fig. 10) for maximal antibody production.

Therefore, β-AR-mediated changes (green arrows in T-cell; Fig. 10) in Th-cell functions

described above can indirectly (black arrows; Fig. 10) affect B-cell function, whereas β-AR

expressed on B-cells can directly affect antibody production (green arrows in B-cell in Fig.

10). Conversely, catecholamines that bind to β2-ARs on B-cells regulate their function (green

arrows in B-cell in Fig. 10), providing another indirect route for the SNS to reciprocally

influence T-cell effector functions (blue arrow in Fig. 10), like cytokine secretion.

“Figure 10 here”

Norepinephrine depletion in severe combined immunodeficiency (SCID) mice before

reconstitution with B-cells and antigen-specific Th2-cells reduces serum antibody and

suppresses the formation of splenic germinal centers (Kohm and Sanders, 1999). These in vivo

effects of catecholamines are, at least in part, due to norepinephrine regulation of CD86

expression, a costimulatory molecule required for germinal centers development (Borriello et al.,

1997). B-cells stimulated with norepinephrine stabilize CD86 mRNA, elevate CD86 transcription,

and increase CD86 surface expression after BCR stimulation (Kohm et al., 2002). Elevated

CD86 expression drives the costimulation of Th2-cells and subsequently CD86-mediated signal

transduction in B-cells to augment IL-4-dependent production of IgG1 and IgE (Fig 11)

(Kasprowicz et al., 2000). Mice that received antigen-specific B-cells from norepinephrine-

depleted hosts and administered CD40L and IL-4 to activate B-cells, have increased serum

IgG1, an effect that is further elevated by anti-CD86 or terbutaline (β2-agonist) treatment (Kohm

et al., 2002). Dual activation of CD86 and β2-AR on B-cells has an additive effect on augmenting

IgG1 production, with the β2-agonist effects mediated via CREB and OCA-B (Fig. 11A). Data

support that CD86 engagement of a B-cell induces a signal in the B-cell that directly affects

IgG1 production per cell, independent of its effect on Th2-cells and subsequent IgG1 switching

(Kasprowicz et al., 2000; Podojil and Sanders, 2003). Elevated IgG1 per B-cell results from

greater 3’IgH enhancer activity and rates of mRNA for IgG1 (Podojil et al., 2004; Podojil and

Sanders, 2003). Additionally, Pongratz and coworkers (2006) reports that in the presence of IL-

4, stimulation of β2-AR in activated B-cells increases IgE expression, an effect that is cAMP-,

PKA- and p38MAPK-dependent; but independent of CREB (Fig. 11B). A β2-AR-mediated

increase in p38MAPK potentiates the B-cell activational rise in p38MAPK and STAT6 the

signals an increase in IgE production and secretion.

Page 37: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

36

“Figure 11A-B here”

Catecholamines have indirect effects on B-cells by modulating T-cell cytokine production

(Figs. 9, 10). While IL-4-mediated Th2-cell differentiation is not affected by catecholamines,

their effect on IFN-γ secretion by Th1-cells indirectly suppresses IL-4 production by Th2-cells

and high IL-10 production by Th2-cells can inhibit Th-cell functions (Fig. 9), which can impact

IgG1 production. Exposing Th1-cell clones or newly generated effector cells to norepinephrine

before they are activated by B-cells also reduces T-cell production of IFN-γ (Sanders, et al.,

1997), and greater IFN-γ by naïve T-cells exposed to norepinephrine augments the B-cell

production of IgG2a (Swanson, et al., 2000). Collectively, norepinephrine augments or

suppresses B-cell function indirectly by altering Th1-cytokine production and this effect is

dependent on when T-cells are exposed to norepinephrine, and their state of activation. Cyclic-

AMP-elevating agents like catecholamines can up-regulate B7 expression on B-cells (Watts et

al., 1993), which binds to the CD28 a T-cell costimulatory molecule that is part of the

“immunological synapse”, indicating increased T-cell-B-cell interaction.

The activation of B-cells requires that a critical threshold of cAMP concentration being

reached for antibody production (Pollok et al., 1991). Elevated cAMPi can sensitize resting B-

cells to immune complex-induced hyporesponsiveness (Stein and Phipps, 1991). Beta2-AR

stimulation-induced cAMP may be contributory to regulation of antibody production under these

conditions; however, future studies are needed to determine whether this is a mechanism

through which norepinephrine affects B-cell function.

Additionally, catecholamines can directly affect B-cell function via binding to ARs

expressed on their cell surface, which are predominantly β2-ARs. Both in vitro and in vivo

studies indicate that catecholamines directly affect B-cell function, and play a critical role in

determining antibody production. Like Th-cells, their effects depend on the activational state of

B-cells. Kasprowicz et al. (2000) find that incubating B-cells with catecholamines during antigen

processing or within 12 h of Th2-cell co-culture potentiates IgG1 and IgE production. However,

when B- and Th2-cell co-cultures are exposed to norepinephrine 12 h after starting the culture,

only IgG2a is elevated. Lastly, adding norepinephrine to plasma cells reduces the amount of

antibody produced (Melmon et al., 1974). These studies demonstrate the ability of

catecholamines to influence B-cell antibody production in an activation-dependent manner.

Summary

The SNS is a major integrative and regulatory pathway between the CNS and the IS. Our

understanding of the central circuits that regulate sympathetic outflow to immune organs is

limited, and in large part based on studies after immune activation with LPS, some

electrophysiological studies, and a couple of studies mapping neuronal transsynaptic infection

Page 38: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

37

with PRV after its injection into the spleen by a single research group. It is well documented that

sympathetic noradrenergic nerves innervate primary and SLOs and that norepinephrine meets

the criteria for neurotransmission, at least in the spleen. Our understanding of the interactions

between these two systems has grown considerably since the criteria for sympathetic

neurotransmission with immune cells as effector cells were first demonstrated. However, this

field is still in its infancy, and many questions remain unanswered. It is clear that the SNS

regulates all aspects of innate and adaptive immune responses, but mechanisms are still being

unraveled. Most studies use in vitro or ex vivo methods in rodent models in the presence or

absence of adrenergic agonist/antagonists or other strategies to manipulate the sympathetic

neurotransmission in selected lymphoid organs. While these studies certainly contributed

valuable information, they may not mimic in vivo functions, particularly, the in vitro studies.

Mixed results can result from differences in species or cell lines used for study, and other

factors such as timing, source and activational state of immune cells, psychological stress, age

and gender. Additionally, sympathetic regulation of blood flow, cellular infiltration and stromal

cell functions cannot be easily mimicked in vitro, and may account for disparate findings

between in vivo, ex vivo and in vitro findings. Moreover, available data supports an intimate

interaction between catecholamine signaling and another key immune regulator, the HPA

pathway suggesting cooperative homeostatic regulation of the IS; however further research is

needed to mechanistically understand this three-way interaction. In this paper, we have focused

on direct sympathetic-to-immune cell interactions via NE released from nerve terminals.

However, sympathetic nerves release other neurotransmitters/neuromodulators, such as

neuropeptide Y, adenosine, and ATP, that can influence immune function, and interact with

noradrenergic signaling to facilitate or dampen the immune response. Some studies suggest

that these mediators are differential released from sympathetic nerves depending on firing

activity, but the role of these mediators either alone or in combination with norepinephrine in

neural-immune regulation remains undefined.

Noradrenergic nerves innervate stromal and immune cells. Alpha2-ARs expressed

presynaptically negatively regulate norepinephrine release. Although, these circuits are often

termed “hardwired”, neural plasticity is a prominent feature of neural-immune interactions

allowing for the SNS to adapt to changes in its microenvironment. Immune cells respond to

norepinephrine preferentially through signaling of β2-ARs, and with lower affinity to α1-ARs, but

α1-ARs under certain conditions play important roles in sympathetic-immune regulation. The

intracellular signaling pathways activated by AR stimulation interact at multiple sites with known

signaling pathways used by immune cells to carry out their cellular functions. It is clear from

existing data that catecholamines regulate hematopoiesis for all types of blood cells and

thymocyte development, the major functions of primary LOs. The SNS also can regulate the

Page 39: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

38

circadian proliferation of BM cells, and the egress/ingress of HSCs and blood cells in the BM. A

rise in circulating epinephrine in response to an acute stressor mobilizes leukocytes from the

BM, but most of the effects of the SNS on BM are mediated by sympathetic nerves in primary,

as well as, SLOs. Sympathetic nerves in the thymus influence key steps in thymocyte

development via AR-mediated regulation of cell surface molecules, like Thy-1, and cytokine

release from stromal cells and thymocytes. Still, only a few research groups are investigating

SNS-immune cell development in primary LOs and remain understudied areas in the field of

psychoneuroimmunology, particularly sympathetic regulation of supporting stromal cells in the

BM and thymus that are critical for normal hematopoietic and thymocyte development.

With regard to adaptive immunity, the SNS clearly regulates the differentiation of Th cells,

particularly Th1- and Th2-cells. The SNS, via β2-ARs, promote differentiation Th0 cells to Th2,

and inhibit differentiation of Th1 cells. Much less is known regarding the influence of

sympathetic nerves on the differentiation of the more recently discovered Th17- and Treg-cells.

However, recent studies indicate these cells express β2-AR, and that the SNS regulates Th0 cell

differentiation toward Th17- and Treg-cells. Future studies are needed to understand the

mechanisms for SNS regulation of these T-cell subsets. Currently, Th17- and Treg-cells are

intensively studied by immunologists due to their importance in autoimmunity and cancer –

diseases linked to SNS dysfunction and where clear relationships between psychosocial stress

and disease outcome are reported.

Sympathetic regulation of mucosal immunity also remains extremely limited (see

McGovern & Mazzone, Sharkey & Savidge in this issue of Autonomic Neuroscience), but a few

studies support current views held for other SLOs, i.e., that the SNS generally suppresses Th1

and drives Th2-type cytokine production and antibody responses, and is anti-inflammatory. It is

apparent from this review that the interactions between these two systems are highly complex,

and that unraveling the mechanisms responsible for SNS-immune interplay is still in its infancy.

The anti-inflammatory properties mediated via β2-ARs have been utilized in a limited way in the

clinical setting, for example in the treatment of asthma, but the research reviewed here

underscore the necessity of future research to better understand the mechanisms for

sympathetic-immune crosstalk under normal and pathological conditions before they can be

translated to treatments that prevent or successfully treat chronic inflammatory diseases.

However, there are exceptions to the anti-inflammatory effects of the SNS under

conditions of chronic inflammation and in autoimmunity, as we have found LOs-dependent

sympathetic response in rats with adjuvant-induced arthritis (Lorton et al., 2013). In DLNs, β2-

AR agonists drive Th1 responses promoting inflammation, while in the spleen they push Th2 or

have no effect. The catecholamine-mediated Th2 drive and Th1 suppression in the spleen can

have beneficial or have detrimental consequence under certain conditions. The Th2-drive may

Page 40: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

39

be protective, particularly systemically, by preventing organ damage and anemia, from high

circulating Th1 proinflammatory cytokines and damaging products produced by the cells of

innate immunity. A chronic Th2 drive may increase susceptibility to infections that are most

efficiently combated by Th1-type response, increasing the risk for infectious complications.

Moreover chronically high SNA increases metabolism, which promote wasting, and taxes the

cardiovascular, renal, and hepatic systems. Defects in the sympathetic-immune interplay, and/or

an abnormal activity of the SNS in either direction, have been reported in many chronic

inflammatory diseases, and likely contributory to their pathophysiology.

The actions of the SNS can be pro- or anti-inflammatory, depending on many factors,

including presence and nature of the antigen, the state of the local microenvironment, where the

antigen is introduced, the activation/differentiation state of cells of the IS, activational state of

the stress pathways, and the G-coupled protein receptor (GCPR) composition, density on

immune cells, the G-proteins to which they are coupled and how they are phosphorylated. The

nature of the SNS as a homeostatic regulatory system is consistent with a proinflammatory role

at the onset of challenge or injury that facilitates the rapid clearing of microbes and/or injured

tissue and promotes healing. During the effector phase, the role of the SNS assumes an anti-

inflammatory function and suppresses immune effector cells, with the goal of “turning off” the IS

and restoring homeostasis to the fullest extent possible. Therefore, sympathetic-mediated

responses are time-dependent relative to the antigen-presentation. An in depth understanding

of disease-mediated changes in β2-AR expression and functioning under chronic inflammatory

conditions is imperative to provide rational neural-immune based therapies for the prevention or

treatment of chronic inflammatory diseases.

Sympathetic modulation of innate immunity has been much less studied than adaptive

immunity. The role of 1-ARs is and their cross-talk with β2-AR in innate immunity, particularly,

DCs, is poorly understood. We know that the intracellular pathways activated by AR stimulation

can influence the activation of immune cells by modifying cell surface molecules critical for

antigen detection and processing/presentation in APCs, and interface with intracellular

pathways used by TLRs, TCRs and BCRs at multiple sites. Diversity in the responses by β-ARs

may result from the presence of multiple isoforms of ACs and PKA, and from promiscuity in

receptor coupling to either Gs or Gi, or uncoupling from ACs to other signaling pathways. Lipid

rafts play an important role in AC-mediated actions. The recent discovery of biased β2-AR

agonists that preferentially signal via Gs or Gi will be useful tools to decipher how this

differential signaling impacts immune responses. Similarly, there are two G-proteins to which α-

ARs can bind and Gq coupling activates three intracellular pathways, each of them interfacing

with signaling pathways that mediate immune cell responses. Collectively, our current

understanding of how and where sympathetically-activated intracellular pathways in immune

Page 41: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

40

cells converge on to those activated with immune stimuli is sparse. Additional biomolecular

research is needed to fully understand how catecholamine-mediated changes in the functional

state of specific immune cells populations that interact via “immunological synapses” across time

and under well-defined microenvironments. With this knowledge we can begin to make

reasonable predictions about local and systemic outcomes in vivo under normal and disease

conditions. Only then will it be possible to design new diagnostic pharmaceuticals tools that are

both safe and efficacious, and are truly tailored for the prevention or successfully treatment of

the underlying causes of inflammatory diseases, rather than the treatment of their symptoms

and/or retarding their progression.

References Cited

Ader, R., Cohen, N., 1982. Behaviorally conditioned immunosuppression and murine systemic

lupus erythematosus. Science 215, 1534–1536.

Aoki, E., Kasahara, T., Hagiwara, H., Sunaga, M., Hisamitsu, N., Hisamitsu T., 2005.

Electroacupuncture and moxibustion influence the lipopolysaccharide-induced TNF-alpha

production by macrophages. In Vivo 19, 495–500.

Alaniz, R.C., Thomas, S.A., Perez-Melgosa, M., Mueller, K., Farr, A.G., Palmiter, R.D., Wilson,

C.B., 1999. Dopamine beta-hydroxylase deficiency impairs cellular immunity. Proc. Natl.

Acad. Sci. U.S.A. 96, 2274–2278.

Altenburg, S.P., Martins, M.A., Silva, A.R., Cordeiro, R.S., Castro-Faria-Neto, H.C., 1997. LPS-

induced blood neutrophilia is inhibited by α1-adrenoceptor antagonists: a role for

catecholamines. J. Leukoc. Biol. 61, 689–694.

Antonica, A., Magni, F., Mearini, L, Paolocci, N., 1994. Vagal control of lymphocyte release from

rat thymus. J. Auton. Nerv. Syst., 48, 187–197.

Badoer, E., 2001. Hypothalamic paraventricular nucleus and cardiovascular regulation. Clin.

Exp. Pharmacol. Physiol. 28, 95–99.

Baranski, G.M., Offin, M.D., Sifri, Z.C., Elhassan, I.O., Hannoush, E.J., Alzate, W.D.,

Rameshwar, P., Livingston, D.H., Mohr, A.M., 2011. β-blockade protection of bone marrow

following trauma: the role of G-CSF. J. Surg. Res. 170, 325–331.

Barnett, C.C., Moore, E.E., Partrick, D.A, Silliman, C.C., 1997. Beta-adrenergic stimulation

down-regulates neutrophil priming for superoxide generation, but not elastase release. J.

Surg. Res. 70, 166–170.

Beiermeister, K.A., Keck, B.M., Sifri, Z.C., ElHassan, I.O., Hannoush, E.J., Alzate, W.D.,

Rameshwar, P., Livingston, D.H., Mohr, A.M., 2010. Hematopoietic progenitor cell

mobilization is mediated through beta-2 and beta-3 receptors after injury. J. Trauma 69,

338–343.

Page 42: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

41

Bellinger, D.L., Felten, S.Y., Collier, T.J., Felten, D.L. 1987. Noradrenergic sympathetic

innervation of the spleen: IV. Morphometric analysis in adult and aged F344 rats. J.

Neurosci. Res. 18, 55–63.

Bellinger, D.L., Felten, S.Y., Felten, D.L., 1988. Maintenance of noradrenergic sympathetic

innervation in the involuted thymus of the aged Fischer 344 rat. Brain Behav. Immun. 2,

133–150.

Bellinger, Denise L., Felten, Suzanne Y., Ackerman, Kurt D., Lorton, Dianne, Madden, Kelly S.,

and Felten, David L. 1992a. “Noradrenergic sympathetic innervation of lymphoid organs

during development, aging, and in immune disease.” In: Aging of the Autonomic Nervous

System, edited by Amenta, F., pp. 243–284. Boca Raton, FL: CRC Press.

Bellinger, D.L., Lorton, D., Felten, S.Y., Felten, D.L., 1992b. Innervation of lymphoid organs and

implications in development, aging, and autoimmunity. Int. J. Immunopharmacol. 14, 329–

344.

Bellinger, D.L., Lorton, D., Hamill, R.W., Felten, S.Y., Felten, D.L., 1993. Acetylcholinesterase

staining and choline acetyltransferase activity in the young adult rat spleen: lack of evidence

for cholinergic innervation. Brain Behav. Immun. 7, 191–204.

Bellinger, D.L., Millar, B.A., Perez, S., Carter, J., Wood, C., ThyagaRajan, S., Molinaro, C.,

Lubahn, C., Lorton, D., 2008a. Sympathetic modulation of immunity: relevance to disease.

Cell. Immunol. 252, 27–56.

Bellinger, D.L., Silva, D., Millar, A.B., Molinaro, C., Ghamsary, M., Carter, J., Perez, S., Lorton,

D., Lubahn, C., Araujoa, G., Thyagarajan, S., 2008b. Sympathetic nervous system and

lymphocyte proliferation in the Fischer 344 rat spleen: a longitudinal study.

Neuroimmunomodulation 15, 260–271

Bellinger, D.L., Nance, D.M., Lorton, D. 2013. Innervation of the Immune System. In: The Wiley-

Blackwell Handbook of Psychoneuroimmunology, First Edition. Kusnecov A.W., Anisman,

H., Eds. John Wiley & Sons, Ltd., pp. 24-49.

Beluli, D.J., Weaver, L.C., 1991a. Areas of rostral medulla providing tonic control of renal and

splenic nerves. Am. J. Physiol. 261, H1687–H1692.

Beluli D.J., Weaver, L.C., 1991b. Differential control of renal and splenic nerves without

medullary topography. Am. J. Physiol. 260, H1072–H1079.

Bender, B.S., Bell, W.E., Taylor, S., Scarpace, P.J. 1993. 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. 264, 1381–1386.

Benschop, R.J., Rodriguez-Feuerhahn, M., Schedlowski, M., 1996. Catecholamine-induced

leukocytosis: early observations, current research, and future directions. Brain Behav.

Page 43: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

42

Immun. 10, 77–91.

Benschop, R.J., Schedlowski, M., Wienecke, H., Jacobs, R., Schmidt, R.E., 1997. Adrenergic

control of natural killer cell circulation and adhesion. Brain Behav. Immun. 11, 321–332.

Bergquist, J., Josefsson, E., Tarkowski, A., Ekman, R., Ewing, A., 1997. Measurements of

catecholamine-mediated apoptosis of immunocompetent cells by capillary electrophoresis.

Electrophoresis 18, 1760–176.

Bermudez, L.E., Wu, M., Young, L.S., 1990. Effect of stress-related hormones on macrophage

receptors and response to tumor necrosis factor. Lymphokine Res. 9, 137–145.

Bhowmick, S., Singh, A., Flavell, R.A., Clark, R.B., O'Rourke, J., Cone, R.E., 2009. The

sympathetic nervous system modulates CD4(+)FoxP3(+) regulatory T cells via a TGF-beta-

dependent mechanism. J. Leukoc. Biol. 86, 1275–1283.

Bissonnette, E.Y., Befus, A.D., 1997. Anti-inflammatory effect of β2-agonists: inhibition of TNF-α

release from human mast cells. J. Allergy Clin. Immunol. 100, 825–831.

Blotta, M.H., DeKruyff, R.H., Umetsu, D.T., 1997. Corticosteroids inhibit IL-12 production in

human monocytes and enhance their capacity to induce IL-4 synthesis in CD4+

lymphocytes. J. Immunol. 158, 5589–5595.

Borger, P., Hoekstra, Y., Esselink, M.T., Postma, D.S., Zaagsma, J., Vellenga, E., Kauffman,

H.F., 1998. Beta-adrenoceptor-mediated inhibition of IFN-gamma, IL-3, and GM-CSF mRNA

accumulation in activated human T lymphocytes is solely mediated by the beta2-

adrenoceptor subtype. Am. J. Respir. Cell. Mol. Biol. 19, 400–407.

Borriello, F., Sethna, M.P., Boyd, S.D., Schweitzer, A.N., Tivol, E.A., Jacoby D., Strom, T.B.,

Simpson, E.M., Freeman, G.J., Sharpe, A.H., 1997. B7-1 and B7-2 have overlapping, critical

roles in immunoglobulin class switching and germinal center formation. Immunity 6, 303–

313.

Botta, F., Maestroni, G.J., 2008. Adrenergic modulation of dendritic cell cancer vaccine in a

mouse model: role of dendritic cell maturation. J. Immunother. 31, 263–270.

Boulet, L-P., Turcotte, H., Boutet, M., Dubé, J., Gagnon, M., Laviolette, M., 1997. Influence of

salmeterol on chronic and allergen-induced airway inflammation in mild allergic asthma: a

pilot study. Curr. Ther. Res. 58, 240–259.

Bovbjerg, D., Cohen, N., Ader. R., 1987. Behaviorally conditioned enhancement of delayed-type

hypersensitivity in the mouse. Brain Behav. Immun. 1, 64–71.

Brady, L.S., Lynn, A.B., Herkenham, M., Gottesfeld, Z., 1994. Systemic interleukin-1 induces

early and late patterns of c-fos mRNA expression in brain. J. Neurosci. 14, 4951–4964.

Bratton, B.O., Martelli, D., McKinley, J.J., Travaks, D., Anderson, C.R., McAllen, R.M., 2012.

Neural regulation of inflammation: No neural connection from the vagus to splenic sympathetic

neurons. Exp. Physiol. 97, 1180–1185.

Page 44: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

43

Brooks, W.H., Cross, R.J., Roszman, T.L., Markesbery, W.R., 1982. Neuroimmunomodulation:

neural anatomical basis for impairment and facilitation. Ann. Neurol. 12, 56–61.

Brown G.L., 1964. The Croonian Lecture 1964. The release and fate of the transmitter liberated

by adrenergic nerves. Proc. R. Soc. B 162, 1–19.

Brown R., Li z., Vriend C.Y., Nirula, R. Janz L., Falk, J., Nance, D.M. Dyck, Dg, Greenberg,

1991. Suppression of splenic macrophage interleukin-1 secretion following

Intracerebroventricular injection of interleukin-1beta: evidence for pituitary-adrenal and

sympathetic control. Cell. Immunol. 132, 84–93.

Burýsek, L., Houstek, J., 1997. Beta-Adrenergic stimulation of interleukin-1alpha and

interleukin-6 expression in mouse brown adipocytes. FEBS. Lett. 411, 83–86.

Butchers, P.R., Vardey, C.J., Johnson, M., 1991. Salmeterol: a potent and long-acting inhibitor

of inflammatory mediator release from human lung. Br. J. Pharmacol. 104, 672–676.

Cano, G., Card, J.P., Rinaman, L., Sved, A.F., 2000. Connections of Barrington's nucleus to the

sympathetic nervous system in rats. J. Auton. Nerv Syst. 79, 117–128.

Cano, G., Sved, A.F., Rinaman, L., Rabin, B.S., Card, J.P., 2001. Characterization of the central

nervous system innervation of the rat spleen using viral transneuronal tracing. J. Comp.

Neurol. 439, 1–18.

Carlson, S.L., Albers, K.M., Beiting, D.J., Parish, M., Conner, J.M., Davis, B.M. 1995. NGF

modulates sympathetic innervation of lymphoid tissues. J. Neurosci. 15, 5892–5899.

Caroleo, M.C., Pulvirenti, L., Arbitrio, M., Lopilato, R., Nisticò, G., 1993. Evidence that CRH

microinfused into the locus coeruleus decreases cell-mediated immune response in rats.

Funct. Neurol. 8, 271–277.

Casale, T.B., Kaliner, M., 1984. Demonstration that circulating human blood cells have no

detectable α1 adrenergic receptors by radioligand binding analysis. J. Allergy Clin. Immunol.

74, 812–818.

Cervi, Lukewich & Lomax 2014. Autonomic Neuroscience

Chao, H.J., Hsu, Y.C., Yuan, H.P., Jiang, H.S., Hsueh, C.M., 2005. The conditioned

enhancement of neutrophil activity is catecholamine dependent. J. Neuroimmunol. 158,

159–169.

Chazan R, Walajtys-Rode E, Grubek-Jaworska H, Madalinska M, Droszcz W. 1991. In vitro and

in vivo effect of salbutamol on histamine release from human basophils. Int. J. Clin.

Pharmacol. Ther. Toxicol. 29, 9–13.

Chen, X., Herbert, J., 1995. Regional changes in c-fos expression in the basal forebrain and

brainstem during adaptation to repeated stress: correlations with cardiovascular,

hypothermic and endocrine responses. Neuroscience 64, 675–685.

Page 45: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

44

Chong, L.K., Morice, A.H., Yeo, W.W., Schleimer, R.P., Peachell, P.T., 1995. Functional

desensitization of beta agonist responses in human lung mast cells. Am. J. Respir. Cell. Mol.

Biol. 13, 540–546.

Chong L.K., Peachell P.T., 1999. Beta-adrenoceptor reserve in human lung: a comparison

between airway smooth muscle and mast cells. Eur. J. Pharmacol., 378, 115–122.

Chong L.K., Suvarna K., Chess-Williams R., Peachell P. T., 2003. Desensitization of beta2-

adrenoceptor-mediated responses by short-acting beta2-adrenoceptor agonists in human

lung mast cells. Br. J. Pharmacol. 138, 512–520.

Chou, R.C., Stinson, M.W., Noble, B.K., Spengler, R.N., 1996. Beta-adrenergic receptor

regulation of macrophage-derived tumor necrosis factor-alpha production from rats with

experimental arthritis. J. Neuroimmunol. 67, 7–16.

Claassen, D.E., Fels, R.J., Kenney, M.J., 1998. Altered frequency characteristics of sympathetic

nerve activity after sustained elevation in arterial pressure. Am. J. Physiol. 274, R694-R703.

Cobelens, P.M., Kavelaars, A., Vroon, A., Ringeling, M., van der Zee, R., van Eden, W.,

Heijnen, C.J., 2002. The beta 2-adrenergic agonist salbutamol potentiates oral induction of

tolerance, suppressing adjuvant arthritis and antigen-specific immunity. J. Immunol. 169,

5028–5035.

Cohen, N., Moynihan, J.A., Ader, R., Pavlovian conditioning of the immune system. Int. Arch.

Allergy Immunol. 1994 105, 101–106.

Cook-Mills, J.M., Mokyr, M.B., Cohen, R.L., Perlman, R.L., Chambers, D.A., 1995.

Neurotransmitter suppression of the in vitro generation of a cytotoxic T lymphocyte response

against the syngeneic MOPC-315 plasmacytoma. Cancer Immunol. Immunother. 40, 79–87.

Cross, R.J., Brooks, W.H., Roszman, T.L., Markesbery, W.R., 1982. Hypothalamic–immune

interactions. Effect of hypophysectomy on neuroimmunomodulation. J. Neurol. Sci. 53, 557–

566.

Cross, R.J., Markesbery, W.R., Brooks, W.H., Roszman, T.L., 1984. Hypothalamic–immune

interactions: neuromodulation of natural killer activity by lesioning of the anterior

hypothalamus. Immunology, 51, 399–405.

Cunningham, E.T. Jr., Miselis, R.R., Sawchenko, P.E., 1994. The relationship of efferent

projections from the area postrema to vagal motor and brain stem catecholamine-containing

cell groups: an axonal transport and immunohistochemical study in the rat. Neuroscience

58, 635–648.

Daaka, Y., Luttrell, L.M., Lefkowitz, R.J., 1997. Switching of the coupling of the beta2-adrenergic

receptor to different G proteins by protein kinase A. Nature (Lond.) 390, 88–91.

Dampney, R.A., 1994. Functional organization of central pathways regulating the cardiovascular

system. Physiol. Rev. 74, 323–364.

Page 46: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

45

Dantzer, R., 2001. Cytokine-induced sickness behavior: mechanisms and implications. Ann N.Y.

Acad. Sci. 933, 222–234.

Dar, A., Schajnovitz, A., Lapid, K., Kalinkovich, A., Itkin, T., Ludin, A., Kao, W.M., Battista, M.,

Tesio, M., Kollet, O., Cohen, N.N., Margalit, R., Buss, E.C., Baleux, F., Oishi, S., Fujii, N.,

Larochelle, A., Dunbar, C.E., Broxmeyer, H.E., Frenette, P.S., Lapidot, T., 2011. Rapid

mobilization of hematopoietic progenitors by AMD3100 and catecholamines is mediated by

CXCR4-dependent SDF-1 release from bone marrow stromal cells. Leukemia 25, 1286–

1296.

Deering, J., Coote, J.H., 2000. Paraventricular neurones elicit a volume expansion-like change

of activity in sympathetic nerves to the heart and kidney in the rabbit. Exp. Physiol. 85. 2,

177–186.

del Rey, A., Roggero, E., Kabiersch, A., Schäfer, M., Besedovsky, H.O. 2006. The role of

noradrenergic nerves in the development of the lymphoproliferative disease in Fas-deficient,

lpr/lpr mice. J. Immunol., 176, 7079–7086.

Dimitrov, S., Lange, T., Born, J., 2010. Selective mobilization of cytotoxic leukocytes by

epinephrine. J. Immunol. 184, 503–511.

Dokur, M., Boyadjieva, N., Sarkar, D.K., 2004. Catecholaminergic control of NK cell cytolytic

activity regulatory factors in the spleen. J. Neuroimmunol. 151, 148–157.

Dresch, C., Minc, J., Poirier, O., Bouvet, D., 1981. Effect of beta adrenergic agonists and beta

blocking agents on hemopoiesis in human bone marrow. Biomedicine 34, 93–98.

Dresch, C., Minc, J., Mary, J.Y., 1984. In vivo protection of normal mouse hematopoiesis by a

beta 2 blocking agent during S-phase chemotherapy. Cancer Res. 44, 493–497.

Duffy, S.M., Cruse, G., Lawley, W.J., Bradding, P., 2005. Beta2-adrenoceptor regulation of the

K+ channel iKCa1 in human mast cells. FASEB J. 19, 1006–1008.

Ebbinghaus, M., Gajda, M., Boettger, M.K., Schaible, H.G., Bräuer, R., 2012. The anti-

inflammatory effects of sympathectomy in murine antigen-induced arthritis are associated

with a reduction of Th1 and Th17 responses. Ann. Rheum. Dis. 71, 253–261.

Eberl, G., Marmon, S., Sunshine, M.J., Rennert, P.D., Choi, Y., Littman, D.R., 2004. An

essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid

tissue inducer cells. Nat. Immunol. 5, 64–73.

Ebong, S.J., Goyert, S.M., Nemzek, J.A., Kim, J., Bolgos, G.L., Remick, D.G., 2001. Critical role

of CD14 for production of proinflammatory cytokines and cytokine inhibitors during sepsis

with failure to alter morbidity or mortality. Infect Immun. 69, 2099–2106.

Elenkov, I.J., D.A. Papanicolaou, R.L. Wilder & G.P. Chrousos. 1996. Modulatory effects of

glucocorticoids and catecholamines on human interleukin-12 and interleukin-10 production:

clinical implications. Proc. Assoc. Am. Physicians 108: 374–381.

Page 47: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

46

Elenkov, I.J., Haskó, G., Kovács, K.J., Vizi, E.S., 1995. Modulation of lipopolysaccharide

induced tumor necrosis factor-alpha production by selective alpha- and beta-adrenergic

drugs in mice. J. Neuroimmunol. 61, 123–131.

Elenkov, I.J., Chrousos, G.P., 1999. Stress hormones, Th1/Th2 patterns, pro/antiinflammatory

cytokines and susceptibility to disease. Trends Endocrinol. Metab. 10, 359–368.

Elenkov, I.J., Wilder, R.L., Chrousos, G.P., Vizi, E.S., 2000. The sympathetic nerve – an

integrative interface between two supersystems: the brain and the immune system.

Pharmacol. Rev. 52, 595–638.

Elenkov IJ, Wilder RL, Bakalov VK, Link AA, Dimitrov MA, Fisher S, Crane M, Kanik KS,

Chrousos GP. 2001. IL-12, TNF-alpha, and hormonal changes during late pregnancy and

early postpartum: implications for autoimmune disease activity during these times. J Clin

Endocrinol. Metab. 86, 4933–4938.

Elhassan, I.O., Hannoush, E.J., Sifri, Z.C., Jones, E., Alzate, W.D., Rameshwar, P., Livingston,

D.H., Mohr, A.M., 2011. Beta-blockade prevents hematopoietic progenitor cell suppression

after hemorrhagic shock. Surg. Infect. 12, 273–278.

Elmquist, J.K., Saper, C.B., 1996. Activation of neurons projecting to the paraventricular

hypothalamic nucleus by intravenous lipopolysaccharide. J. Comp. Neurol. 374, 315–331.

Engler, H., Doenlen, R., Riether, C., Engler, A., Besedovsky, H.O., Del Rey, A., Pacheco-López,

G., Schedlowski, M., 2010. Chemical destruction of brain noradrenergic neurons affects

splenic cytokine production. J. Neuroimmunol. 219, 75–80.

Ericsson, A., Kovacs, K.J., Sawchenko, P.E., 1994. A functional anatomical analysis of central

pathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons.

J. Neurosci. 14, 897–913.

Ericsson, A., Arias, C., Sawchenko, P.E., 1997. Evidence for an intramedullary prostaglandin-

dependent mechanism in the activation of stress-related neuroendocrine circuitry by

intravenous interleukin-1. J. Neurosci. 17, 7166–7179

Ericsson, A., Liu, C., Hart, R.P., Sawchenko, P.E., 1995. Type 1 interleukin-1 receptor in the rat

brain: distribution, regulation, and relationship to sites of IL-1-induced cellular activation. J.

Comp. Neurol. 361, 681–698.

Ermirio, R., Ruggeri, P., Molinari, C., Weaver, L.C., 1993. Somatic and visceral inputs to

neurons of the rostral ventrolateral medulla. Am. J. Physiol. 265(1 Pt 2), R35–R40.

Farkas, E., Jansen, A.S., Loewy, A.D., 1998. Periaqueductal gray matter input to cardiac-related

sympathetic premotor neurons. Brain Res. 792, 179–192.

Felderhoff-Mueser, U., Schmidt, O.I., Oberholzer, A., Bührer, C., Stahel, P.F., 2005. IL-18: a key

player in neuroinflammation and neurodegeneration? Trends Neurosci. 28, 487–493.

Page 48: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

47

Felten, D.L., Cohen, N., Ader, R., Felten, S.Y, Carlson, S.L., Roszman, T.L. 1991. Central

neural circuits involved in neural-immune interactions. In: Psychoneuroimmunology, 2nd

edition, Ader, R., Felten, D.L., Cohen, N. San Diego, CA: Academic Press, pp. 3–25.

Felten D.L., Felten, S.Y., Carlson, S.L., Olschowka, J.A, and Livnat, S. 1985. Noradrenergic and

peptidergic innervation of lymphoid tissue. J. Immunol. 135, 755s–765s.

Felten, D.L., Livnat, S., Felten, S.Y., Carlson, S.L., Bellinger, D.L., Yeh, P. 1984. Sympathetic

innervation of lymph nodes in mice. Brain Res. Bull. 13, 693–699.

Felten, D.L, Overhage, J.M., Felten, S.Y., Schmedtje, J.F. 1981. Noradrenergic sympathetic

innervation of lymphoid tissue in the rabbit appendix: further evidence for a link between the

nervous and immune systems. Brain Res. Bull., 7, 595–612.

Felten, S.Y., Olschowka, J., 1987. Noradrenergic sympathetic innervation of the spleen: II.

Tyrosine hydroxylase (TH)-positive nerve terminals form synaptic-like contacts on

lymphocytes in the splenic white pulp. J. Neurosci. Res. 18, 37–48.

Fletcher, A.L., Lukacs-Kornek, V., Reynoso, E.D., Pinner, S.E., Bellemare-Pelletier, A., Curry,

M.S., Collier, A.R., Boyd, R.L., Turley, S.J., 2010. Lymph node fibroblastic reticular cells

directly present peripheral tissue antigen under steady-state and inflammatory conditions. J.

Exp. Med. 207, 689–697.

Fonseca, R.B., Mohr, A.M., Wang, L., Sifri, Z.C., Rameshwar, P., Livingston, D.H., 2005. The

impact of a hypercatecholamine state on erythropoiesis following severe injury and the role

of IL-6. J. Trauma 59, 884–889, discussion 889–890.

Franceschini, D., Orr-Urtreger, A., Yu, W., Mackey, L.Y., Bond, R.A., Armstrong, D., Patrick,

J.W., Beaudet, A.L., De Biasi, M., 2000. Altered baroreflrex responses in alpha7 deficient

mice. Behav. Brain Res. 113, 3–10.

Freier, E., Weber, C.S., Nowottne, U., Horn, C., Bartels, K., Meyer, S., Hildebrandt, Y.,

Luetkens, T., Cao, Y., Pabst, C., Muzzulini, J., Schnee, B., Brunner-Weinzierl, M.C.,

Marangolo, M., Bokemeyer, C., Deter, H.C., Atanackovic, D., 2010. Decrease of

CD4(+)FOXP3(+) T regulatory cells in the peripheral blood of human subjects undergoing a

mental stressor. Psychoneuroendocrinology 35, 663–673.

Furukawa, H., Yamashita, A., del Rey, A., Besedovsky, H., 2004. c-Fos expression in the rat

cerebral cortex during systemic GvH reaction. Neuroimmunomodulation 11, 425–433.

Giang, D.W., Goodman, A.D., Schiffer, R.B., Mattson, D.H., Petrie, M., Cohen, N., Ader, R.,

1996. Conditioning of cyclophosphamide-induced leukopenia in humans. J. Neuropsychiatry

Clin. Neurosci. 8, 194–201.

Gibson-Berry, K.L., Whitin JC and Cohen HJ (1993) Modulation of the respiratory burst in

human neutrophils by isoproterenol and dibutyryl cyclic AMP. J Neuroimmunol 43, 59–68.

Goyarts, E., Matsui, M., Mammone, T., Bender, A.M., Wagner, J.A., Maes, D., Granstein, R.D.

Page 49: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

48

(2008) Norepinephrine modulates human dendritic cell activation by altering cytokine

release. Exp. Dermatol. 17, 188–196

Guereschi, M.G., Araujo, L.P., Maricato, J.T., Takenaka, M.C., Nascimento, V.M., Vivanco,

B.C., Reis, V.O., Keller, A.C., Brum, P.C., Basso, A.S., 2013. Beta2-adrenergic receptor

signaling in CD4(+) Foxp3(+) regulatory T cells enhances their suppressive function in a

PKA-dependent manner. Eur J Immunol. 43, 1001–1012.

Harvath, L., Robbins, J.D., Russell, A.A., Seamon, K.B. 1991. cAMP and human neutrophil

chemotaxis. Elevation of cAMP differentially affects chemotactic responsiveness. J.

Immunol. 146, 224–232.

Haskó, G., Szabo, C., Nemeth, Z.H., Salzman, A.L., Vizi, E.S., 1998. Stimulation of beta-

adrenoceptors inhibits endotoxin-induced IL-12 production in normal and IL-10 deficient

mice. J. Neuroimmunol. 88, 57–61.

Hatfield, S.M., Petersen, B.H., DiMicco, J.A., 1986. Beta adrenoceptor modulation of the

generation of murine cytotoxic T lymphocytes in vitro. J. Pharmacol. Exp. Ther. 239, 460–

466.

Hayes, K., Weaver, L.C., 1990. Selective control of sympathetic pathways to the kidney, spleen

and intestine by the ventrolateral medulla in rats. J. Physiol. 428, 371–385.

Hayes, K., Weaver, L.C., 1992. Tonic sympathetic excitation and vasomotor control from

pontine reticular neurons. Am. J. Physiol. 263, H1567–H1575.

Heijnen, C.J., Rouppe van der Voort, C., van de Pol, M., Kavelaars, A., 2002. Cytokines

regulate α1-adrenergic receptor mRNA expression in human monocytic cells and endothelial

cells. J. Neuroimmunol., 125, 66–72.

Hellstrand, K., Hermodsson, S., 1989. An immunopharmacological analysis of adrenaline-

induced suppression of human natural killer cell cytotoxicity. Int. Arch. Allergy Appl.

Immunol. 89, 334–341.

Helwig, B.G., Parimi, S., Ganta, C.K., Cober, R., Fels, R.J., Kenney, M.J., 2006. Aging alters

regulation of visceral sympathetic nerve responses to acute hypothermia. Am. J. Physiol.

Regul. Integr. Comp. Physiol. 291, R573–R579.

Herman, F.F., Goldberg, B.J., Lad, P.M., Neidzin, H., Kaplan, M., Easton, J.G., 1990. Effects of

histamine on alpha adrenergic receptor expression on the lymphocytes of normal and

asthmatic subjects. Ann. Allergy 65, 32–36.

Herman, J.P., Eyigor, O., Ziegler, D.R., Jennes, L., 2000. Expression of ionotropic glutamate

receptor subunit mRNAs in the hypothalamic paraventricular nucleus of the rat. J. Comp.

Neurol. 422, 352–362.

Hervé, J., Dubreil, L., Tardif, V., Terme, M., Pogu, S., Anegon, I., Rozec, B., Gauthier, C., Bach,

J.M., Blancou, P., 2013. β2-Adrenoreceptor agonist inhibits antigen cross-presentation by

Page 50: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

49

dendritic cells. J. Immunol. 190, 3163–3171.

Hetier, E., Ayala, J., Bousseau, A., Prochiantz, A., 1991. Modulation of interleukin-1 and tumor

necrosis factor expression by beta-adrenergic agonists in mouse ameboid microglial cells.

Exp. Brain Res. 86, 407–413.

Hilbert, T., Bongartz, J., Weisheit, C., Knüfermann, P., Baumgarten, G., Hoeft, A., Poth, J.M.,

2013. Beta2-adrenoceptor stimulation suppresses TLR9-dependent ifna1 secretion in

human peripheral blood mononuclear cells. PLoS One 8(5), e65024.

Hosking, K.G., Fels, R.J., Kenney, M.J., 2009. Inhibition of RVLM synaptic activation at peak

hyperthermia reduces visceral sympathetic nerve discharge. Auton. Neurosci. 150, 104–

110.

Howarth, P.H., Durham, S.R., Lee, T.H., Kay, A.B., Church, M.K., Holgate, S.T. 1982. Influence

of albuterol, cromolyn sodium and ipratropium bromide on the airway and circulating

mediator response to allergen bronchial provocation in asthma. Am. Rev. Respir. Dis. 132,

986–992.

Hu, Z., Chen, R., Cai, Z., Yu, L., Fei, Y., Weng, L., Wang, J., Ge, X., Zhu, T., Wang, J., Bai, C.,

2012. Salmeterol attenuates the inflammatory response in asthma and decreases the pro-

inflammatory cytokine secretion of dendritic cells. Cell. Mol. Immunol. 9, 267–275.

Hueber, A.O., Bernard, A.M., Battari, C.L., Marguet, D., Massol, P., Foa, C., Brun, N., Garcia,

S., Stewart, C., Pierres, M., He, H.T., 1997. Thymocytes in Thy-1-/- mice show augmented

TCR signaling and impaired differentiation. Curr. Biol., 7, 705–708.

Imaki, T., Nahan, J.L., Rivier, C., Sawchenko, P.E., Vale, W., 1991. Differential regulation of

corticotropin-releasing factor mRNA in rat brain regions by glucocorticoids and stress. J.

Neurosci. 11, 585–599.

Imaki, T., Shibasaki T., Hotta, M., Demura, H., 1993. Intracerebroventricular administration of

corticotropin-releasing factor induces c-fos mRNA expression in brain regions related to

stress responses: comparison with pattern of c-fos mRNA induction after stress. Brain Res.

616, 114–125.

Irwin, M., 1994. Stress-induced immune suppression: role of brain corticotropin releasing

hormone and autonomic nervous system mechanisms. Adv. Neuroimmunol. 4, 29–47.

Jänig, W., McLachlan E.M., 2013. Neurobiology of the autonomic nervous system. In: Mathias,

C.J., Bannister, R. (Eds.) Autonomic Failure. 5th edition, Oxford University Press, NY Oxford,

pp. 21-34.

Jansen, A.S., Farwell, D.G., Loewy, A.D., 1993. Specificity of pseudorabies virus as a

retrograde marker of sympathetic preganglionic neurons: implications for transneuronal

labeling studies. Brain Res. 617, 103–112.

Javierre, M.Q., Pinto, L.V., Lima, A.O., Sassine, W.A., 1975. Immunologic phagocytosis by

Page 51: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

50

macrophages: effect by stimulation of alpha adrenergic receptors. Rev. Bras. Pesqui. Med.

Biol. 8, 271–274.

Johnson, M., 2002. Effects of β2-agonists on resident and infiltrating inflammatory cells. J.

Allergy Clin. Immunol. 110, S262–S290.

Jondal, M., Pazirandeh, A., Okret, S., 2004. Different roles for glucocorticoids in thymocyte

homeostasis? Trends Immunol. 25, 595–600.

Kalinichenko, V.V., Mokyr, M.B., Graf, L.H. Jr., Cohen, R.L., Chambers, D.A., 1999.

Norepinephrine-mediated inhibition of antitumor cytotoxic T lymphocyte generation involves

a beta-adrenergic receptor mechanism and decreased TNF-alpha gene expression. J.

Immunol. 163, 2492–2499.

Kanemi, O., Zhang, X., Sakamoto, Y., Ebina M., Nagatomi, R., 2005. Acute stress reduces

intraparenchymal lung natural killer cells via beta-adrenergic stimulation. Clin. Exp. Immunol.

139, 25–34.

Kannan, H., Hayashida, Y., Yamashita, H., 1989. Increase in sympathetic outflow by

paraventricular nucleus stimulation in awake rats. Am. J. Physiol. Regulatory Integrative

Comp. Physiol. 256, R1325–R1330.

Kasprowicz, D.J., Kohm, A.P., Berton, M.T., Chruscinski, A.J., Sharpe, A., Sanders, V.M., 2000.

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. 165, 680–

690.

Katafuchi, T., Oomura, Y., Kurosawa, M., 1988. Effects of chemical stimulation of

paraventricular nucleus on adrenal and renal nerve activity in rats. Neurosci. Lett. 86, 195–

200.

Katafuchi, T., Take, S., Hori, T., 1993a. Roles of sympathetic nervous system in the

suppression of cytotoxicity of splenic natural killer cells in the rat. J. Physiol. (Lond.) 465,

343–357.

Katafuchi, T., Ichijo, T., Take, S., Hori, T., 1993b. Hypothalamic modulation of splenic natural

killer cell activity in rats. J. Physiol. 471, 209–221.

Katayama, Y., Battista, M., Kao, W.M., Hidalgo, A., Peired, A.J., Thomas, S.A., Frenette, P.S.,

2006. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress

from bone marrow. Cell 124, 407–421.

Kavelaars, A., 2002. Regulated expression of alpha-1 adrenergic receptors in the immune

system. Brain Behav. Immun. 16, 799–807.

Kemeny, M.E., Schedlowski, M., 2007. Understanding the interaction between psychosocial

stress and immune-related diseases: a stepwise progression. Brain Behav. Immun. 21,

1009–1018.

Page 52: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

51

Kendall, M.D., Atkinson, B.A., Muñoz, F.J., de la Riva, C., Clarke, A.G., von Gaudecker, B.,

1994. The noradrenergic innervation of the rat thymus during pregnancy and in the

postpartum period. J. Anat. 185, 617–625.

Kenney, M.J. 1994. Frequency characteristics of sympathetic nerve discharge in anesthetized

rats. Am J Physiol Regulatory Integrative Comp Physiol 267, R830–R840.

Kenney, M.J., Claassen, D.E., Fels, R.J., Saindon, C.S. 1999. Cold stress alters characteristics

of sympathetic nerve discharge bursts. J. Appl. Physiol. 87, 732–742.

Kenney, M.J., Blecha, F., Morgan, D.A., Fels, R.J., 2001a. Interleukin-1 beta alters brown

adipose tissue but not renal sympathetic nerve responses to hypothermia. Am. J. Physiol.

Heart Circ. Physiol. 281, H2441–H2445.

Kenney, M.J., Weiss, M.L., Patel, K.P., Wang, Y., Fels, R.J., 2001b. Paraventricular nucleus

bicuculline alters frequency components of sympathetic nerve discharge bursts. Am. J.

Physiol. Heart Circ. Physiol. 281, H1233–H1241.

Kenney, M.J., Blecha, F., Fels, R.J., Morgan, D.A., 2002. Altered frequency responses of

sympathetic nerve discharge bursts after IL-1beta and mild hypothermia. J. Appl. Physiol.

93, 280–288.

Kenney, M.J., Fels, R.J., 2002. Sympathetic nerve regulation to heating is altered in senescent

rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 283, R513–R520.

Kenney, M.J., Musch, T.I., 2004. Senescence alters blood flow responses to acute heat stress.

Am J Physiol. Heart Circ. Physiol. 286, H1480–H1485.

Kenney, M.J., Weiss, M.L., Mendes, T., Wang, Y., Fels, R.J., 2003a. Role of paraventricular

nucleus in regulation of sympathetic nerve frequency components. Am. J. Physiol. Heart

Circ. Physiol. 284, H1710–H1720.

Kenney, M.J., Weiss, M.L., Haywood, J.R., 2003b. The paraventricular nucleus: an important

component of the central neurocircuitry regulating sympathetic nerve outflow. Acta Physiol.

Scand. 177, 7–15.

Killeen, N., 1997. Thy-1-hiding in full view. Curr. Biol. 7, R774–R777.

Kim, B.J., Jones, H.P., 2010. Epinephrine-primed murine bone marrow-derived dendritic cells

facilitate production of IL-17A and IL-4 but not IFN-γ by CD4+ T cells. Brain Behav. Immun.

24, 1126–1136.

Kleine-Tebbe J, Frank G, Josties C, Kunkel G. 1994. Influence of salmeterol, a long-acting beta

2-adrenoceptor agonist, on IgE-mediated histamine release from human basophils. J.

Investig. Allergol. Clin. Immunol. 4, 12–17.

Klooster, J., Beckers, H.J., Vrensen, G.F., van der Want, J.J., 1993. The peripheral and central

projections of the Edinger-Westphal nucleus in the rat: a light and electron microscopic

tracing study. Brain Res. 632, 260–273.

Page 53: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

52

Koff, W.C., Dunegan, M.A., 1985. Modulation of macrophage-mediated tumoricidal activity by

neuropeptides and neurohormones. J. Immunol. 135, 350–354.

Koff, W.C., Fann, A.V., Dunegan, M.A., Lachman, L.B., 1986. Catecholamine-induced

suppression of interleukin-1 production. Lymphokine Res. 5, 239–247.

Koff, W.C., Dunegan, M.A., 1986. Neuroendocrine hormones suppress macrophage-mediated

lysis of herpes simplex virus-infected cells. J. Immunol. 136, 705–709.

Kohm, A.P., Mozaffarian, A., Sanders, V.M., 2002. B cell receptor- and beta 2-adrenergic

receptor-induced regulation of B7-2 (CD86) expression in B cells. J. Immunol. 168, 6314–

6322.

Kohm, A.P., Sanders, V.M., 1999. Suppression of antigen-specific Th2 cell-dependent IgM and

IgG1 production following norepinephrine depletion in vivo. J. Immunol. 162, 5299–5308.

Kollet, O., Dar, A., Shivtiel, S., Kalinkovich, A., Lapid, K., Sztainberg, Y., Tesio, M., Samstein,

R.M., Goichberg, P., Spiegel, A., Elson, A., Lapidot, T., 2006. Osteoclasts degrade

endosteal components and promote mobilization of hematopoietic progenitor cells. Nat.

Med. 12, 657–664.

Koyama, S., Sato, E., Masubuchi, T., Takamizawa, A., Kubo, K., Nagai, S., Isumi, T., 1999.

Procaterol inhibits IL-1beta- and TNF-alpha-mediated epithelial cell eosinophil chemotactic

activity. Eur. Respir. J. 14, 767–775.

Kroemer G. 1995. The pharmacology of T cell apoptosis. Adv. Immunol. 58, 211–296.

Krukoff, T.L., Harris, K.H., Linetsky, E., Jhamandas, J.H., 1994. Expression of c-fos protein in

rat brain elicited by electrical and chemical stimulation of the hypothalamic paraventricular

nucleus. Neuroendocrinology 59, 590–602.

Kuroki, K., Takahashi, H.K., Iwagaki, H., Murakami, T., Kuinose, M., Hamanaka, S., Minami, K,

Nishibori, M., Tanaka, N., Tanemoto, K., 2004. Beta2-adrenergic receptor stimulation-

induced immunosuppressive effects possibly through down-regulation of co-stimulatory

molecules, ICAM-1, CD40 and CD14 on monocytes. J. Int. Med. Res. 32, 465–483.

Kwon, Y.B., Kim, H.W., Ham, T.W. Yoon, S.Y., Roh, D.H., Han, H.J., Betz, A.J., Yang, I.S., Lee,

J-H. 2003. The anti-inflammatory effect of bee venom stimulation in a mouse air pouch

model is mediated by adrenal medullary activity. J. Neuroendocrinol. 15, 93–96.

LaJevic, M.D., Suleiman, S., Cohen, R.L., Chambers, D.A., 2011. Activation of p38 mitogen-

activated protein kinase by norepinephrine in T-lineage cells. Immunology 132, 197–208.

Leposavić, G., Arsenović-Ranin, N., Radojević, K., Kosec, D., Pešić, V., Vidić-Danković, B.,

Plećas-Solarović, B., Pilipović, I., 2006. Characterization of thymocyte phenotypic alterations

induced by long-lasting beta-adrenoceptor blockade in vivo and its effects on thymocyte

proliferation and apoptosis. Mol. Cell. Biochem. 285, 87–99.

Leposavić, G., Plećas, B., Kosec, D., 2000. Differential effects of chronic propranolol treatment

Page 54: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

53

on the phenotypic profile of thymocytes from immature and adult rats. Immunopharmacology

46, 79–87.

Leposavić, G., Pešić, V., Stojić-Vukanić, Z., Radojević, K., Arsenović-Ranin, N., Kosec, D.,

Perišić, M., Pilipović, I., 2010. Age-associated plasticity of α1-adrenoceptor-mediated tuning

of T-cell development. Exp. Gerontol. 45, 918–935.

Leposavić, G., Perisić, M., 2008. Age-associated remodeling of thymopoiesis: role for gonadal

hormones and catecholamines. Neuroimmunomodulation 15, 290–322.

Lever, J.D., Graham, J.D., Irvine, G., Chick, W.J., 1965. The vesiculated axons in relation to

arteriolar smooth muscle in the pancreas. A. fine structural and quantitative study. J. Anat.

99, 299–313.

Li, Y., Novotny, G.E., 2001. Regional differences in innervation of lymph nodes in the Wistar rat.

J. Anat.199, 735–739.

Ling, Y.L., Meng, A.H., Zhao, X.Y., Shan, B.E., Zhang, J.L., Zhang, X.P., 2001. Effect of

cholecystokinin on cytokines during endotoxic shock in rats. World J. Gastroenterol. 7, 667–

671.

Link, A., Hardie, D.L., Favre, S., Britschgi, M.R., Adams, D.H., Sixt, M., Cyster, J.G., Buckley,

C.D., Luther, S.A., 2011. Association of T-zone reticular networks and conduits with ectopic

lymphoid tissues in mice and humans. Am. J. Pathol. 178, 1662–1675.

Livnat, S., Madden, K.S., Felten, D.L., Felten, S.Y. 1987. Regulation of the immune system by

sympathetic neural mechanisms. Prog. Neuropsychopharmacol. Biol. Psychiatry, 11, 145–

152.

Lochner, M., Ohnmacht, C., Presley, L., Bruhns, P., Si-Tahar, M., Sawa, S., Eberl, G., 2011.

Microbiota-induced tertiary lymphoid tissues aggravate inflammatory disease in the absence

of RORgamma t and LTi cells. J. Exp. Med. 208, 125–134.

Loewy, A.D., 1990. Central autonomic pathways. In: Loewy, A.D., Spyer, K.M, editors. Central

Regulation of Autonomic Functions. New York: Oxford University Press. pp. 88–103.

Lorton, D., Bellinger, D.L., Schaller, J.A. Shewmaker, E., Osredkar T., Lubahn, C., 2013. Altered

Sympathetic-to-immune cell signaling via beta2-adrenergic receptors in adjuvant arthritis

Clin. Dev. Immun., 2013, 764395.

Lorton, D., Lubahn, C., Felten, S.Y., Bellinger, D.L. 1997. Norepinephrine content in primary and

secondary lymphoid organs is altered in rats with adjuvant-induced arthritis. Mech. Ageing

Dev. 94, 145–163. Erratum in: Mech. Ageing Dev. 1997. 98, 181–183.

Lorton, D., Lubahn, C., Sweeney, S., Major, A., Lindquist, C.A., Schaller, J., Washington, C.,

Bellinger, D.L., 2009. Differences in the injury/sprouting response of splenic noradrenergic

nerves in Lewis rats with adjuvant-induced arthritis compared with rats treated with 6-

hydroxydopamine. Brain Behav. Immun. 23, 276–285.

Page 55: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

54

Lorton, D., Lubahn, C., Lindquist, C.A., Schaller, J., Washington, C., Bellinger, D.L. 2005.

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., 489, 260–273.

Loveland, B.E., Jarrott, B., McKenzie, I.F., 1981. The detection of beta-adrenoceptors on murine

lymphocytes. Int. J. Immunopharmacol. 3, 45–55.

Lu, N., Wang, Y., Blecha, F., Fels, R.J., Hoch, H.P., Kenney, M.J., 2003. Central interleukin-

1beta antibody increases renal and splenic sympathetic nerve discharge. Am. J. Physiol.

Heart Circ. Physiol. 284, H1536–H1541.

Lu, X.-Z., Sun, X.-Y. & Yao, T. 1991. Inhibition of renal nerve activity induced by chemical

stimulation of the paraventricular nucleus: mediation of the vasopressinergic spinally-

projecting pathway. Chi. J. Physiol. Sci. 7, 215–221.

Lubahn, C.L., Schaller, J.A., Bellinger, D.L., Sweeney, S., Lorton, D., 2004. The importance of

timing of adrenergic drug delivery in relation to the induction and onset of adjuvant-induced

arthritis. Brain Behav. Immun. 18, 563–571.

Lubbers, T., de Haan, J.J., Luye,r M.D., Verbaeys, I., Hadfoune, M., Dejong, C.H., Buurman,

W.A., Greve, J.W.. 2010. Cholecystokinin/Cholecystokinin-1 receptor-mediated peripheral

activation of the afferent vagus by enteral nutrients attenuates inflammation in rats. Ann.

Surg.. 252, 376–382.

Luitten, P.G., terHorst, G.J., Karst, H., Steffens, A.B., 1985. The course of paraventricular

hypothalamic efferents to autonomic structures in medulla and spinal cord. Brain Res. 329,

374–378.

Luppi, P.H., Sakai, K., Fort, P., Salvert, D., Jouvet, M., 1988. The nuclei of origin of

monoaminergic, peptidergic, and cholinergic afferents to the cat nucleus reticularis

magnocellularis: a double-labeling study with cholera toxin as a retrograde tracer. J. Comp.

Neurol. 277, 1–20.

Luyer, M.D., Greve, J.W., Hadfoune, M., Jacobs, J.A., Dejong, C.H., Buurman, W.A., 2005.

Nutritional stimulation of cholecystokinin receptors inhibits inflammation via the vagus nerve.

J. Exp. Med. 202, 1023–1029.

MacNeil, B.J., Jansen, A.H., Greenberg, A.H., Nance, D.M., 1996. Activation and selectivity of

splenic sympathetic nerve electrical activity response to bacterial endotoxin. Am. J. Physiol.

270, R264–R270.

MacNeil, B.J., Jansen, A.H., Janz, L.J., Greenberg, A.H., Nance, D.M., 1997. Peripheral

endotoxin increases splenic sympathetic nerve activity via central prostaglandin synthesis.

Am. J. Physiol. 273, R609–R614.

Madden, K.S., Felten, D.L., 2001. Beta-adrenoceptor blockade alters thymocyte differentiation

Page 56: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

55

in aged mice. Cell. Mol. Biol. (Noisy-le-grand). 47, 189–196.

Maestroni, G.J., 1995. Adrenergic regulation of haematopoiesis. Pharmacol. Res. 32, 249–253.

Maestroni, G.J., 2000a. Neurohormones and catecholamines as functional components of the

bone marrow microenvironment. Ann. N. Y. Acad. Sci. 917, 29–37.

Maestroni, G.J., 2000b. Dendritic cell migration controlled by alpha 1b-adrenergic receptors. J.

Immunol. 165, 6743–6747.

Maestroni, G.J., 2002. Short exposure of maturing, bone marrow-derived dendritic cells to

norepinephrine: impact on kinetics of cytokine production and Th development. J.

Neuroimmunol. 129, 106–114.

Maestroni, G.J. 2005. Adrenergic modulation of dendritic cells function: relevance for the

immune homeostasis. Curr. Neurovasc. Res. 2, 169–173.

Maestroni, G.J., 2006. Sympathetic nervous system influence on the innate immune response.

Ann. N.Y. Acad. Sci. 1069, 195–207.

Maestroni, G.J., Conti A., 1994a. Modulation of hematopoiesis via alpha 1-adrenergic receptors

on bone marrow cells. Exp. Hematol. 22, 313–320.

Maestroni, G.J., Conti, A., 1994b. Noradrenergic modulation of lymphohematopoiesis. Int. J.

Immunopharmacol. 16, 117–122.

Maestroni, G.J., Cosentino, M., Marino, F., Togni, M., Conti, A., Lecchini S, Frigo G., 1998.

Neural and endogenous catecholamines in the bone marrow. Circadian association of

norepinephrine with hematopoiesis? Exp. Hematol. 26, 1172–1177.

Maimone, D., Cioni, C., Rosa, S., Macchia, G., Aloisi, F., Annunziata, P., 1993. Norepinephrine

and vasoactive intestinal peptide induce IL-6 secretion by astrocytes: Synergism with IL-1

beta and TNF alpha. J. Neuroimmunol. 47, 73–81.

Maier, S.F., Goehler, L.E., Fleshner, M., Watkins, L.R. 1998. The role of the vagus nerve in

cytokine-to-brain communication. Ann. N.Y. Acad. Sci., 840, 289–300.

Manni, M., Granstein, R.D., Maestroni, G. 2011. β2-Adrenergic agonists bias TLR-2 and NOD2

activated dendritic cells towards inducing an IL-17 immune response. Cytokine 55, 380–386.

Manni, M., Maestroni, G.J., 2008. Sympathetic nervous modulation of the skin innate and

adaptive immune response to peptidoglycan but not lipopolysaccharide: involvement of

beta-adrenoceptors and relevance in inflammatory diseases. Brain Behav. Immun. 22, 80–

88.

Marchetti, B., Morale, M.C., Paradis, P., Bouvier, M., 1994. Characterization, expression, and

hormonal control of a thymic beta 2-adrenergic receptor. Am. J. Physiol. 267, E718–31.

Martelli, McKinley & McAllen 2014, Autonomic Neuroscience.

Martin, D.S., Haywood, J.R., 1992. Sympathetic nervous system activation by glutamate

injections into the paraventricular nucleus. Brain Res., 577, 261–267.

Page 57: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

56

Masuyama, K., Ishikawa, T., 1985. Direct interaction of guinea pig eosinophils and adrenergic

agents. Int. Arch. Allergy Appl. Immunol. 78, 243–428.

McDonald, K.G., McDonough, J.S., Newberry, R.D., 2005. Adaptive immune responses are

dispensable for isolated lymphoid follicle formation: antigen-naive, lymphotoxin-sufficient B

lymphocytes drive the formation of mature isolated lymphoid follicles. J. Immunol. 174,

5720–5728.

McGovern & Mazzone, Sharkey & Savidge. Autonomic Neuroscience.

McGraw, D.W., Liggett, S.B., 1997. Heterogeneity of β-adrenergic receptor kinase expression in

the lung accounts for cell-specific desensitisation of the β2-adrenergic receptor. J. Biol.

Chem. 272, 7338–7344.

Meckler, R.L., Weaver, L.C., 1985. Splenic, renal, and cardiac nerves have unequal

dependence upon tonic supraspinal inputs. Brain Res. 338, 123–135.

Meckler, R.L., Weaver, L.C., 1988a. Characteristics of ongoing and reflex discharge of single

splenic and renal sympathetic postganglionic fibres in cats. J. Physiol. 396, 139–153.

Meckler, R.L., Weaver, L.C., 1988b. Persistent firing of splenic and renal nerves after acute

decentralization but failure to produce ganglionic reflexes. Neurosci. Lett. 88, 167–172.

Melmon, K.L., Bourne, H.R., Weinstein, Y., Shearer, G.M., Kram, J., Bauminger, S., 1974.

Hemolytic plaque formation by leukocytes in vitro. Control by vasoactive hormones. J. Clin.

Invest. 53, 13–21.

Meng, A.-H., Ling, Y.-L.., Zang, X.-P., Zhao, X.Y., Zhang, J.-L., 2002. CCK8 inhibits expression of

TNF-α in the spleen of endotoxic shock rats and signal transduction mechanism of p38 MAPK.

World J. Gastroenterol. 8, 139–143.

Méndez-Ferrer, S., Lucas, D., Battista, M., Frenette, P.S., 2008. Haematopoietic stem cell

release is regulated by circadian oscillations. Nature 452, 442–447.

Miles, B.A., Lafuse, W.P., Zwilling, B.S., 1996. Binding of alpha-adrenergic receptors stimulates

the anti-mycobacterial activity of murine peritoneal macrophages. J. Neuroimmunol. 71, 19–

24.

Mills, P.J., Goebel, M., Rehman, J., Irwin, M.R., Maisel, A.S., 2000. Leukocyte adhesion

molecule expression and T-cell naïve/memory status following isoproterenol infusion. J.

Neuroimmunol. 102, 137–144.

Morita Y, Miyamoto T., 1987. Fenoterol, a selective beta 2-adrenergic agonist, and inhibition of

IgE-mediated basophil histamine release. Allergy 42, 524–528.

Moroni, F., Fantozzi, R., Masini, E., Mannaioni, P.F., 1977. The modulation of histamine release

by alpha-adrenoceptors: evidences in murine neoplastic mast cells. Agents Actions 7, 57–

61.

Page 58: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

57

Morrison, S.F. 2001. Differential control of sympathetic outflow. Am J Physiol Regulatory

Integrative Comp Physiol 281, R683–R698.

Moyron-Quiroz, J.E., Rangel-Moreno, J., Kusser, K., Hartson, L., Sprague, F., Goodrich, S.,

Woodland, D.L., Lund, F.E., Randall, T.D., 2004. Role of inducible bronchus associated

lymphoid tissue (iBALT) in respiratory immunity. Nat. Med. 10, 927–934.

Muthu, K., Deng, J., Gamelli, R., Shankar, R., Jones, S.B., 2005. Adrenergic modulation of

cytokine release in bone marrow progenitor-derived macrophage following polymicrobial

sepsis. J. Neuroimmunol. 158, 50–57.

Muthu, K., Lyer, S., He, L., Szilagyi, A., Gamelli, R., Shankar, R., Jones, S., 2007. Murine

hematopoietic stem cells and progenitors express adrenergic receptors. J. Neuroimmunol.

186, 27–36.

Nakamura, A., Johns, E.J., Imaizumi, A., Abe, T., Kohsaka, T., 1998. Regulation of tumour

necrosis factor and interleukin-6 gene transcription by beta2-adrenoceptor in the rat

astrocytes. J. Neuroimmunol. 88, 144–153.

Nance, D.M., Burns, J., 1989. Innervation of the spleen in the rat: evidence for absence of

afferent innervation. Brain Behav. Immun. 3, 281–290.

Nance, D.M., Hopkins, D.A., Bieger, D., 1987. Re-investigation of the innervation of the thymus

gland in mice and rats. Brain Behav. Immun. 1, 134–147.

Nance, D.M., Luczy-Bachman, G., Min, P., Lee, Y-B., Cho, Z-H. 2005. The immunomodulatory

effects of counter-irritation are mediated via the sympathetic nervous system. Brain Behav.

Immun. 19, 4, S1, e52–e53.

Nance, D.M., Sanders, V.M., 2007. Autonomic innervation and regulation of the immune system

(1987-2007). Brain Behav. Immun. 21, 736–745.

Németh, Z.H., Szabó, C., Haskó, G., Salzman, A.L., Vizi, E.S., 1997. Effect of the

phosphodiesterase III inhibitor amrinone on cytokine and nitric oxide production in

immunostimulated J774.1 macrophages. Eur. J. Pharmacol. 339, 215–221.

Neumann H., 2001. Control of glial immune function by neurons. Glia 36, 191–199.

Neyt, K., Perros, F., GeurtsvanKessel, C.H., Hammad, H., Lambrecht, BN., 2012. Tertiary

lymphoid organs in infection and autoimmunity. Trends Immunol. 33, 297–305.

Nielson, C.P., 1987. Beta-adrenergic modulation of the polymorphonuclear leukocyte respiratory

burst is dependent upon the mechanism of cell activation. J Immunol 139:2392–2397.

Nohr, D., Weihe, E. 1991. The neuroimmune link in the bronchus-associated lymphoid tissue

(BALT) of cat and rat: peptides and neural markers. Brain Behav. Immun., 5, 84–101.

Norris, J.G., Benveniste, E.N. 1993. Interleukin-6 production by astrocytes: Induction by the

neurotransmitter norepinephrine. J. Neuroimmunol. 45, 137–145.

Novotny, G.E., Sommerfeld, H., Zirbes, T., 1990. Thymic innervation in the rat: a light and

Page 59: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

58

electron microscopical study. J. Comp. Neurol. 302, 552–561.

Novotny, G.E., Hsu, Y., 1993. The effect of immunostimulation on thymic innervation in the rat.

J. Hirnforsch. 34, 155–163.

Novotny, G.E., Heuer, T., Schöttelndreier, A., Fleisgarten, C., 1994. Plasticity of innervation of

the medulla of axillary lymph nodes in the rat after antigenic stimulation. Anat. Rec. 238,

213–224.

Nunn, N., Womack, M., Dart, C., Barrett-Jolley, R., 2011. Function and pharmacology of

spinally-projecting sympathetic pre-autonomic neurones in the paraventricular nucleus of the

hypothalamus. Curr. Neuropharmacol. 9, 262–277.

Okamoto, S., Ishikawa, I., Kimura, K., Saito, M., 1998. Potent suppressive effects of urocortin

on splenic lymphocyte activity in rats. Neuroreport 9, 4035–4039.

Osborn, J.W., Livingstone, R.H. & Schramm, L.P. 1987. Elevated renal nerve activity after

spinal transection: effects on renal function. Am. J. Physiol. Regulatory Integrative Comp.

Physiol. 253, R619–R625.

Ottaway, C.A., Husband, A.J., 1992. Central nervous system influences on lymphocyte

migration. Brain Behav. Immun. 6, 97–116.

Ottaway CA, Husband AJ. 1994. The influence of neuroendocrine pathways on lymphocyte

migration. Immunol. Today 15, 511–517.

Pabst. R., Tschernig, T., 2010. Bronchus-associated lymphoid tissue: an entry site for antigens

for successful mucosal vaccinations? Am. J. Respir. Cell. Mol. Biol.43, 137–141.

Palkovits, M., Baffi, J.S., Pacak, K., 1997. Stress-induced Fos-like immunoreactivity in the pons

and the medulla oblongata of rats. Stress 1, 155–168.

Panina-Bordignon, P., Mazzeo, D., Lucia, P.D., Ambrosio, D. D., Lang, R., Fabbri, L., Self, C.,

Sinigaglia, F., 1997. β2-agonists prevent Th1 development by selective inhibition of

interleukin 12. J. Clin. Invest. 100, 1513–1519.

Pardini, B.J., Lund, D.D., Schmid, P.G., 1989. Organization of the sympathetic postganglionic

innervation of the rat heart. J. Auton. Nerv. Syst. 28:193–201.

Parikh, V., Singh, M., 1999. Possible role of adrenergic component and cardiac mast cell

degranulation in preconditioning-induced cardioprotection. Pharmacol. Res. 40, 129–137.

Patel, K., Taub, D.D., 2009. Role of neuropeptides, hormones, and growth factors in regulating

thymopoiesis in middle to old age. F1000 Biol. Rep. 1:42.

Peduto, L., Dulauroy, S., Lochner, M., Späth, G.F., Morales, M.A., Cumano, A., Eberl, G., 2009.

Inflammation recapitulates the ontogeny of lymphoid stromal cells. J. Immunol. 182, 5789–

5799.

Pešić, V., Kosec, D., Radojević, K., Pilipović, I., Perisić, M., Vidić-Danković, B., Leposavić, G.,

2009. Expression of alpha1-adrenoceptors on thymic cells and their role in fine tuning of

Page 60: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

59

thymopoiesis. J. Neuroimmunol. 214, 55–66.

Pešić, V., Plećas-Solarović, B., Radojević, K., Kosec, D., Pilipović, I., Perisić, M., Leposavić, G.,

2007. Long-term beta-adrenergic receptor blockade increases levels of the most mature

thymocyte subsets in aged rats. Int. Immunopharmacol. 7, 674–686.

Petit, I., Szyper-Kravitz, M., Nagler, A., Lahav, M., Peled, A., Habler, L., Ponomaryov, T.,

Taichman, R.S., Arenzana-Seisdedos, F., Fujii, N., Sandbank, J., Zipori, D., Lapidot, T.,

2002. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-

regulating CXCR4. Nat. Immunol. 3, 687–694. Erratum in: Nat. Immunol. 2002 3, 787.

Pezzone, M.A., Dohanics, J., Rabin, B.S., 1994. Effects of footshock stress upon spleen and

peripheral blood lymphocyte mitogenic responses in rats with lesions of the paraventricular

nuclei. J. Neuroimmunol. 53, 39–46.

Plećas-Solarović, B., Lalić, L., Leposavić, G., 2004. Age-dependent morphometrical changes in

the thymus of male propranolol-treated rats. Ann. Anat. 186, 141–147.

Plećas-Solarović, B., Hristić-Zivković, I., Radojević, K., Kosec, D., Leposavić, G., 2005. Chronic

alpha1-adrenoreceptor blockade produces age-dependent changes in rat thymus structure

and thymocyte differentiation. Histol. Histopathol. 20, 833–841.

Podojil, J.R., Kin, N.W., Sanders, V.M. 2004. CD86 and beta2-adrenergic receptor signaling

pathways, respectively, increase Oct-2 and OCA-B Expression and binding to the 3'-IgH

enhancer in B cells. J. Biol. Chem. 279, 23394–23404.

Podojil, J.R., Sanders, V.M., 2003. Selective regulation of mature IgG1 transcription by CD86

and beta 2-adrenergic receptor stimulation. J Immunol. 170, 5143–5151.

Pollok, K.E., O’Brien, V., Marshall, L., Olson, J.W., Noelle, R.J., Snow, E.C. 1991. The

development of competence in resting B cells. The induction of cyclic AMP and ornithine

decarboxylase activity after direct contact between B and T helper cells. J. Immunol. 146,

1633–1641.

Pongratz, G., McAlees, J.W., Conrad, D.H., Erbe, R.S., Haas, K.M., Sanders, V.M., 2006. The

level of IgE produced by a B cell is regulated by norepinephrine in a p38 MAPK- and CD23-

dependent manner. J. Immunol. 177, 2926–2938.

Proud, D., Reynolds, C.J., Lichtenstein, L.M., Kagey-Sobotka, A., Togias, A., 1998. Intranasal

salmeterol inhibits allergen-induced vascular permeability but not mast cell activation or

cellular infiltration. Clin. Exp. Allergy 28, 868–875.

Qu, L., Sherebrin, R., Weaver, L.C., 1988. Blockade of spinal pathways decreases pre- and

postganglionic discharge differentially. Am. J. Physiol. 255, R946–R951.

Ramer-Quinn, D.S., Baker, R.A., Sanders, V.M., 1997. Activated Th1 and Th2 cells differentially

express the beta-2-adrenergic receptor: a mechanism for selective modulation of Th1 cell

cytokine production. J. Immunol. 159, 4857–4867.

Page 61: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

60

Ramer-Quinn, D.S., Swanson, M.A., Lee, W.T., Sanders, V.M., 2000. Cytokine production by

naive and primary effector CD4(+) T cells exposed to norepinephrine. Brain Behav. Immun.

14, 239–255.

Rangel-Moreno, J., Carragher, D.M., de la Luz Garcia-Hernandez, M., Hwang, J.Y., Kusser, K.,

Hartson, L., Kolls, J.K., Khader, S.A., Randall, T.D., 2011. The development of inducible

bronchus-associated lymphoid tissue depends on IL-17. Nat. Immunol. 12, 639–646.

Rangel-Moreno, J., Moyron-Quiroz, J.E., Hartson, L., Kusser, K., Randall, T.D., 2007.

Pulmonary expression of CXC chemokine ligand 13, CC chemokine ligand 19, and CC

chemokine ligand 21 is essential for local immunity to influenza. Proc. Natl. Acad. Sci.

U.S.A. 104, 10577–10582.

Rassnick, S., Sved, A.F., Rabin, B.S., 1994. Locus coeruleus stimulation by corticotropin-

releasing hormone suppresses in vitro cellular immune responses. J. Neurosci. 14, 6033–

6040.

Rauski, A., Kosec, D., Vidić-Danković, B., Radojević, K., Plećas-Solarović, B., Leposavić, G.,

2003. Thymopoiesis following chronic blockade of beta-adrenoceptors. Immunopharmacol.

Immunotoxicol. 25, 513–528.

Rehman, J., Mills, P.J., Carter, S.M., Chou, J., Thomas, J., Maisel, A.S., 1997. Dynamic

exercise leads to an increase in circulating ICAM-1: further evidence for adrenergic

modulation of cell adhesion. Brain Behav. Immun. 11, 343–351.

Ricci, A., Bronzetti, E., Conterno, A., Greco, S., Mulatero, P., Schena, M., Schiavone, D.,

Tayebati ,S.K., Veglio, F., Amenta, F., 1999. alpha1-adrenergic receptor subtypes in human

peripheral blood lymphocytes. Hypertension 33, 708–712.

Riether, C., Doenlen, R., Pacheco-López, G., Niemi, M.B., Engler, A., Engler, H., Schedlowski,

M., 2008. Behavioural conditioning of immune functions: how the central nervous system

controls peripheral immune responses by evoking associative learning processes. Rev.

Neurosci. 19, 1–17.

Rosas-Ballina, M., Tracey, K.J., 2009. Cholinergic control of inflammation. J. Intern. Med. 265,

663–679.

Rosas-Ballina, M., Ochani, M., Parrish, W.R., Ochani, K., Harris, Y.T., Huston, J.M., Chavan, S,

Tracey, K.J., 2008. Splenic nerve is required for cholinergic antiinflammatory pathway

control of TNF in endotoxemia. Proc. Natl. Acad. Sci. U.S.A. 105, 11008–11013.

Rothwell, N.J., 1991. Functions and mechanisms of interleukin 1 in the brain. Trends

Pharmacol. Sci. 12, 430–436.

Rouppe van der Voort, C., Kavelaars, A., van de Pol, M., Heijnen, C.J., 1999. Neuroendocrine

mediators upregulate α1b- and α1d-adrenergic receptor subtypes in human monocytes. J.

Neuroimmunol. 95, 165–173.

Page 62: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

61

Rouppe van der Voort, C., Kavelaars, A., van de Pol, M., Heijnen, C.J., 2000a. Noradrenaline

induces phosphorylation of ERK-2 in human peripheral blood mononuclear cells after

induction of α1-adrenergic receptors. J. Neuroimmunol. 108, 82–91.

Rouppe van der Voort, C., Heijnen, C.J., Wulffraat, N., Kuis, W., Kavelaars, A., 2000b. Stress

induces increases in IL-6 production by leucocytes of patients with the chronic inflammatory

disease juvenile rheumatoid arthritis: a putative role for alpha(1)-adrenergic receptors. J.

Neuroimmunol. 110, 223–229.

Roszman. T.L., Cross, R.J., Brooks, W.H., Markesbery, W.R., 1982. Hypothalamic-immune

interactions. II. The effect of hypothalamic lesions on the ability of adherent spleen cells to

limit lymphocyte blastogenesis. Immunology 45:737–742.

Russo, D., Fantaguzzi, C.M., Di Guardo, G., Clavenzani, P., Costerbosa, G.L., Ligios, C.,

Chiocchetti, R., 2009. Characterization of sheep (Ovis aries) palatine tonsil innervation.

Neuroscience, 161, 813–826.

Sanders, V.M., 2012. The beta2-adrenergic receptor on T and B lymphocytes: do we

understand it yet? Brain Behav. Immun. 26, 195–200.

Sanders, V.M., Baker, R.A., Ramer-Quinn, D.S., Kasprowicz, D.J., Fuchs, B.A., Street, N.E.,

1997. Differential expression of the beta2-adrenergic receptor by Th1 and Th2 clones:

implications for cytokine production and B cell help. J. Immunol. 158, 4200–4210.

Sanders, V.M., Kasprowicz, D.J., Kohm, A.P., Swanson, M.A., 2001. Neurotransmitter receptors

on lymphocytes and other lymphoid cells. In: Ader, R., Felten, D., Cohen, N. (Eds.).

Psychoneuroimmunology Academic Press, San Diego, CA, pp. 161–196.

Saper, C., 1995. Central autonomic system. In: Paxinos, G. (Ed.). The rat nervous system. San

Diego: Academic Press, pp. 107–135.

Sato, Y., 2004. Modulation of PMN-endothelial cells interactions by cyclic nucleotides. Curr.

Pharm. Des. 10, 163–170.

Sawchenko, P.E., Swanson, L.W., 1981. Central noradrenergic pathways for the integration of

hypothalamic neuroendocrine and autonomic responses. Science 214, 685–687.

Sawchenko, P.E., Swanson, L.W., 1982. The organization of noradrenergic pathways from the

brainstem to the paraventricular and supraoptic nuclei in the rat. Brain Res. 257, 275–325.

Saxena, M., Williams, S., Tasken, K., Mustelin, T., 1999. Crosstalk between cAMP-dependent

kinase and MAP kinase through a protein tyrosine phosphatase. Nat. Cell Biol. 1, 305–311.

Schäfer, M.K., Eiden, L.E., Weihe, E., 1998. Cholinergic neurons and terminal fields revealed by

immunohistochemistry for the vesicular acetylcholine transporter. II. The peripheral nervous

system. Neuroscience, 84, 361–376.

Schedlowski, M., Hosch, W., Oberbeck, R., Benschop, R.J., Jacobs, R., Raab, H.R., Schmidt,

R.E. 1996. Catecholamines modulate human NK cell circulation and function via spleen-

Page 63: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

62

independent beta 2-adrenergic mechanisms. J. Immunol. 156, 93–99.

Schramm, L.P., Strack, A.M., Platt, K.B., Loewy, A.D., 1993. Peripheral and central pathways

regulating the kidney: a study using pseudorabies virus. Brain Res. 616, 251–262.

Seiffert, K., Hosoi, J., Torii, H., Ozawa, H., Ding, W., Campton, K., Wagner, J.A., Granstein,

R.D., 2002. Catecholamines inhibit the antigen-presenting capability of epidermal

Langerhans cells. J. Immunol. 168, 6128–6135.

Severn, A., Rapson, N.T., Hunter, C.A., Liew, F.Y., 1992. Regulation of tumor necrosis factor

production by adrenaline and beta-adrenergic agonists. J. Immunol. 148, 3441–3445.

Shakhar, G., Ben-Eliyahu, S. 1998. In vivo beta-adrenergic stimulation suppresses natural killer

activity and compromises resistance to tumor metastasis in rats. J. Immunol. 160, 3251–

3258.

Sharkey and Savidge Autonomic Neuroscience

Siegmund, B., Eigler, A., Hartmann, G., Hacker, U., Endres, S., 1998. Adrenaline enhances

LPS-induced IL-10 synthesis: evidence for protein kinase A-mediated pathway. Int. J.

Immunopharmacol. 20, 57–69.

Singh, U. 1985a. Lymphopoiesis in the nude fetal thymus following sympathectomy. Cell.

Immunol. 93, 222–228.

Singh, U., 1985b. Effect of sympathectomy on the maturation of fetal thymocytes grown within

the anterior eye chambers in mice. Adv. Exp. Biol. Med. 186, 349–356,

Sirot'áková, M., Schmidtová, K., Kocisová, M., Kuchta, M., 2002. Adrenergic and

acetylcholinesterase-positive innervation of palatine tonsils in mammals. Acta Histochem.

104, 349–352.

Spengler, R.N., Allen, R.M., Remick, D.G., Strieter, R.M., Kunkel, S.L., 1990. Stimulation of

alpha-adrenergic receptor augments the production of macrophage-derived tumor necrosis

factor. J. Immunol. 145, 1430–1434.

Spengler, R.N., Chensue, S.W., Giacherio, D.A., Blenk, N., Kunkel, S.L., 1994. Endogenous

norepinephrine regulates tumor necrosis factor-alpha production from macrophages in vitro.

J. Immunol. 152, 3024–3031.

Stein, S.H., Phipps, R.P., 1991. Elevated levels of intracellular cAMP sensitize resting B

lymphocytes to immune complex-induced unresponsiveness. Eur. J. Immunol. 21, 313–318.

Stein, R.D., Weaver, L.C., 1988. Multi- and single-fibre mesenteric and renal sympathetic

responses to chemical stimulation of intestinal receptors in cats. J. Physiol. 396, 155-172.

Stevens-Felten, S.Y., and Bellinger, D.L. 1997. Noradrenergic and peptidergic innervation of

lymphoid organs. Chem. Immunol. 69, 99–131.

Page 64: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

63

Strack, A.M., Sawyer, W.B., Hughes, J.H., Platt, K.B., Loewy, A.D., 1989a. A general pattern of

CNS innervation of the sympathetic outflow demonstrated by transneuronal pseudorabies

viral infections. Brain Res 491:156–162.

Strack, A.M., Sawyer, W.B., Platt, K.B., Loewy, A.D., 1989b. CNS cell groups regulating the

sympathetic outflow to adrenal gland as revealed by transneuronal cell body labeling with

pseudorabies virus. Brain Res. 491, 274–296.

Straub, R.H., Grum, F., Strauch, U., Capellino, S., Bataille, F., Bleich, A., Falk, W., Schölmerich,

J., Obermeier, F., 2008. Anti-inflammatory role of sympathetic nerves in chronic intestinal

inflammation. Gut 57, 911–921.

Straub, R.H., Mayer, M., Kreutz, M., Leeb, S., Schölmerich, J., Falk, W., 2000.

Neurotransmitters of the sympathetic nerve terminal are powerful chemoattractants for

monocytes. J. Leukoc. Biol. 67, 553–558.

Su, C., Bevan, J.A., 1970. The release of H3-norepinephrine in arterial strips studied by the

technique of superfusion and transmural stimulation. J. Pharmacol. Exp. Ther. 172, 62–68.

Suberville, S., Bellocq, A., Fouqueray, B., Philippe, C., Lantz, O., Perez, J., Baud, L., 1996.

Regulation of interleukin-10 production by beta-adrenergic agonists. Eur. J. Immunol. 26,

2601–2605.

Sun, M.K. 1995. Central neural organization and control of sympathetic nervous system in

mammals. Prog. Neurobiol. 47, 157–233.

Sundar, S.K., Becker, K.J., Cierpial, M.A., Carpenter, M.D., Rankin, L.A., Fleener, S.L., Ritchie,

J.C., Simson, P.E., Weiss, J.M., 1989. Intracerebroventricular infusion of interleukin 1 rapidly

decreases peripheral cellular immune responses. Proc. Natl. Acad. Sci. U.S.A. 86, 6398–

6402.

Sved, A.F., Cano, G., Passerin, A.M., Rabin, B.S., 2002. The locus coeruleus, Barrington's

nucleus, and neural circuits of stress. Physiol. Behav. 77, 737–742.

Swanson, L.W., Sawchenko, P.E., 1983. Hypothalamic integration: Organization of the

paraventricular and supraoptic nuclei. Annu. Rev. Neurosci. 6, 269–324.

Swanson, M.A., Lee, W.T., Sanders, V.M., 2001. IFN-gamma production by Th1 cells generated

from naive CD4(+) T cells exposed to norepinephrine. J. Immunol. 166, 232–240. Erratum

in: J. Immunol. 166, 6992.

Szelényi, J., Kiss, J.P., Vizi, E.S., 2000. Differential involvement of sympathetic nervous system

and immune system in the modulation of TNF-alpha production by alpha2- and beta-

adrenoceptors in mice. J. Neuroimmunol. 103, 34–40.

Takahashi, H.K., Iwagaki, H., Tamura, R., Katsuno, G., Xue, D., Sugita, S., Mori, S., Yoshino,

T., Tanaka, N., Nishibori, M., 2005. alpha1-Adrenergic receptor antagonists induce

production of IL-18 and expression of ICAM-1 and CD40 in human monocytes. J.

Page 65: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

64

Immunother. 28, 40–43.

Takahashi, H.K., Morichika, T., Iwagaki, H., Yoshino, T., Tamura, R., Saito, S., Mori, S., Akagi,

T., Tanaka, N., Nishibori, M., 2003. Effect of beta 2-adrenergic receptor stimulation on

interleukin-18-induced intercellular adhesion molecule-1 expression and cytokine

production. J. Pharmacol. Exp. Ther. 304, 634–642.

Take S, Mori T, Katafuchi T, Hori T. 1993. Central interferon-alpha inhibits natural killer

cytotoxicity through sympathetic innervation. Am. J. Physiol. 265, R453–R459.

Tarr, A.J., Powell, N.D., Reader, B.F., Bhave, N.S., Roloson, A.L., Carson, W.E. 3rd, Sheridan,

J.F., 2012. β-Adrenergic receptor mediated increases in activation and function of natural

killer cells following repeated social disruption. Brain Behav. Immun. 26, 1226–1238.

Taylor, R.F., Schramm, L.P. 1987. Differential effects of spinal transection on sympathetic nerve

activities in rats. Am. J. Physiol. 253, R611–R618.

Taylor, R.B., Weaver, L.C., 1992. Spinal stimulation to locate preganglionic neurons controlling

the kidney, spleen, or intestine. Am. J. Physiol. 263, H1026–H1033.

Terao, A., Oikawa, M., Saito M., 1994. Tissue-specific increase in norepinephrine turnover by

central interleukin-1, but not by interleukin-6, in rats. Am. J. Physiol. 266, R400–R404.

Tesio, M., Golan, K., Corso, S., Giordano S., Schajnovitz, A., Vagima, Y., Shivtiel, S.,

Kalinkovich, A., Caione, L., Gammaitoni, L., Laurenti, E., Buss, E.C., Shezen, E., Itkin, T.,

Kollet, O., Petit, I., Trumpp, A., Christensen J., Aglietta, M., Piacibello, W., Lapidot, T., 2011.

Enhanced c-Met activity promotes G-CSF-induced mobilization of hematopoietic progenitor

cells via ROS signaling. Blood. 117, 419–428.

ThyagaRajan, S., Madden, K.S., Teruya, B., Stevens, S.Y., Felten, D.L., Bellinger, D.L., 2011.

Age-associated alterations in sympathetic noradrenergic innervation of primary and

secondary lymphoid organs in female Fischer 344 rats. J. Neuroimmunol. 233, 54–64.

Togni, M., Maestroni, G., 1996. Hematopoietic rescue in mice via alpha 1-adrenoceptors on

bone marrow B cell precursors. Int. J. Oncol. 9, 313–318.

Tool, A.T., Mul, F.P., Knol, E.F., Verhoeven, A.J., Roos, D., 1996. The effect of salmeterol and

nimesulide on chemotaxis and synthesis of PAF and LTC4 by human eosinophils. Eur.

Respir. J. Suppl. 22, 141s–145s.

Tracey, K.J., 2007. Physiology and immunology of the cholinergic antiinflammatory pathway. J.

Clin. Invest. 117, 289–296.

Tracey, K.J. 2009. Reflex control of immunity. Nat. Rev. Immunol. 9, 418–428.

Tsuji, T., Kato, T., Kimata, M., Miura, T., Serizawa, I., Inagaki, N., Nagai, H., 2004. Differential

effects of β2-adrenoceptor desensitization on the IgE-dependent release of chemical

mediators from cultured human mast cells. Biol. Pharm. Bull. 27, 1549–1554.

Ueyama, T., Kozuki, K., Houtani, T., Ikeda, M., Kitajiri, M., Yamashita, T., Kumazawa, T.,

Page 66: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

65

Nagatsu, I., Sugimoto, T., 1990. Immunolocalization of tyrosine hydroxylase and vasoactive

intestinal polypeptide in nerve fibers innervating human palatine tonsil and paratonsillar

glands. Neurosci. Lett. 116, 70–74.

Uvnäs-Moberg, K., Lundeberg, T., Bruzelius, G., Alster, P., 1992. Vagally mediated release of

gastrin and cholecystokinin following sensory stimulation. Acta Physiol. Scand. 146, 349–

356.

van de Pavert, S.A., Olivier, B.J., Goverse, G., Vondenhoff, M.F., Greuter, M., Beke, P., Kusser,

K., Höpken, U.E., Lipp, M., Niederreither, K., Blomhoff, R., Sitnik, K., Agace, W.W., Randall,

T.D., de Jonge, W.J., Mebius, R.E., 2009. Chemokine CXCL13 is essential for lymph node

initiation and is induced by retinoic acid and neuronal stimulation. Nat. Immunol. 10, 1193–

1199

van der Poll, T., Jansen, J., Endert, E., Sauerwein, H.P., van Deventer, S.J., 1994.

Noradrenaline inhibits lipopolysaccharide-induced tumor necrosis factor and interleukin 6

production in human whole blood. Infect. Immun. 62, 2046–2050.

van der Poll, T., Coyle, S.M., Barbosa, K., Braxton, C.C., Lowry, S.F., 1996. Epinephrine inhibits

tumor necrosis factor-alpha and potentiates interleukin 10 production during human

endotoxemia. J. Clin. Invest. 97, 713–719.

van der Poll, T., Lowry, S.F., 1997. Epinephrine inhibits endotoxin-induced IL-1beta production:

Roles of tumor necrosis factor-alpha and IL-10. Am. J. Physiol. 273, R1885–R1890.

Vaughan, J., Donaldson, C., Bittencourt, J., Perrin, M.H., Lewis, K., Sutton, S., Chan, R.,

Turnbull, A.V., Lovejoy, D., Rivier, C., Rivier, J., Sawchenko, P.E., Vale, W., 1995. Urocortin,

a mammalian neuropeptide related to fish urotensin I and to corticotropin-releasing factor.

Nature 378, 287–292.

Veiga-Fernandes, H., Coles, M.C., Foster, K.E., Patel, A., Williams, A., Natarajan, D., Barlow,

A., Pachnis, V., Kioussis, D., 2007. Tyrosine kinase receptor RET is a key regulator of

Peyer’s patch organogenesis. Nature 446, 547–551.

Vendetti, S., Riccomi, A., Sacchi, A., Gatta, L., Piolo C., De Magistris, M.T. 2002. Cyclic

adenosine 5'-monophosphate and calcium induce CD152 (CTLA-4) up-regulation in resting

CD4(+) T lymphocytes. J. Immunol. 169, 6231–6235.

Verbalis, J.G., Stricker, E.M., Robinson, A.G., Hoffman, G.E., Verbalis, Joseph G., Stricker,

Edward M., Robinson, Alan G., Hoffman, Gloria E., 1991. Cholecystokinin activates c-fos

expression in hypothalamic oxytocin and corticotropin-releasing hormone neurons. J.

Neuroendocrinol. 3, 205–213.

Vida, G., Peña, G., Deitch, E.A., Ulloa, L., 2011a. α7-Cholinergic receptor mediates vagal

induction of splenic norepinephrine. J. Immunol. 186, 4340–4346.

Page 67: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

66

Vida, G., Peña, G., Kanashiro, A., del Rocio, M., Thompson-Bonilla, D.P., Deitch, E.A., Ulloa, L.,

2011b. β2-Adrenoreceptors of regulatory lymphocytes are essential for vagal

neuromodulation of the innate immune system. FASEB J. 25, 4476–4485.

Vizi, E.S., Orsó, E., Osipenko, O.N., Haskó, G., Elenkov, I.J., 1995. Neurochemical,

electrophysiological and immunocytochemical evidence for a noradrenergic link between the

sympathetic nervous system and thymocytes. Neuroscience 68, 1263–1276.

Vondenhoff, M.F., Greuter, M., Goverse, G., Elewaut, D., Dewint, P., Ware, C.F., Hoorweg, K.,

Kraal, G., Mebius, R.E., 2009. LTbetaR signaling induces cytokine expression and up-

regulates lymphangiogenic factors in lymph node anlagen. J. Immunol. 182, 5439–5445.

von Patay, B., Loppnow, H., Feindt, J., Kurz, B., Mentlein, R., 1998. Catecholamines and

lipopolysaccharide synergistically induce the release of interleukin-6 from thymic epithelial

cells. J. Neuroimmunol. 86, 182–189.

Vriend, C.Y., Zuo, L., Dyck, D.G., Nance, D.M., Greeberg A.H. 1993. Central administration of

interleukin-1 beta increase norepinephrine turnover in the spleen. Brain Res. Bull. 31, 39–

42.

Wajeman-Chao, S.A., Lancaster, S.A., Graf, L.H. Jr., Chambers, D.A., 1998. Mechanism of

catecholamine-mediated destabilization of messenger RNA encoding Thy-1 protein in T-

lineage cells. J. Immunol. 161, 4825–4833.

Wan, W., Vriend, C.Y., Wetmore, L., Gartner, J.G., Greenberg, A.H., Nance, D.M., 1993. The

effects of stress on splenic immune function are mediated by the splenic nerve. Brain Res.

Bull. 30, 101–105.

Wang, H., Yu, M., Ochani, M., Amella, C.A., Tanovic, M., Susarla, S., Li, J.H., Wang, H., Yang,

H., Ulloa, L., Al-Abed, Y., Czura, C.J., Tracey, K.J., 2003. Nicotinic acetylcholine receptor α7

subunit is an essential regulator of inflammation. Nature 421, 384–388.

Watkins, L.R., Maier, S.F., 2000. The pain of being sick: implications of immune-to-brain

communication for understanding pain. Annu. Rev. Psychol. 51, 29–57.

Watkins, L.R., Maier, S.F., Goehler, L.E., 1995. Cytokine-to-brain communication: a review and

analysis of alternative mechanisms. Life Sci. 57, 1011–1026.

Watts, T.H., Alaverdi, N., Wade, W.F., Linsley, P.S., 1993. Induction of costimulatory molecule

B7 in M12 B lymphomas by cAMP or MHC-restricted T cell interaction. J. Immunol. 150,

2192–2202.

Weber, R., Pert, A., 1990. Suppression of natural killer cell activity following electrical

stimulation of the rat mesencephalon. Soc. Neurosci. Abstr, 16, 972.

Weiss, M., Schneider, E.M., Tarnow, J., Mettler, S., Krone, M., Teschemacher, A., Lemoine, H.,

1996. Is inhibition of oxygen radical production of neutrophils by sympathomimetics

mediated via beta-2 adrenoceptors? J. Pharmacol. Exp. Ther. 278, 1105–1113.

Page 68: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

67

Wengner, A.M., Höpken, U.E., Petrow, P.K., Hartmann, S., Schurigt, U., Bräuer, R., Lipp, M.,

2007. CXCR5- and CCR7-dependent lymphoid neogenesis in a murine model of chronic

antigen-induced arthritis. Arthritis Rheum. 56, 3271–3283.

Weninger, S.C., Peters, L.L., Majzoub, J.A., 2000. Urocortin expression in the Edinger-Westphal

nucleus is up-regulated by stress and corticotropin-releasing hormone deficiency.

Endocrinology 141:256–263.

Wenner, M., Kawamura, N., Miyazawa, H., Ago, Y., Ishikawa, T., Yamamoto, H., 1996. Acute

electrical stimulation of lateral hypothalamus increases natural killer cell activity in rats. J.

Neuroimmunol. 67, 67–70.

Whalen, M.M., Bankhurst, A.D., 1990. Effects of beta-adrenergic receptor activation, cholera

toxin and forskolin on human natural killer cell function. Biochem. J. 272, 327–331.

Wright, S.D., Ramos, R.A., Tobias, P.S., Ulevitch, R.J., Mathison, J.C., 1990. CD14, a receptor

for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 249, 1431–

1433.

Xiao, J., Huang, H.W., Peng, Y.P., Bao, J.Y., Huang, Y., Qiu, Y.H., 2010. Modulation of natural

killer cell function by alpha-adrenoreceptor-coupled signalling. Neuro. Endocrinol. Lett. 31,

635–644.

Yardley, C.P., Stein, R.D., Weaver, L.C., 1989. Tonic influences from the rostral medulla affect

sympathetic nerves differentially. Am. J. Physiol. 256, R323–R331.

Yamaguchi, M., Hirai, K., Ohta, K., Ito, K., Morita, Y., 1995. A beta 2-agonist, procaterol, inhibits

basophil migration. J. Asthma. 32, 125–130.

Yamazaki, S., Ema, H., Karlsson, G., Yamaguchi, T., Miyoshi, H., Shioda, S., Taketo, M.M.,

Karlsson, S., Iwama, A., Nakauchi, H., 2011. Nonmyelinating Schwann cells maintain

hematopoietic stem cell hibernation in the bone marrow niche. Cell 147, 1146–1158.

Yanagawa, Y., Matsumoto, M., Togashi, H., 2010. Enhanced dendritic cell antigen uptake via

alpha2 adrenoceptor-mediated PI3K activation following brief exposure to noradrenaline. J.

Immunol. 185, 5762–5768.

Yanagawa, Y., Matsumoto, M., Togashi, H., 2011. Adrenoceptor-mediated enhancement of

interleukin-33 production by dendritic cells. Brain Behav. Immun. 25, 1427–1433.

Yoon, S.-Y., Kim, H.-W.., Roh, D.-H., Kwon, Y.-B., Han, H.-J., Beitz, A.J., Lee, J.-H., 2006.

Intrathecal clonidine suppresses zymosan-induced peripheral leukocyte migration in a

mouse air pouch model via activation of spinal muscarinic type 2 receptors and

sympathoadrenal medullary activity. Neuropharmacology 51, 829–837.

Yoshimatsu, H., Egawa, M., Bray, G.A., 1993. Sympathetic nerve activity after discrete

hypothalamic injections of l-glutamate. Brain Res. 601, 121–128.

Zurier, R.B., Weissmann, G., Hoffstein, S., Kammerman, S., Tai, H.H., 1974. Mechanisms of

Page 69: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

68

lysosomal enzyme release from human leukocytes. II. Effects of cAMP and cGMP,

autonomic agonists, and agents which affect microtubule function. J. Clin. Invest. 53, 297–

309. Figure Legends

Figure 1. Overview of SNS-Immune System Cross-Talk. The brain responds to sensory information from the periphery, including circulating cytokines (particularly, interleukin (IL)-1, IL-6, tumor necrosis factor (TNF)-α) and sensory neurons that project to the tissues (lower left corner). Central nervous system (CNS) responses to sensory information are transmitted through many CNS pathways, thus alter the firing of preganglionic neurons to regulate SNS tone to sympathetic target organs, including lymphoid organs. Preganglionic sympathetic neurons in the intermediolateral cell column of the thoracolumbar region of the spinal cord (green circles) send their axons to postganglionic sympathetic neurons (green circles) located in sympathetic ganglia and to chromaffin cells in the adrenal medulla. Postganglionic neurons send their axons to lymphoid organs and tissues where they signal cells of the immune system and smooth muscle cells of the vasculature by releasing norepinephrine. Immune cells express predominantly β2-ARs (orange); however, some immune cells do express α-AR subtypes (pink). Preganglionic sympathetic nerves supply adrenal medullary chromaffin cells. These cells release predominantly epinephrine (~80%) and to a lesser extent norepinephrine (~20%) into the circulation (i.e., hormonal release) to interact with ARs expressed on immune cells, subsequently activating signaling pathways that alters their cellular functions. Through these connections, the SNS regulates immune system homeostasis, and under conditions of stress or immune activation. Figure 2. SNS Regulation of Hematopoiesis. Schematic illustrating sites where hematopoiesis of specific type of blood cells is either positively or negatively influenced by catecholamines or adrenergic receptor (AR) agonists. Catecholamines activating via α- or β-ARs promote lymphopoiesis, and β-agonists promote erythropoiesis (upper and lower left side). The β2-AR-mediated drive in erythropoiesis requires the presence of erythropoietin. In contrast, α- or β-agonists suppress in vivo granulocyte/monocyte production (lower left side). Catecholamines also promote the proliferation of pluripotent hematopoietic stem cells (HSCs) via β-AR-mediated mechanisms (upper left side). Additionally, both β3- and β2-ARs expressed

on bone marrow stromal and bone cells, respectively, regulate circadian (~) proliferation of

HSCs and trafficking into the circulation, and subsequently back into the bone marrow (upper right corner). Sympathetic regulation of bone marrow functions may be exploited clinically to improve bone marrow transplantation, or prevent anemia that can result under conditions of

chronic stress and/or inflammation. 1

Byron, 1971; 2

Maestroni et al., 1992, Maestroni & Conti,

1994a,b; 3

Maestroni et al., 1998, Mendez-Ferrier et al., 2010; 4

Mladenovic & Adamson, 1984. Figure 3. Sympathetic Regulation of Circadian HSC Proliferation and HSC Egress from BM. Photic cues are conveyed from the eye to the suprachiasmatic nucleus (SCN) through the retinal-hypothalamic tract (RHT), the central pacemaker in the brain (upper right side). Clock gene expression (like Bmal) in neurons of the suprachiasmatic nucleus (SCN), orchestrates the β3-AR-mediated circadian proliferation and release of hematopoietic stem cells (HSCs)/hematopoietic progenitor stem cell (HPSCs) (purple cells in large box). These signals are transmitted to the bone marrow (BM) via the sympathetic nervous system (SNS, green),

Page 70: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

69

causing the circadian (~) release of norepinephrine into the BM microenvironment. During the

rest period (day for mice; night for humans indicated by open or closed eye on the right side of the diagram, respectively), the rise in sympathetic activity in the bone marrow is driven by increased G-CSF release in the BM (upper large box). NE binding to β2-ARs on endosteal osteoblasts increase chemokine (C-X-C motif) 12 (CXCL12) secretion (also known as stromal cell-derived growth factor (SDF-1) (lower left of large box). CXCL12 subsequently binds to CXC receptor type 4 (CXCR4; see small box inset) on quiescent HSCs/HPSCs that are “tethered’ to the BM by the adhesion molecules, very late antigen 4 (VLA4) and vascular cell cellular adhesion molecules (VCAM-1)). CXCL12-CXCR4 binding increases metalloprotease 9 (MMP9) by stromal cells, which degrade the adhesion molecules and releases HSC for mobilization into the bloodstream. Clinically, G-CSF is used to increase recovery from depleted white blood cell counts. NE also binds to β3-adrenergic receptor (AR) on mesenchymal stromal cells (MSCs) (left side of large box), which reduces CXCL12 expression (lower left of large box). Beta2-ARs upregulate CXCL12 expression, whereas β3-ARs down-regulate CXCL12 expression, which permits or inhibits HSC/HPSC egress, respectively. As a result of β2- AR stimulation, protein kinase A phosphorylates specificity protein 1 (Sp1), which enhances its DNA binding activity and subsequently transcription of target genes that down-regulate CXCL12 expression (not shown). Conversely, circadian stimulation of β3-AR-Gi/o protein signaling pathway reduces cAMP production and consequently Sp1 phosphorylation, which increases Cxcl12 and CXCL12 expression and prevents HSC and HPSC release from BM. Based on findings from Méndez-Ferrer et al. 2008. Figure 4. HF Staining in Primary and Secondary Lymphoid Organs from Rodents. Fluorescence histochemical (HF) staining demonstrates noradrenergic innervation of the thymus (A), spleen (B), and lymph node (C). A. Thymic noradrenergic nerves derive from the superior cervical and stellate ganglia, and form neurovascular plexuses from which nerves extend into the surrounding parenchyma, particularly in the cortex (CTX). Sympathetic nerves form neuroeffector junctions with thymocytes and cells in the stroma throughout the cortex, so they are “positioned” for regulating key mechanisms responsible for ingress of HSPC and their migration from inner to outer cortex, thymocyte proliferation and maturation, T-cell receptor gene rearrangement, and positive and negative selection. Additionally, noradrenergic nerves closely appose immune cells that associate with the vasculature and connective tissue (i.e., capsule and septae) of the thymus, including perivascular mast cells, macrophages, eosinophils, fibroblast and other accessory cells. In comparison to the cortex, sympathetic nerves sparsely supply the medulla (M), and mainly associate with the vasculature, where hematopoietic progenitor stem cells ingress and mature T-cells egress. The close relationship between medullary vasculature and sympathetic nerves suggest a role for noradrenergic nerves in the ingress of hematopoietic stem cells, and egress of mature thymocytes. In man, but not rodents, noradrenergic nerves closely associate with medullary epithelial cells that form Hassall’s corpuscles; the significance of this finding is not clear. YAC, yellow autofluorescent cells. B. Noradrenergic fibers enter the spleen around the splenic artery, and travel with the vasculature in plexuses (not shown). Noradrenergic nerves continue into the spleen along the trabeculae in trabecular plexuses. Fibers continue mainly along the central artery (ca) and its branches. Noradrenergic nerves extend from these plexuses into the surrounding white pulp among lymphocytes in the periarteriolar lymphatic sheath (pals), but avoid the nodular areas containing naïve B cells and the parenchyma of the red pulp. Noradrenergic nerves are closely associated with CD4+ and CD8+ T-cells. In the marginal (mz) and the parafollicular zones that surround the pals, noradrenergic nerves course among B-cells and macrophages. Close associations between noradrenergic nerves and follicular dendritic cells, or stromal cells (i.e., reticuloendothelial cells and fibroblasts) have not been described, but are likely to occur given

Page 71: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

70

the overlapping distribution of these nerves and these cells. C. Noradrenergic innervation of a popliteal lymph node (PLN) showing fluorescent noradrenergic nerves traveling along a blood vessel (BV), in the paracortical regions (PC). Some fibers (arrowhead) branch from the vascular plexuses into the surrounding parenchyma and end among lymphocytes and other cells in this region (arrowheads). In the medulla (M) of LNs, sympathetic nerves reside adjacent to the medullary sinuses among T-cells, reticuloendothelial cells, macrophages and plasma cells of the medullary cords, and in the cortex, noradrenergic nerves reside among T-cells and autofluorescent cells, presumed to be macrophages. Sympathetic nerves provide only minimal innervation of B-lymphocyte-rich follicles and germinal centers. Figure 5A. SNS and Thymocyte Development. Schematic illustration of the proposed α- and β-adrenoceptor-mediated modulation of thymocyte development (upper and lower panel, respectively). Also in the upper pale yellow panel, the gray arrows represent the regions in the thymus where the following developmental changes occur in immature thymocytes: 1. β selection and rearrangement in the outer cortex; 2. Positive selection mid-cortex to the corticomedullary junction (CMJ); and 3. Negative selection in the CMJ and medulla. In the lower panel the cortex, corticomedullary junction (CMJ) and medulla are indicated. The major developmental stages in thymocytes are shown from left to right. Catecholamines have very little effect on the early developmental stages, i.e., double negative (DN) T-cell receptor (TCR)― thymocytes (right side of figure) either by α- or β-adrenergic receptors (ARs) stimulation (upper or lower panels, respectively). Stimulation of α-ARs increases (+) the frequency of double-positive (DP, CD4+CD8+) T-cell receptor (TCR)low thymocytes without affecting the frequency of double-negative (DN, CD4‒CD8‒ ) TCR‒ thymocytes. Activation of α-ARs also increases the frequency of single-positive (SP) CD8+ and reduces (‒ ) the frequency of CD4+- and CD4+CD25+RT6.1+ Treg-thymocytes (left side of upper panel). In contrast, stimulation of β-ARs (lower panel) negatively (‒ ) regulates both positive and negative selection, reducing the frequency of DP TCRhigh thymocytes and SP CD8+TCRhigh-, CD4+TCRhigh-, and CD8+CD25+RT6.1+-Treg-thymocytes. B. Cross-talk between the β-AR-Gs-cAMP-PKA and the p38 MAPK pathways occurs in the thymus. Although the β-AR signaling with the MAPK pathway in thymocytes has not been clearly determined, norepinephrine is proposed to binds to β-ARs expressed on thymocytes to activate the canonical Gs protein-AC-cAMP—PKA pathway. Cyclic AMP signals PKA then phosphorylates p38, which phosphorylates activating transcription factor-2 (ATF-2). ATF-2 travels to the nucleus to upregulate Fas ligand (FasL) mRNA, which increases cell death. This is a proposed mechanism by which the SNS promotes reduced cellularity and thymocyte numbers and increased apoptosis of thymocytes. Signaling is terminated by the degradation of cAMP to 5′-AMP by phosphodiesterase (PDE). Adapted from LaJevic et al. (2011). Figure 6. Summary of β- and α-AR-Mediated Effects on Cells of Innate Immunity. Sympathetic nerves (center) release norepinephrine as the major neurotransmitter (pale green circles), which binds to β- (left side, orange) or α-adrenergic receptors (ARs) (right side, pink) expressed on cells of the innate immune system to positively (dark green text) or negatively (red text) affect specific cellular functions of (from top to bottom) eosinophils, neutrophils, mast cells/basophils, natural killer (NK) cells, and macrophages/dendritic cells (MAC/DC). EC, endothelial cells; FMLP, formyl-methionyl-leucyl-phenylalanine; Ag, antigen; IFN-γ; LPS,

lipopolysaccharide; TNF-α, tumor necrosis factor-α; IL, interleukin; MIP-1α, macrophage inflammatory protein-1α; I-CAM, intercellular adhesion molecule; Figure 7. Circulating and Marginating Pools of Effector Cells and Release into the Circulation. A. The circulating pool of peripheral blood leukocytes consist of lymphocytes, polymorphonuclear (PMN) cells, granulocytes (top of 7A), and basophils (not shown). Five

Page 72: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

71

cytotoxic effector leukocyte subsets make up the marginal pool: CD8+ effector T-cells, DN (CD4─/CD8─

) TCRγ/δ or gamma/delta T-cells, CD3+CD56+ NKT-like cells, CD16+CD56dim

cytotoxic NK-cells, and CD14dimCD16+ proinflammatory monocytes (bottom of 7A). These subsets are attached to the basement membrane of capillary and venule endothelial cells by adhesion molecules, CX3CR1, and CD11a expressed on immune cells. B. Catecholamines induce the mobilization of lymphocytes subsets from the marginal pool by activating β2-ARs expressed on immune cells, not on the endothelial cells (EC), resulting in a breakdown in adhesion between CD11a expressed on leukocytes and CXC3R1 expressed on EC (4th cell from the left). Activation of β2-ARs stimulates the release of proteases (scissors). Beta-AR agonists mobilize memory/activated CD8+CD29high T-cells within 30 min, but not CD8+ CD45RA+ CD62L+ naïve T-cells, an effect blocked by β-AR antagonists. Epinephrine rapidly attenuates endothelium attachment via CD11a and CX3CR1, permitting demargination and release into the circulation, to rapidly mobilize protective host defense against invading pathogens. Based on findings from Dimitrov et al. (2010). Figure 8. Effects of CD28 and CTLA4 (CD152) Recognition in the “Immunological Synapse on APC Functions. A. Recognition of the T-cell receptor (TCR) with a specific major histocompatibility complex (MHC)–peptide (Ag) complex (signal 1) in the absence of CD28 binding to B7.1 or B7.2 (collectively B7; CD80 or CD86, respectively) causes T-cell anergy or apoptosis. B. Simultaneous recognition of the TCR with a specific MHC-peptide complex and CD28-B7 (signal 2) induces T-cell proliferation (clonal expansion) and differentiation in to mature, functional effector T-cells. C. The upregulation and subsequent cell surface expression of cytotoxic T-lymphocyte antigen 4 (CTLA-4) and its binding to CD40 in the presence of TCR recognition of a specific MHC-peptide complex prevents T cell activation and clonal expansion. Figure 9. Summary of the β-AR-Mediated Effects on Th-Cell Differentiation. In secondary lymphoid organs, antigen-presenting cells (APCs; i.e., dendritic cells, macrophages or B-cells) drive the differentiation of T-helper (Th)0-cells toward specific subtypes of Th effector-cells, depending on the type of antigen, the cytokine milieu and adrenergic receptor (AR) activation. This figure illustrates the influence of AR stimulation at the time of antigen presentation and Th0-cell activation (left side of figure) or after Th0-cell differentiation into effector Th-cells (right side of figure). A-B. Cytokines regulate Th1- and Th-2-cell drive. IL-12 from APCs and IFN-γ

from Th0 cells drive Th1-cell and inhibit Th-2 cell differentiation, whereas tumor necrosis factor-α (TNF-α) and IL-10 from APCs and IL-4 from Th0 cells drive Th2-cell and inhibit Th1-cell differentiation (red lines). A. APC presentation of microbial/viral antigens to Th0-cells induces a

cell-mediated response by promoting APC production of TNF- and IL-12. These cytokines

promote IFN- secretion by Th0-cells. Activation of α1-ARs increases APC TNF-α, while activation of Th0 cell β2-ARs enhances IFN-γ production to drive differentiation towards a Th1-cell phenotype and promote cell-mediated immunity. Also, by increasing IFN-γ production by

Th0-cell, β2-AR activation indirectly inhibits Th2-cell differentiation by reducing IL-4. Th1-cells are characterized by expression of the transcription factor T-bet and cell surface expression of IL-12 receptor (IL-12R). Activation of Th0 β2-ARs results in greater IFN-γ production per Th1-cell. In contrast, β2-AR stimulation of effectorTh1-cells (pink) reduces IFN-γ production by the differentiated Th1-cells. By decreasing IFN-γ in Th1-cells, the SNS inhibits cell-mediated immunity and acts as a negative feedback mechanism to restore homeostasis. B. APC presentation of parasitic or allergenic antigens in the presence of APC produced IL-10, Th0-cell released IL-4 (green circles) drives Th0-cell differentiation towards a Th2-cell phenotype (GATA-3 and IL-4R expression) and an increase in IL-4 production by the differentiating Th-cells. Activation of β2-ARs increases the production of APC IL-10, to promote humoral immunity. Beta2-AR stimulation of effector Th1-cells (right side of figure) indirectly promotes cell-mediated immunity, by reducing IFN-γ production, which would inhibit IL-4 production. C. In the context of

Page 73: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

72

elevated IL-6 and norepinephrine, APC presentation of self-antigens to Th0-cells can drive Th-cell differentiation towards a Th17-cell phenotype (RORγt and IL-23R expression), characterized

by the secretion of transforming growth factor-β (TGF-β), IL-17A, and IL-23. However, β2-AR stimulation of effector Th17-cells (right side of figure) suppresses IL-17A production. D. In the context of elevated retinoic acid and TGF-β, Th0-cells differentiation into Treg-cells. The SNS promotes Th0- to Treg-cell differentiation and β-agonists promote TGF-β and IL-10 production, but the cell type(s) that produce(s) these cytokines is/are not clear. Treg-cells are characterized by increased FoxP3 expression and production of TGF-β and IL-10. FoxP3, forkhead box P3; RORγt, retinoic acid-related orphan receptor γt. Figure 10. Direct and indirect effects of β-adrenergic receptor (AR) stimulation on APC and T-cell functions are illustrated. Simultaneous TCR recognition of a specific MHC-Ag complex and B7 by CD28 induces gene transcription (gray arrows) in both the T-cell and the antigen-presenting cell (APC - dendritic cells (DCs), macrophages or B-cells), which directs specific cellular responses necessary to coordinate an appropriate immune response. Activation of β-ARs via norepinephrine (green circles) binding directly effects gene transcription in the APC (on right) or the T-cell (on left), and the subsequent changes in gene transcription (green arrows) which influences their cellular responses. Additionally, β-AR-mediated changes in the cellular response of the APC can indirectly affect T-cell response (blue arrows). Similarly, β-AR-mediated changes in the cellular response of the activated T-cell can indirectly affect APC response (black arrows). BCR, B-cell receptor. Figure 11. Sympathetic Influence on Antibody Production. A. Schematic showing the molecular mechanism proposed by Sanders et al. (2012) of β2-adrenergic receptor (β2-AR) convergence with the CD86 signaling pathway in an activated B-cell. CD86 ligation increases NF-κB by removing IκB-α-mediated inhibition, and increasing the expression OCA-B, a co-activator protein (a) (left side of figure). CD86 also activates Lyn kinase, CD19, and Akt (not shown). At the time of CD86 ligation, β2-AR stimulation (b) activates adenylate cyclase (AC), which increases cAMP production and consequently activates PKA, which in turn phosphorylates CREB and increases OCA-B, a transcription factor (c). More OCA-B-Oct-2 complexes (d) translocate to the nucleus where they bind to the 3′-IgH-enhancer. OCA-B/Oct-2 binding augments the rate of IgG1 transcription (e), synthesis and levels secreted by the B-cell (f). Beta2-AR stimulation does not affect the number of naïve B-cell that undergo IgM-to-IgG1 switching. B. Schematic demonstrating the molecular mechanisms through which β2-AR (R) signaling modulates the production of IgE in an activated B-cell. STAT6 is essential for IL-4-induced IgE transcription. Upon B-cell activation (a), simultaneous stimulation of β2-ARs (b) activates the cAMP-PKA signal transduction pathway (c). Phosphorylation of PKA activates hematopoietic protein tyrosine phosphatase (HePTP), which increases the available pool of p38MAPK (d) that can be activated by CD40 ligation. Increased p38MAPK phosphorylation along with STAT6 (e) causes an increase in CD23 synthesis and its cleavage from the cell surface (not shown), leading to greater soluble CD23 (sCD23). STAT6/phosphorylated p38MAPK-mediated shedding of CD23 creates a microenvironment that promotes IgE gene transcription (f) and synthesis (g) per B-cell. Blue double-line, cell membrane; blue double, dashed-line, nuclear membrane. L, ligand (norepinephrine); IgE, immunoglobulin E; Gs, Gs protein; AC, adenylate cyclase, PKA, protein kinase A; p38-mitogen-activated protein kinase; STAT6, signal transducer and activator of transcription 6. Both panels are adapted from Sanders et al., 2012.

Page 74: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

73

Figure 1

Page 75: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

74

Figure 2

Page 76: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

75

Figure 3

Page 77: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

76

Figure 4

Page 78: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

77

Figure 5A

Page 79: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

78

Figure 5B

Page 80: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

79

Figure 6

Page 81: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

80

Figure 7

Page 82: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

81

Figure 8

Page 83: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

82

Figure 9

Page 84: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

83

Figure 10

Page 85: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

84

Figure 11A

Page 86: Autonomic regulation of cellular immune function

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

85

Figure 11B


Recommended