+ All Categories
Home > Documents > Neuroendocrine Regulationof Immunity

Neuroendocrine Regulationof Immunity

Date post: 30-Apr-2017
Category:
Upload: nepretip
View: 224 times
Download: 0 times
Share this document with a friend
41
Annu. Rev. Immunol. 2002. 20:125–63 DOI: 10.1146/annurev.immunol.20.082401.104914 NEUROENDOCRINE REGULATION OF IMMUNITY * Jeanette I. Webster, Leonardo Tonelli, and Esther M. Sternberg § National Institute of Mental Health, Section on Neuroimmune Immunology and Behavior, Bldg 36, Room 1A 23 (MSC 4020), 36 Convent Drive, Bethesda, Maryland 20892-4020; e-mail: [email protected]; [email protected]; [email protected] Key Words glucocorticoid, immune response, inflammatory/autoimmune disease, HPA axis, cytokines Abstract A reciprocal regulation exists between the central nervous and im- mune systems through which the CNS signals the immune system via hormonal and neuronal pathways and the immune system signals the CNS through cytokines. The primary hormonal pathway by which the CNS regulates the immune system is the hypothalamic-pituitary-adrenal axis, through the hormones of the neuroen- docrine stress response. The sympathetic nervous system regulates the function of the immune system primarily via adrenergic neurotransmitters released through neu- ronal routes. Neuroendocrine regulation of immune function is essential for survival during stress or infection and to modulate immune responses in inflammatory dis- ease. Glucocorticoids are the main effector end point of this neuroendocrine system and, through the glucocorticoid receptor, have multiple effects on immune cells and molecules. This review focuses on the regulation of the immune response via the neuroendocrine system. Particular details are presented on the effects of interrup- tions of this regulatory loop at multiple levels in predisposition and expression of immune diseases and on mechanisms of glucocorticoid effects on immune cells and molecules. GENERAL INTRODUCTION The immune system has for many years been known to be influenced by gluco- corticoids, and glucocorticoids have been used in the treatment of inflammatory diseases since the 1940s. In fact, Kendall, Reichstein, and Hench received the Nobel Prize for this discovery in 1950 (1). Since then extensive research has shown the pharmacological effects of glucocorticoids on many aspects of immune cell function (2,3). However, until recently the fact that glucocorticoids play an * The US Government has the right to retain a nonexclusive, royalty-free license in and to any copyright covering this paper. § Corresponding author. 125 Annu. Rev. Immunol. 2002.20:125-163. Downloaded from arjournals.annualreviews.org by University of Bern on 12/05/05. For personal use only.
Transcript
Page 1: Neuroendocrine Regulationof Immunity

8 Feb 2002 9:39 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC10.1146/annurev.immunol.20.082401.104914

Annu. Rev. Immunol. 2002. 20:125–63DOI: 10.1146/annurev.immunol.20.082401.104914

NEUROENDOCRINE REGULATION OF IMMUNITY∗

Jeanette I. Webster, Leonardo Tonelli,and Esther M. Sternberg§National Institute of Mental Health, Section on Neuroimmune Immunology and Behavior,Bldg 36, Room 1A 23 (MSC 4020), 36 Convent Drive, Bethesda, Maryland 20892-4020;e-mail: [email protected]; [email protected]; [email protected]

Key Words glucocorticoid, immune response, inflammatory/autoimmune disease,HPA axis, cytokines

■ Abstract A reciprocal regulation exists between the central nervous and im-mune systems through which the CNS signals the immune system via hormonaland neuronal pathways and the immune system signals the CNS through cytokines.The primary hormonal pathway by which the CNS regulates the immune systemis the hypothalamic-pituitary-adrenal axis, through the hormones of the neuroen-docrine stress response. The sympathetic nervous system regulates the function ofthe immune system primarily via adrenergic neurotransmitters released through neu-ronal routes. Neuroendocrine regulation of immune function is essential for survivalduring stress or infection and to modulate immune responses in inflammatory dis-ease. Glucocorticoids are the main effector end point of this neuroendocrine systemand, through the glucocorticoid receptor, have multiple effects on immune cells andmolecules. This review focuses on the regulation of the immune response via theneuroendocrine system. Particular details are presented on the effects of interrup-tions of this regulatory loop at multiple levels in predisposition and expression ofimmune diseases and on mechanisms of glucocorticoid effects on immune cells andmolecules.

GENERAL INTRODUCTION

The immune system has for many years been known to be influenced by gluco-corticoids, and glucocorticoids have been used in the treatment of inflammatorydiseases since the 1940s. In fact, Kendall, Reichstein, and Hench received theNobel Prize for this discovery in 1950 (1). Since then extensive research hasshown the pharmacological effects of glucocorticoids on many aspects of immunecell function (2, 3). However, until recently the fact that glucocorticoids play an

∗The US Government has the right to retain a nonexclusive, royalty-free license in and toany copyright covering this paper.§Corresponding author.

125

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 2: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

126 WEBSTER ET AL.

essential physiological role in regulation of the immune system in health and dis-ease was not fully appreciated. An understanding of the physiological mechanismsinvolved in glucocorticoid secretion and their regulation of the immune systemunder both normal and disease conditions is fundamental to our understanding ofthe pathogenesis of inflammatory disease and ultimately for the development ofeffective therapies for such diseases.

Recent studies have shown that this physiological regulation of the immunesystem by glucocorticoids is only one part of an extensive regulatory networkbetween the central nervous system (CNS), neuroendocrine system, and immunesystem. This network of connections through nerve pathways, hormonal cascades,and cellular interactions allows the CNS to regulate the immune system locallyat sites of inflammation, regionally in immune organs, and systemically thoughhormonal routes. In turn through similar connections, the immune system alsoregulates the CNS. During inflammation, cytokines produced at the inflammatorysite can signal to the brain and produce the symptoms of sickness behavior andfever (4, 5). Cytokines are also expressed in areas of the brain, e.g., glia, neurons,and macrophages, and play a role in both neuronal cell death (6, 7) and survival(8). Cytokine-mediated neuronal cell death is thought to play an important rolein several neuro-degenerative diseases, such as neuro-AIDS, Alzheimer’s, mul-tiple sclerosis, stroke, and nerve trauma. In addition to this growth-factor rolewhen expressed within the CNS, cytokines produced in the periphery can func-tion as hormones and can stimulate the CNS by several mechanisms. They canpass the blood-brain barrier (BBB) at leaky points, for example at the organumvasculosum lamina terminalis (OVLT) or median eminence. They may be activelytransported across the BBB in small amounts (9). In addition they can rapidlysignal the CNS through the vagus nerve (10, 11). They can influence the brainby activation of second messengers, such as nitric oxide and prostaglandins, afterbinding to receptors on endothelial cells (12, 13). While a full understanding of thisbidirectional communication between the CNS and immune systems is important,this chapter focuses only on the regulation of the immune system by the CNS,mainly though the neuroendocrine system and the adrenergic system. For furtherinformation on the regulation of the CNS by the immune system see the review byMulla & Buckingham (14).

The CNS regulates the immune system through two major mechanisms: (a) thehormonal stress response and the production of glucocorticoids, and (b) the auto-nomic nervous system with the release of noradrenalin. The CNS can also regulatethe immune system locally via the peripheral nerves with release of neuropeptidessuch as substance P and locally produced corticotrophin-releasing hormone (CRH).This latter mechanism is not the focus of this review; for further information onthese refer to the following articles (15, 16).

The main regulator of the glucocorticoid effect on the immune system is thehypothalamic-pituitary-adrenal axis (HPA axis) (Figure 1). The main componentsof the HPA axis are the paraventricular nucleus (PVN) in the hypothalamus of thebrain, the anterior pituitary gland located at the base of the brain, and the adrenal

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 3: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 127

Figure 1 Diagram of the routes of communication between the brain and immune system,including the HPA axis, sympathetic nervous system, and cytokine feedback to the brain.

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 4: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

128 WEBSTER ET AL.

glands. CRH is secreted from the PVN of the hypothalamus into the hypophysealportal blood supply and stimulates the expression of adrenocorticotropin hormone(ACTH) in the anterior pituitary gland. ACTH then circulates in the bloodstream tothe adrenal glands where it induces the expression and release of glucocorticoids.

The HPA axis is subject to regulation both from within the central nervous sys-tem and from the periphery. Glucocorticoids, themselves, feed back to negativelyregulate the HPA axis, exerting their negative feedback at the hypothalamic andpituitary level. The HPA axis can also be regulated by other factors such as the sym-pathetic nervous system, cytokines, and other neuropeptides, such as arginine va-sopressin (AVP) (17). CRH is also negatively regulated by ACTH and itself, as wellas by other neuropeptides and neurotransmitters in the brain, e.g.,γ -aminobutyricacid-benzodiazopines (GABA-BDZ) and opioid peptide systems. CRH is posi-tively regulated by serotonergic, cholinergic, and histaminergic systems (18).

The connections between the neuroendocrine system and immune system pro-vide a finely tuned regulatory system required for health. Disturbances at anylevel of the HPA axis or glucocorticoid action lead to an imbalance of this systemand enhanced susceptibility to infection and inflammatory or autoimmune disease.Overstimulation of the HPA axis with excessive amounts of circulating glucocorti-coids and overall suppression of immune responses lead to enhanced susceptibilityto infection, whereas understimulation results in lower circulating levels of glu-cocorticoids and susceptibility to inflammation. Dysregulation may also occur atthe molecular level and in this instance would result in glucocorticoid resistanceat a molecular level leading to enhanced susceptibility to inflammation. A detailedunderstanding, therefore, by which the CNS and neuroendocrine systems regu-late the immune system at the systemic, anatomical, cellular, and molecular levelswill inform not only the pathogenesis and treatment of inflammatory/autoimmuneconditions and infectious disease but also conditions predisposing to susceptibilityand resistance to these illnesses.

EVIDENCE FOR ROLE OF ENDOGENOUSGLUCOCORTICOIDS IN REGULATIONOF IMMUNE FUNCTION

Changes in the levels of circulating glucocorticoids, such as during exercise andwith circadian rhythms, are associated with changes in cytokine levels and pro-duction by leukocytes (19–23). Such studies provide circumstantial evidence thatphysiological fluctuations in glucocorticoid levels, in the range of those seen withcircadian variations or stress, are associated with altered immune function. How-ever, animal models have provided the strongest proof that endogenous glucocor-ticoids are essential physiological regulators of the immune response and inflam-matory/autoimmune disease.

Inbred rat strains, in which altered neuroendocrine responsiveness is associ-ated with differential susceptibility and resistance to autoimmune/inflammatory

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 5: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 129

disease, provide a naturalistic, genetically uniform system that can be systemi-cally manipulated to test the role of neuroendocrine regulation of various aspectsof immunity. Lewis (LEW/N) rats are an inbred strain of rats that are highly sus-ceptible to development of a wide range of autoimmune/inflammatory diseasesin response to a variety of antigenic or proinflammatory stimuli. Largely histo-compatible Fischer (F344/N) rats are relatively resistant to these same illnessesafter exposure to the same dose of antigens. These two strains also show relateddifferences in HPA axis responsiveness, with inflammatory-susceptible LEW/Nexhibiting a blunted response, compared to inflammatory-resistant F344/N ratswith an excessive HPA response compared to outbred rats (24–28). Differences inthe expression of hypothalamic CRH (26), pro-opiomelanocortin (POMC) (28),corticosterone-binding globulin (CBG) (27), and glucocorticoid receptor (GR)expression and activation (27, 29, 30) have been shown in these two rat strains.A variety of surgical or pharmacological HPA axis interventions in these strainsof rats, as well as in mouse strains, alter the course and severity of inducibleautoimmune/inflammatory disease. Thus, in F344/N rats, treatment with the glu-cocorticoid antagonist RU486 is associated with high mortality and developmentof arthritis in response to injection of streptococcal cell walls (25). Approximately50% of rats die after infection withSalmonella typhimurium, but adrenalectomizedrats suffer 100% lethality after infection with this bacteria (31). Similarly, in mice,adrenalectomy before infection with CMV virus results in lethality but glucocorti-coid replacement prevents virus-induced lethality (32). The high rates of mortalitywithin 12–24 h of exposure to this wide range of antigenic, proinflammatory, orinfectious stimuli across species and strains in which the HPA axis has been inter-rupted pharmacologically or surgically, indicates the importance of an intact HPAaxis to protect against septic shock.

It is also possible in animal models to attenuate inflammatory disease by re-constituting the HPA axis, pharmacologically with glucocorticoids, or surgicallyby intracerebral fetal hypothalamic tissue transplantation. In LEW/N rats eithertreated with low-dose dexamethasone (25) or transplanted intracerebroventricu-larly with F344/N hypothalamic tissue (33), arthritis and carageenan inflammationare significantly attenuated. Experimental allergic encephalomyelitis (EAE) is in-ducible in LEW/N rats by immunization with myelin basic protein. After initialimmunization these animals experience transient paralysis but then recover to someextent. During the development of the disease and recovery, the production of en-dogenous corticosterone increases, which is essential for survival of the animal. Ifthis endogenous production is interrupted by adrenalectomy, the development ofEAE is fatal. Whereas a subcutaneous steroid implant equivalent to the basal cor-ticosterone levels does not reduce mortality, a dose equivalent to the EAE-inducedcorticosterone levels results in survival rates similar to that of normal animals, butEAE still develops. If a subcutaneous implant of even higher corticosterone levelsis used, however, the animal will undergo complete remission (34).

Such animal studies showing that interruption of the HPA axis predisposesto worse inflammation, while reconstitution attenuates autoimmune/inflammatory

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 6: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

130 WEBSTER ET AL.

disease, lend support to the role of glucocorticoid imbalance in exacerbation ofhuman autoimmune, inflammatory, allergic, or infectious diseases. Thus, a bluntedHPA axis response to pituitary adrenal axis stimulation with CRH or insulin, orby psychological stress, has been shown in a variety of autoimmune diseases in-cluding rheumatoid arthritis, SLE, Sjogren’s syndrome, fibromyalgia, and chronicfatigue syndrome, as well as in allergic asthma and atopic dermatitis. Conversely,in humans, excess chronic stimulation of the hormonal stress response with con-comitant chronically elevated glucocorticoids, as occurs in chronic stress situationsis associated with an enhanced susceptibility to viral infection, prolonged woundhealing, or decreased antibody production after vaccination (35–38). Such stressis experienced by caregivers of Alzheimer’s patients, students taking exams, cou-ples during marital conflict, and Army Rangers undergoing extreme exercise andstress. One important mechanism by which activation of the HPA axis regulatesthese immune responses and severity of expression of resultant disease is throughthe effects of glucocorticoids at the molecular level though the glucocorticoid re-ceptor (GR). The next sections describes this important receptor system and itsregulation of target gene expression.

PHARMACOLOGICAL VERSUS PHYSIOLOGICALEFFECTS OF GLUCOCORTICOID

When considering the physiological relevance of the effects of glucocorticoidson immunity, it is important to recognize that glucocorticoids in pharmacologi-cal doses or forms exert different effects than they do under physiological condi-tions. Physiological concentrations of glucocorticoids in the range of 350 nmol/l to950 nmol/l, such as occur during physical or psychological stress, result in mod-ulation of transcription of genes involved in the inflammatory response, whereaspharmacological doses (higher concentration than physiological) result in a totalsuppression of the inflammatory response. There are also differences in potency ofimmune suppression by synthetic glucocorticoids, such as dexamethasone, versusthe effects on immune response to natural glucocorticoids, such as hydrocorti-sone. For example, the synthetic glucocorticoid dexamethasone exerts a greatersuppression of IL-12 than does the natural glucocorticoid hydrocortisone, whichis consistent with the greater affinity of dexamethasone than hydrocortisone forGR (39).

MODULATION OF THE IMMUNE SYSTEMBY GLUCOCORTICOIDS

There are two receptors for glucocorticoids, the glucocorticoid receptor and themineralocorticoid receptor (MR). Corticosterone has a lower affinity for MRthan for GR. Thus, at low levels, glucocorticoids bind preferentially to MR, and

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 7: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 131

only at high levels, i.e., during stress, is GR occupied (39, 41). MR and GR, whichare colocalized in some areas of the brain and in lymphocytes, can bind as het-erodimers to DNA (42, 43), which could have implications for gene transcriptionor transrepression. In the brain, MR has been implicated in a proactive mode, inthe prevention of disturbance of homeostasis, whereas GR has been suggested towork in a reactive mode in the recovery from a disturbance (39, 44). The primaryreceptor for glucocorticoids in immune cells is GR. The availability of glucocorti-coids is also dependent on the expression of 11β-hydroxysteroid dehydrogenase,an enzyme responsible for the conversion of steroids from the active form, e.g.,cortisol and corticosterone, into an 11-keto inactive form, e.g., cortisone and 11-dehydrocortisone. Two forms of this enzyme exist. The type I enzyme is expressedin liver, brain, adipose tissue, lung, and other glucocorticoid-target tissues and cat-alyzes the regeneration of active glucocorticoids from the inactive 11-keto form.Conversely, the type II enzyme catalyzes the inactivation of glucocorticoids to theinert 11-keto form (45).

Glucocorticoid Receptor and Mechanism of Action

The end point tissue effect of glucocorticoids is mediated by GR (NR3C1) (46).This is a member of the steroid and thyroid hormone receptor superfamily alongwith the progesterone, estrogen, mineralocorticoid, and thyroid receptors, and GRessentially is a ligand-dependent transcription factor. These receptors all have asimilar structure that can be divided into three distinct regions (Figure 2). TheN-terminal domain is involved in transactivation; the middle section is termedthe DNA-binding domain (DBD) and is involved in DNA binding mediated viatwo zinc fingers; and the C-terminal domain or ligand-binding domain (LDB) isresponsible for ligand binding as well as being also involved in transactivation,dimerization, and hsp90 binding (2, 47).

Glucocorticoids circulate in the plasma associated with CBG or albumin. It isgenerally thought that glucocorticoids enter the cell by passive diffusion,

Figure 2 Structure of the glucocorticoid receptor.

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 8: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

132 WEBSTER ET AL.

although there is evidence for an active transport out of the cell. GR is located inthe cytoplasm in the unactivated state in a multiprotein complex containing hsp90and immunophilins. These are thought to hold the GR in a conformation that isavailable to the ligand. Upon ligand binding, GR dissociates from this complexand translocates to the nucleus where it binds as a homodimer to target elementsor glucocorticoid response elements (GREs) via its zinc fingers of the DBD. Thebound GR homodimer can then modulate gene expression by modulating the basaltranscription machinery either directly or via cofactors (Figure 3). GR has beeninvolved in both the upregulation and downregulation of genes. Downregulation ofgene expression can occur via so-called negative glucocorticoid response elements(nGRE), e.g., the POMC gene, but mostly gene repression by GR occurs via its in-teraction with other transcription factors such as AP-1 and NFκB (2, 48). It is note-worthy that the reverse can also occur, i.e., AP-1 and NFκB are able to repress GRfunction.

When GR was first cloned, two clones for GR were found that differed in theC terminus (49). This second receptor, GRβ, is a splice variant of GR that isidentical to GR but is lacking the last 50 amino acids. Instead it has a unique 15–amino acid C terminus. This receptor is located in the nucleus regardless of ligand

Figure 3 Schematic diagram of the mechanisms of action ofthe glucocorticoid receptor.

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 9: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 133

status, but it can also be found in the cytoplasm complexed to hsp90. It does not bindligand and does not activate gene transcription but may act as a dominant negativereceptor in vitro by forming transcriptionally inactive heterodimers with GRα

(50–52). This mechanism of a dominant negative receptor is still under dispute asother studies have shown no effect of GRβ on GRα-mediated transactivation ortransrepression (53–55).

AP-1

GR can modulate gene expression via interaction with other factors, such as NFκBand AP-1. AP-1 consists of a heterodimer of the oncoproteins c-Fos and c-Jun thatbind to an AP-1 consensus binding site in DNA to activate gene transcriptionin response to ligands such as phorbol esters. Although the high-affinity Fos/Junheterodimers predominate, the lower-affinity Jun/Jun homodimers are also capableof binding to the AP-1 site. Glucocorticoids can reduce the DNA-binding abilityof the Fos and Jun complexes by protein-protein interactions and thereby repressAP-1-mediated gene activation (56, 57) (Figure 4). This can also occur through acomposite GRE. This sequence contains a binding site for the receptor and alsofor a nonreceptor factor. For example, GR was shown to repress AP-1 activity atthe composite GRE in the promoter of the plfG gene; this repression was mediatedby the N-terminal domain of GR and could not be mediated by MR (58, 59).

One example of a gene where a composite HRE is involved in the repressionof the gene by glucocorticoids is the hypothalamic hormone CRH. The promoterof this gene contains an AP-1 site closely located to a GRE, and repression of theAP-1-activated gene transcription by glucocorticoids has been shown (60). Thiscould be the mechanism by which glucocorticoids exert their negative feedbackon the hypothalamus to downregulate the HPA axis.

NFκB

Glucocorticoids play a role in immunosuppression through their repression ofNFκB, which is a major factor involved in the regulation of cytokines and other im-mune responses. [For comprehensive reviews on NFκB, see Baldwin (61), Ghoshet al. (62), and McKay & Cidlowski (48)]. NFκB was originally described as adimer of two proteins p65 (RelA) and p50 (NF-κB1), but a whole family of theseproteins has now been described. These proteins all contain a highly conserved300–amino acid domain termed the Rel homology domain (RHD), which is impor-tant for DNA binding and nuclear translocation and dimerization. The proteins p65(Rel A), C Rel, and Rel B contain a C-terminal transactivation domain, whereasp50 (NF-κB1) and p52 (NF-κB2) are present as precursors (p105 and p100, respec-tively), which contain a C-terminal IκB-like region that is removed by proteolysis.The ability of these proteins to form various dimer partners and the specificityof these dimers for different DNA sequences may be fundamental to the cell-specific responses mediated by NFκB. These proteins are present in the cytoplasmas complexes together with the inhibitory protein IκB. This is also now knownto be part of a family of proteins that include IκBα, IκBβ, IκBγ , IκB-R, and the

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 10: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

134 WEBSTER ET AL.

Figure 4 Schematic diagram of the mechanism(s) by which the glucocorticoid receptorinhibits the action of NFκB and AP-1.

C-termini of p105/p50 and p100/p52. These all contain multiple ankyrin repeats,which are involved in protein-protein interactions between the IκB and NFκB, anda C-terminal PEST sequence, which is a signal for protein degradation (48).

NFκB is normally located in the cytoplasm associated with the inhibitor IκBprotein. Upon activation, IκB is phosphorylated, ubiquinated, and then degraded.The free NFκB can then translocate to the nucleus where it activates gene expres-sion. NFκB can be stimulated by a variety of factors, including proinflammatorycytokines such as TNF-α and IL-1, physical or oxidative stress, and bacterial orviral proteins (48, 63) (Figure 4).

There have been two schools of thought regarding GR-mediated repression ofNFκB, and data exist to support both. One is that glucocorticoids via GR inducethe expression of the inhibitory protein IκB that then sequesters NFκB in thecytoplasm and prevents it from translocating to the nucleus and inducing gene

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 11: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 135

activation (63, 64). This mechanism of repression of NFκB by GR may be limitedto certain cell types, particularly monocytes and lymphocytes. The other modelsuggests that there is a physical interaction or cross-talk between NFκB and GRthat prevents gene expression (63, 65–71). In some cases GR-induced transcriptionis not required, as the anti-glucocorticoid RU486 is also capable of inhibitingNFκB to some extent (67, 72). The region of the GR required for GR repression ofNFκB has been located to the zinc finger region of the DNA-binding region of GR(63, 67). This region has been further defined as two amino acids in the C-terminalzinc finger that are absolutely critical for GR-mediated repression of NFκB; theseresidues are not involved in GR-mediated repression of AP-1 (73). It is plausiblethat these two models are not mutually exclusive or that they are dependent oncell type. One experiment in the A549 pulmonary epithelial cell line has shownthat both these models exist in the same system. In this case, dexamethasone doesinduce IκB expression, but when protein synthesis and therefore IκB productionis blocked, GR is still able to repress NFκB-mediated gene activation although notto the same extent as when protein synthesis is allowed (74).

It has also been suggested that glucocorticoid repression of NFκB activitycould be caused by competition between GR and NFκB for limited cofactors suchas CREB-binding protein (CBP) and steroid receptor coactivator 1 (SRC-1), asit has been shown in Cos cells that this repression can be alleviated by excesscofactor (75). McKay & Cidlowski, however, showed that CBP did not mediateGR repression of NFκB by a competitive model, but rather that CBP functioned asan integrator to enhance the physical interaction between GR and NFκB (76). Thecatalytic subunit of protein kinase A (PKAc) has also been suggested to promotethe cross-talk between GR and NFκB (77).

REGULATION OF IMMUNE-RELATED GENES

Glucocorticoids regulate a wide variety of immune cell functions and expressionof immune molecules through the molecular mechanisms described above. Thus,glucocorticoids modulate cytokine expression, adhesion molecule expression andimmune cell trafficking, immune cell maturation and differentiation, expressionof chemoattractants and cell migration, and production of inflammatory mediatorsand other inflammatory molecules [for reviews see Barnes (78) and Adcock (2)].

Cytokine Expression

Glucocorticoids modulate the transcription of many cytokines. They suppress theproinflammatory cytokines IL-1, IL-2, IL-6, IL-8, IL-11, IL-12, TNF-α, IFN-γ ,and GM-CSF while upregulating the anti-inflammatory cytokines IL-4 andIL-10.

PROINFLAMMATORY CYTOKINES Glucocorticoids mediate the anti-inflammatoryresponse by downregulating the expression of proinflammatory cytokines such

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 12: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

136 WEBSTER ET AL.

as IL-1, IL-6, and TNF-α. Glucocorticoids have been shown to downregulate thetranscription of IL-1β, to destabilize IL-1β mRNA (79–81), and to downregulateIL-1α (82). They also upregulate the expression of the decoy IL-1 receptor, IL-1RII (83).

Glucocorticoids regulate IL-6 production not only directly, but also indirectly,through their effects on the cytokines that regulate IL-6. IL-1 or TNF induction ofIL-6 is mediated via NFκB and by another nuclear factor, NF-IL6, in transfectionstudies of these nuclear factors and the IL-6 promoter in HeLa and F9 cells. ThisIL-1-induced induction of IL-6 can be inhibited by glucocorticoid-activated GRvia protein-protein interactions at the C-terminal transactivation domain of NFκB(65, 69, 84). This repression does not require protein synthesis and does not affectthe DNA-binding capacity of NFκB (69). Glucocorticoids also inhibit expressionof IL-11, a member of the IL-6 cytokine family, by inhibition of gene expressionand by destabilization of mRNA (85).

Evidence for these molecular mechanisms of GC regulation of cytokines hasappeared in vivo in disease states. In rheumatoid arthritis patients, administrationof glucocorticoids leads to a decrease in the release of TNF-α into the bloodstream(86). The LPS-stimulated production of TNF-α in monocytes is reduced by gluco-corticoid treatment. The promoter of TNF-α does not contain a typical GRE site,and so this repression by glucocorticoids may occur via other factors involved inthe regulation of TNF-α which have binding sites in the promoter, such as NFκBand AP-1 (87). However, some evidence suggests that the glucocorticoid-mediatedrepression of TNF-α could occur at the level of translation rather than transcription(88).

The transcription of other proinflammatory cytokines is also affected by gluco-corticoids. IFN-γ expression in spleen cells, and blood monocytes is inhibited byglucocorticoids (89, 90). Dexamethasone inhibits basal IL-8 in airway epithelialcells by destabilizing the mRNA via new protein synthesis (91). The expressionof the Th1 cytokine IL-12 and its receptor is downregulated by glucocorticoids(92–94). Effects of glucocorticoids on IL-12 are fully discussed in the section onTh1-Th2 shift.

IL-2 expression in spleen cells is inhibited by glucocorticoids (89), and dexam-ethasone inhibits the ionomycin- and TPA-induced IL-2 expression in FJ8.1 cells.With the promoter for IL-2, the dexamethasone-induced repression inhibits thebinding of NFκB and reduces the binding of AP-1 to DNA. Glucocorticoids notonly affect the action of cytokines by affecting their expression, but they also caninhibit their signaling mechanisms. Thus IL-2 signaling via the Jak-STAT cascadecan be inhibited by dexamethasone (95).

Granulocyte-macrophage colony stimulating factor (GM-CSF) is a proinflam-matory mediator. IL-1β-stimulated GM-CSF in bronchial epithelial cells is inhib-ited by dexamethasone by transcriptional mechanisms (96).

ANTIINFLAMMATORY CYTOKINES The major Th2 cytokine is IL-10. In blood mono-cytes varying effects of glucocorticoids, both synthetic and natural, on IL-10 have

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 13: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 137

been reported that are dependent on the dose. Low physiological doses of glucocor-ticoids suppress IL-10 expression, whereas high pharmacological doses enhanceIL-10 expression (39, 97a).

IL-4 is a Th2-associated cytokine that is important for the proliferation ofmast cells and attraction of eosinophils to areas of inflammation during responseto IgE stimuli or allergic reactions. Dexamethasone downregulates IL-4 mRNAin mast cells (98, 99) and in lymphocytes (82). The effect of dexamethasone onIL-4 production in T cells is a matter of some controversy and can be at leastpartially explained by consideration of the doses used experimentally. As IL-4is a Th2-associated cytokine and since glucocorticoids induce a shift from Th1-Th2 immunity (see later section), one might expect dexamethasone to induceIL-4 expression. Indeed, in the lymph nodes and spleen of mice IL-4 is inducedin response to physiological concentrations of glucocorticoids both in vivo and invitro (100, 101). On the other hand, consistent with the efficiency of glucocorticoidsin the treatment of allergic diseases, data also exist showing that stress levels ofdexamethasone downregulate IL-4 expression in T cells (89).

Cell Adhesion Molecules and Immune Cell Trafficking

Glucocorticoids reduce the trafficking of leukocytes to areas of inflammation.This occurs via the downregulation of protein molecules involved in the attractionand adhesion of leukocytes to these areas. Dexamethasone inhibits the expressionof intracellular adhesion molecule 1 (ICAM-1), endothelial-leukocyte adhesionmolecule 1 (ELAM-1) (102), and vascular adhesion molecule 1 (VCAM-1) (103).Glucocorticoids can inhibit the expression of ICAM-1 through inhibition of theNFκB site in the promoter of the ICAM-1 gene (66, 71, 74). Glucocorticoids in-hibit TNF-α–induced VCAM-1 expression but not ICAM-1 in bronchial epithe-lial cells. NFκB sites have been identified in the promoter of VCAM-1, and ithas been suggested that glucocorticoid repression of this gene may occur throughGR-mediated repression of NFκB (103).

E-selectin is an endothelial cell surface adhesion molecule that is important forthe recruitment of leukocytes from the blood. It is upregulated by IL-1β and TNF-αand its expression can be repressed by glucocorticoids. Repression by glucocor-ticoids does not affect NFκB translocation or binding to DNA, which suggestsinterference in transcriptional activation of NFκB (70). L-selectin is involved inthe attachment of blood leukocytes during inflammation, and its expression is de-creased by glucocorticoid treatment in bone marrow cells and polymorphonuclearcells (104).

Chemoattractants and Cell Migration

Cytokine-induced neutrophil chemoattractant (CINC)/gro is a chemoattractant forneutrophils and is therefore important for the accumulation of neutrophils at sitesof inflammation. It is also proposed to be a member of the family that includesIL-8. mRNA for CINC/gro is induced by IL-1β via NFκB, and this induction can

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 14: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

138 WEBSTER ET AL.

be inhibited by glucocorticoids, which prevent the translocation of NFκB to thenucleus and therefore prevent DNA binding and gene transcription (105).

IL-5 is a chemoattractant for eosinophils and is important for the accumula-tion of eosinophils in inflammatory diseases such as asthma. IL-5 mRNA can bedownregulated by glucocorticoids in mast cells (99) and T cells (106). IL-5 canbe induced by TCR and IL-2 by different mechanisms, and both of these canbe repressed by glucocorticoids. The mechanism of glucocorticoid repression ofTCR-induced IL-5 possibly occurs through AP-1 and NFκB, but these factors arenot involved in the glucocorticoid repression of IL-2-induced IL-5 (107).

RANTES (regulated upon activation normal T cell expressed and secreted) is achemoattractant involved in the recruitment of eosinophils to areas of inflamma-tion. Dexamethasone inhibits mRNA expression in activated and nonactivated cells(108, 109). The cytokines monocyte chemoattractant protein 1 (MCP-1), MCP-2, and MCP-3 are downregulated by dexamethasone (108). Downregulation ofMCP-1 occurs posttranscriptionally by destabilization of the mRNA at the 5′ end,although the exact mechanism is unknown (110).

Eotaxin is an eosinophil chemoattractant involved in the accumulation of eosin-ophils in the airways in response to allergic stimuli. Eotaxin mRNA is stimulatedby cytokines such as TNF-α and IL-1β and this cytokine-induced induction isrepressed by glucocorticoids (111).

Production of Inflammatory Mediators

Glucocorticoids also affect the production of inflammatory mediators includingprostaglandins and nitric oxide. Glucocorticoids suppress prostaglandin synthesisat sites of inflammation by several mechanisms. The key stages during the synthesisof prostaglandin are the release of arachadonic acid from membrane phospholipids,which is catalyzed by phospholipase A2 (PLA2), and the subsequent conversion toPGH2 by the action of cyclooxygenase (COX). There are two isoforms of PLA2,secretory PLA2 and cytosolic PLA2. There are also two COX enzymes; COX-1is constitutively expressed, and COX-2 is inducible and thought to be involvedin stimuli-induced prostaglandin synthesis. COX-2 induction by inflammatorystimuli is mediated though NF-IL6 and NFκB regulatory sequences in its promoter(112). Glucocorticoids exert their effect by repression of cytosolic PLA2 and COX-2 mRNA levels (113–115).

Nitric oxide synthase II (NOS II) is the enzyme that produces nitric oxide (NO).NO has been implicated in autoimmune and inflammatory responses. The promoterof NOS II contains a binding site for NFκB, which is required for the cytokineinduction of this gene. Glucocorticoids repress the cytokine induction of NOS II,and this repression does not correspond with an induction of the IκB gene, therebyimplicating a method of protein-protein interaction for this repression (68). iNOSis an inducible form of nitric oxide synthase and is another enzyme involved in thesynthesis of nitric oxide. Dexamethasone inhibits transcription of the iNOS genein rat hepatocytes by the induction of IκB (116).

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 15: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 139

Lipocortin 1 (or annexin 1) is expressed in many immune cells and is alsoexpressed in the neuroendocrine system in the hypothalamus and pituitary. It hasanti-inflammatory actions although these mechanisms are not fully understood.One way in which lipocortin 1 exerts its anti-inflammatory action in the periph-ery is by the inhibition of prostaglandin synthesis via the inhibition of release ofarachadonic acid, although the exact mechanism is not known. Glucocorticoidshave a dual action on lipocortin 1. Lipocortin 1 is normally expressed and local-ized mainly in the cytoplasm, but it is also localized to the external surface of cellmembranes. Upon glucocorticoid exposure, the amount of lipocortin 1 on the ex-ternal membrane rapidly increases. This externalization is followed by an increasein lipocortin 1 synthesis by the cells in response to glucocorticoids (117).

Other Factors Involved in the Inflammatory Response

Theβ2-adrenoreceptor is involved in the adrenergic control of the immune system(see later section). Glucocorticoids increase the transcription of theβ2-adrenore-ceptor in many cell types, including airway epithelial cells. This transcriptionalregulation occurs via a GRE sequence in the promoter of the gene (118, 119).Other receptors involved in the regulation of the immune system are regulated byglucocorticoids. The transcription of the NK1-receptor, the receptor for substanceP, is inhibited by glucocorticoids probably through an AP-1 site (120). The NK2-receptor is also downregulated by glucocorticoids (121).

EFFECTS ON IMMUNE CELLS

Glucocorticoids, in general, suppress maturation, differentiation, and proliferationof immune cells involved in all aspects of immunity, including innate, T cell, andB cell function and chronic allergic reactions.

Innate Immunity

Monocytes are produced in the bone marrow. Once mature they leave, circulate inthe bloodstream, and after a time enter the tissues and become macrophages.During inflammation, circulating monocytes migrate to the inflammatory sitewhere they become activated. Glucocorticoids interfere with the protective anddefense mechanisms of activated macrophages, rather than resident macrophages.This occurs via their effects on cytokines and other inflammatory mediators suchas prostaglandin synthesis (122). Glucocorticoids at pharmacological concentra-tions induce apoptosis in macrophages and monocytes; and the pro-inflammatorycytokine IL-1β may be involved in prevention of apoptosis, whereas the anti-inflammatory cytokines IL-4 and IL-10 induce apoptosis in these cells (81). Thus,pharmacological or stress levels of glucocorticoids reduce circulating numbersof monocytes, inhibit secretion of IL-1, IL-6, TNF-α, and monocyte chemotacticactivating factors, and impair synthesis of collagenase, elastase, and tissue plas-minogen activator (123).

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 16: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

140 WEBSTER ET AL.

Neutrophils are involved in the tissue damage that occurs during the innateinflammatory phase of inflammatory disease. In response to injury, neutrophilsmigrate out of the circulating bloodstream, extravasate between endothelial cells,and move to the site of inflammation. Glucocorticoids affect the activation ofneutrophils and also the functions of neutrophils, such as chemotaxis, adhesion,transmigration, apoptosis, and phagocytosis (123, 124). Glucocorticoid regulationof these functions in neutrophils is due to numerous components including regula-tion of cytokines. However, lipocortin 1 also plays a pivotal role in glucocorticoid-induced responses. Glucocorticoids have a dual effect on neutrophils. On onehand pharmacological doses are inhibitory and suppress the inflammatory responsecaused by neutrophil activation and migration. On the other hand, neutrophils arerequired for the response to bacterial infections, and as such their circulating num-bers are increased by pharmacological doses of glucocorticoids through inhibitionof apoptosis (124).

Th1-Th2

Glucocorticoids induce a shift from a Th1 to a Th2 pattern of immunity. Th1immunity or cellular immunity is characterized by the expression of proinflamma-tory cytokines, such as IFN-γ , IL-2, and TNF-β, which lead to the differentiationof macrophages, natural killer cells (NK cells), and cytotoxic T cells that are in-volved in phagocytosis and destruction of invading bacteria or foreign bodies.Th2 immunity or humoral immunity is characterized by the production of anti-inflammatory cytokines such as IL-4, IL-10, and IL-13, resulting in the differenti-ation of eosinophils, mast cells, and B cells, which lead to an antibody-mediateddefense against foreign antigens. The main inducer of Th1 immunity is IL-12.This can induce the expression of IFN-γ and inhibit the expression of IL-4, whichplays a critical role in the immune response. [For a detailed review on IL-4, seeNelms et al. (125)].

As previously mentioned, glucocorticoids can affect the transcription of manycytokines, generally upregulating antiinflammatory cytokines and downregulatingproinflammatory cytokines, thereby causing a shift from Th1 to Th2 immunity.The glucocorticoid-induced shift of Th1 to Th2 may be due mainly to down-regulation of the Th1 cytokines, thus allowing dominant expression of the Th2cytokines (126, 127). They modulate the expression of IL-12 or its receptor, andthis regulation of IL-12 is thought to be a major mechanism by which glucocor-ticoids mediate the Th1-Th2 shift (94). This glucocorticoid-mediated downreg-ulation of IL-12 occurs by inhibition of Stat4 phosphorylation in the signalingcascade downstream of IL-12, and this inhibition is mediated by GR. This is spe-cific for the Th1-mediated immunity because phosphorylation of Stat6, whichis involved in the signal cascade from IL-4, is not affected by glucocorticoids(93). Glucocorticoids downregulate the IL-12 receptorβ1- andβ2-chains, butthis downregulation occurs after 3 days of treatment, whereas the inhibition of

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 17: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 141

Stat4 phosphorylation is seen within 2 h, suggesting that IL-12 receptordownregulation occurs in response to prolonged glucocorticoid exposure (92, 93).Several other neural factors besides glucocorticoids can also induce a Th1-Th2shift; these neural factors include the sympathetic neuropeptides, noradrenalin,and adrenalin (94).

Glucocorticoids differentially affect the survival of Th1 and Th2 cells. Th2NK1.1+T cells, which produce IL-4, are resistant to dexamethasone-induced apop-tosis, and this resistance may be due to the expression of the proto-oncogene Bcl-2(128).

Alterations in Th1-Th2 immunity are characteristic of some autoimmune dis-eases. Rheumatoid arthritis, multiple sclerosis (MS), and type I diabetes mellitusare examples of autoimmune diseases in which there is a shift toward Th1-mediatedimmunity with an excess of IL-12 and TNF-α production. Systemic lupus erythe-matosus (SLE) is shifted toward Th2-mediated immunity with an excess of IL-10production (94). In situations where there is an excess of glucocorticoid produc-tion, e.g., in animal models with a hyperactive HPA axis (F344/N rats) or in womenin the third trimester of pregnancy, there is a relative resistance to Th1-associatedautoimmune diseases. Conversely, animals with a lack of glucocorticoids or a hy-poactive HPA axis (LEW/N rats) are susceptible to Th1-associated autoimmunediseases (94, 129). LEW/N rats are susceptible to EAE, a Th1-mediated autoim-mune disease. Spontaneous recovery of these animals is correlated with an increasein glucocorticoids, whereas adrenalectomy results in fatal regression of the diseasebut can be reversed by administration of glucocorticoids, indicating the shift fromTh1 to Th2 immunity (34).

Allergy

Eosinophils are critical in the response to allergic stimuli, and glucocorticoidsboth systemically and topically applied are effective in the treatment of thesediseases. Pharmacological doses of glucocorticoids reduce the circulating numbersof eosinophils probably through many factors including IL-5, IL-3, and GM-CSF.Glucocorticoids also sequester eosinophils in primary and secondary lymphoidtissues, induce apoptosis, and inhibit the recruitment of eosinophils to areas ofinflammation (123, 130, 131).

Basophils are the least abundant circulating leukocyte but are important in IgE-mediated allergic inflammation. Their activation leads to the synthesis and releaseof lipid mediators such as leukotriene C4 (LTC4), which induces changes in muscleand vascular tissue. Glucocorticoids reduce circulating numbers of basophils, im-pair histamine and leukotriene release, and inhibit basophil migration (123, 131).

Mast cells are important for the mediation of inflammatory disease particularlyin response to IgE. Glucocorticoid treatment results in a decrease in mast cells inairways, but the mechanism for this is not certain, and a direct inhibitory effect onmediator release has been disproven (132, 133).

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 18: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

142 WEBSTER ET AL.

Resident Tissue Immune Cells

Dendritic cells are involved in antigen presentation, and this could assist in theTh1-Th2 shift. Glucocorticoids reduce the number of dendritic cells in an animalmodel and also in human nasal mucosa after topical glucocorticoid application(134, 135). Epithelial cells are involved in inflammation in asthmatic airways,and they are also a target for glucocorticoid therapy in this disease. As describedearlier, glucocorticoids inhibit the expression of cytokines, chemoattractants, andmediators of inflammation in the epithelial cells of the airways. Glucocorticoidsreduce the inflammatory mediator–induced leakage of plasma proteins into thealveolar bronchi (136). Interestingly, endothelial cells of the airways show thehighest expression of GR in human lung (137). Mucosal cells secrete mucus duringinflammation. Glucocorticoids act directly on these cells and inhibit the expressionof MUC2andMUC5AC, the genes that encode mucin (138, 139). Glucocorticoidsalso inhibit mucus secretion via their inhibition of inflammatory mediators.

Effects of Glucocorticoids on Lymphocyte Selectionat the Cellular and Immune Organ Level

Glucocorticoids play an important role in lymphocyte selection via several mech-anisms, including apoptosis and thymocyte differentiation during development.

APOPTOSIS Apoptosis is a mechanism of programmed cell death, which is neces-sary for a homeostasis to exist between cell death and proliferation. For a review onapoptosis, see King & Cidlowski (140). Glucocorticoids induce G1 arrest and apop-tosis in lymphoid cells. There appear to be two mechanisms for glucocorticoid-mediated apoptosis in thymocytes, one for proliferating thymocytes and one fornonproliferating thymocytes. Although separate mechanisms, these two pathwaysdo share some common features such as the activation of GR and the activation ofcaspases. The mechanism of glucocorticoid-mediated cell cycle arrest and apop-tosis is not fully understood, but some cell cycle genes, e.g.,c-myc, cyclin D3, andCdk4, are regulated by glucocorticoids (140, 141). Glucocorticoids upregulate theexpression of the cyclin-dependent kinase inhibitor, p57Kip2, which is important inthis glucocorticoids-mediated cell cycle arrest (142).

In T cells, glucocorticoid-induced apoptosis is dependent on protein synthesis,but there is also evidence that it is dependent on repression of genes, such asc-myc. Comparison of the glucocorticoid-sensitive T-cell line 6TG1.1 and theglucocorticoid-resistant variant ICR27TK.3, which contains GR that is incapableof gene activation, indicates that repression of AP-1-induced gene activity is notinvolved in glucocorticoid-induced apoptosis in these cells. DNA fragmentation inthe sensitive 6TG1.1 cells was inhibited by cyclohexamide, indicating that proteinsynthesis is required for glucocorticoid-induced apoptosis. Because inhibitory IκBmolecule is induced by dexamethasone in these cells and becausec-mycis regulatedby NFκB, it has been suggested that glucocorticoid-induced apoptosis could occur

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 19: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 143

by repression of gene transcription of thec-mycgene via glucocorticoid-inducedtranscription of IκB or another similar inhibitory factor (64).

Glucocorticoid-mediated apoptosis has also been described in monocytes. Thisoccurs via the death receptor, CD95, a cascade for apoptosis that does not occur inT cells or thymocytes in response to glucocorticoids. In monocytes, glucocorticoidsincrease the expression of membrane CD95 and the ligand, CD95L, and alsoenhance their release. This activates a cascade involving caspase 8 and caspase 3to induce apoptosis (143).

EFFECTS OF GLUCOCORTICOIDS ON THYMOCYTE DIFFERENTIATION AND SELECTION

During the process of thymocyte differentiation, immature CD4− CD8− thymo-cytes rearrange theβ gene locus of the T cell receptor (TCR) to express pre-TCR.These then divide and begin to express CD4+ and CD8+ and rearrange theα geneof TCR to express matureα/β TCR but at low levels. These CD4+ CD8+ lowTCR cells then undergo selection, either positive or negative selection. Preciselywhat factors determine positive versus negative selection is not understood. Invitro in the presence of glucocorticoids alone, thymocytes/hybridomas undergoglucocorticoid-mediated apoptosis, whereas in the absence of glucocorticoids andthe presence of activated TCR, these cells undergo activated-mediated apopto-sis. Culture in the presence of both results in survival, and this balance betweenglucocorticoids and TCR has been termed mutual antagonism. To explain howselection occurs, it has been proposed that where the CD4+ CD8+ low TCR thy-mocytes do not recognize self-antigen/major histocompatability complex (MHC),there is a greater ratio of GR to TCR stimulation, and these cells will die of neglectthrough glucocorticoid-mediated apoptosis. If these thymocytes strongly recog-nize self-MHC, with a resultant higher ratio of TCR to GR activation, then thesecells will die by negative selection through activated-mediated apoptosis. If thereis an intermediate recognition of the MHC, a balance exists between the GR andTCR activation, and these cells will be selected by positive selection and willfurther differentiate into mature CD4+ or CD8+ thymocytes (144, 145). Evidencefor this mutual antagonism has been provided by studies in which inhibiting glu-cocorticoids results in thymocytes with a low-to-moderate avidity for self-MHCbeing forced into activated-mediated apoptosis (negative selection), rather thanbeing rescued and undergoing positive selection as would occur in the presenceof glucocorticoids (146). Furthermore, in these conditions thymocytes that do notrecognize self-MHC are rescued rather than undergoing glucocorticoid-mediatedapoptosis (147).

Thymocyte selection is active during fetal and neonatal period, but the levelsof glucocorticoids circulating in the fetus are low because some level of pro-tection from maternal glucocorticoids is provided by the placenta that containshigh levels of the GC-catabolizing enzyme 11β-hydroxysteroid dehydrogenase.This apparent lack of glucocorticoids in the fetus and the necessity of them forthymocyte differentiation has led to the hypothesis that there may be local produc-tion of glucocorticoids in the thymus during development (145). Indeed, cultured

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 20: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

144 WEBSTER ET AL.

thymic epithelium cells do produce pregnenolone and deoxycorticosterone, andthis production can be increased by ACTH. This production of glucocorticoidswas specific to thymic epithelium cells and was not observed in cultures of thymo-cytes, macrophages, or dendritic cells. The P450 hydroxylase enzymes involvedin the production of glucocorticoids, specifically corticosterone from cholesterol,have been shown in thymic epithelium cells (144, 145, 148). Murine thymus alsocontains all the enzymes required for the production of glucocorticoids fromcholesterol (149).

In hybridomas, activated-mediated apoptosis is caused by the upregulation ofthe ligand for Fas, Fas ligand (FasL). Glucocorticoids prevent this upregulationof FasL, probably by the glucocorticoid-induced leucine zipper (GILZ) gene,but do not interfere with Fas itself. Another mechanism by which glucocorti-coids may inhibit activated-mediated apoptosis is the activation of glucocorticoid-induced TNFR family related (GITR), which can inhibit CD3-mediated but notFas-mediated apoptosis. In some diseases there could be a shift in this balance.Thus, in cases where glucocorticoids are overexpressed, this balance could beshifted such that a higher level of activated TCR cells survive and thus a greaterpopulation of thymocytes that recognize self could survive (145). Indeed, in au-toimmune disease models in animals and in patients with the autoimmune diseasemultiple sclerosis, there is a higher basal level of glucocorticoids (145, 150).

The involvement of glucocorticoids in thymocyte differentiation and selectionis still a matter of some controversy. Generation of transgenic antisense mice forthe 3′ untranslated region of GR under the control of a T cell–specific promoterled to mice with a twofold reduction of GR mRNA and protein. The thymus oftransgenic homozygotes were 90% smaller than controls, and there was a decreasein CD4+ CD8+ thymocytes and a secondary decrease in CD4+ CD8− and CD4−

CD8+ thymocytes, which indicates the necessity of glucocorticoids in thymocytedevelopment and differentiation (144, 145). However, in similar transgenics underthe control of a different promoter (a neurofilament promoter), GR levels weredecreased in all tissues, and there was an associated increase in circulating ACTHand corticosterone. Unlike the T cell–specific antisense animals, no differencein CD4+ CD8+ thymocytes was found (151). In the GR knockin mice, whichare deficient in dimerization and are therefore unable to activate gene transcrip-tion by GR, the thymocyte population was described as being normal althoughglucocorticoid-mediated apoptosis was reduced and no extensive analysis wasperformed (152).

Recently, GR knockout mice have been described that die at birth [day 19 of em-bryogenesis (E19)]. However, during embryogenesis, i.e., up to E18, these animalshave apparently normal thymocyte populations and T cell development, suggestingthat GR in this case in not necessary for the entire process of T cell developmentand differentiation (153, 154). The reason for these differences between animalmodels is not completely clear, but one significant difference between these dif-ferent transgenic animals and knockin animals may result from secondary effectsof the disrupted HPA axis (145).

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 21: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 145

DISRUPTION OF THE HPA AXIS AND DISEASE

Animal Models

Animal models have been useful in understanding the pathogenesis of autoim-mune/inflammatory disease. Many animal models exist in which a blunted HPAaxis response has been associated with susceptibility to autoimmune/inflammatorydiseases. These include the obese strain (OS) chicken as a model for autoimmunethyroiditis (155), certain mouse lupus (SLE) models (156, 157), and the inbredrat strains, LEW/N and F344/N rats, as described earlier. For a review on theseand other animal models for inflammatory diseases, refer to Jafarian-Tehrani &Sternberg (158). It should be noted, however, that many genes and many chromo-some regions, each with small effect, contribute to susceptibility to autoimmunediseases, such as SLE (158a) and inflammatory arthritis (158b). While some ofthese linkage regions contain genes that regulate the HPA axis (158c), many areunrelated or unknown. Such genetic linkage and segregation studies also indi-cate that environmental factors play a large role in variance of disease expression.Thus, many different genetic and environmental factors contribute to autoimmunesusceptibility in addition to HPA or neuroendocrine factors.

Human Diseases

HPA axis responsiveness differs greatly among individuals. Even in healthy in-dividuals considerable intra-individual variability appears in HPA axis responses.Normal healthy volunteers have been subgrouped into high or low responders de-pending on the response of their HPA axis to stimuli (159, 160). Disruptions in theHPA axis or glucocorticoid response could occur at many levels, including at thelevels of the hypothalamus, pituitary, or adrenals, with changes in the expressionof CRH, ACTH or cortisol, or changes in the sensitivity of the system to stimulior suppressive factors. Once cortisol reaches its target tissue, there are then manysteps to inducing gene activation, including entry into the cell, binding to GR,dimerization, translocation to the nucleus, and interaction with cofactors and thebasal transcription machinery. It is conceivable that interruption of any of thesepathways could result in a defective HPA axis or glucocorticoid response leadingeither to a lack of glucocorticoid production or to glucocorticoid resistance andresultant enhanced autoimmune/inflammatory disease. Depending on the specificdefect, patients might or might not respond to exogenous glucocorticoid therapy.For further reading on glucocorticoid resistance syndromes, see the recent reviewby Kino & Chrousos (161).

Disruptions in the HPA Axis or Glucocorticoid ResponseLeading to Glucocorticoid Sensitivity and Resistance

HPA AXIS In patient populations it is often difficult to dissect where a problemlies within the HPA axis, as many of its regulatory components cannot be directly

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 22: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

146 WEBSTER ET AL.

measured in blood. Furthermore, inflammatory illness itself stimulates the HPAaxis and alters its regulation (162, 163). Since CRH cannot be measured directlyin peripheral venous blood, CRH responsiveness must be deduced by measuringlevels of ACTH and cortisol, which can be directly measured in peripheral blood. Ablunted HPA axis resulting in low levels of glucocorticoids or a low glucocorticoidresponse to HPA axis stimulation has been implicated in a number of inflammatorydiseases, including rheumatoid arthritis (164–167), systemic lupus erythematosus(SLE) (168), Sjogren’s syndrome (169), allergic asthma and atopic skin disease(170), and chronic fatigue syndrome (171, 172). Patients with fibromyalgia havea low 24-h urinary free cortisol and a reduced response in the secretion of cortisolto HPA axis challenge (173). In patients with multiple sclerosis and a high plasmabasal cortisol level, a normal HPA axis response to oCRH but a reduced HPA axisresponse to AVP were observed (150).

ADRENAL GLANDS Isolated glucocorticoid deficiency (IGD) is an autosomal re-cessive disorder, characterized by adrenocortical but not mineralocorticoid defi-ciency. Patients generally have low, undetectable cortisol levels that do not increasewith treatment with exogenous ACTH. Also, the endogenous ACTH levels arehigh. Seventeen-point mutation and frameshifts in the receptor for ACTH havebeen identified that lead to this condition (174–176). Thus, this disease resultsbecause defective ACTH receptors render the adrenal gland incapable of sens-ing the high ACTH levels that continue to be secreted by the pituitary. Thus,despite high levels of ACTH, little or no cortisol is produced. Novel mutationsin the ACTH receptor have also been described in a patient with ACTH hyper-sensitivity syndrome. In this case the patient had normal cortisol levels but low,undetectable ACTH levels (177). There have been some reports of patients withIGD developing autoimmune disease, such as organ-specific autoimmunity andautoimmune-mediated hypothroidism (178, 179).

CORTISOL BINDING GLOBULIN (CBG) The level of CBG in the blood limits theamount of free cortisol available. Changes in the expression of this protein orin its binding capacity could thus also affect the availability of cortisol. In NewWorld primates, the lower levels of CBG and the lower affinity of cortisol for CBGhave been associated with the higher circulating levels of glucocorticoids, whichhave been suggested to compensate for target organ resistance present in thesemammals (180). In some patients with long-standing Crohn’s disease, a partialor complete resistance to steroids has been described; this resistance could bedue to the increased expression of CBG in these patients, thereby limiting thebioavailability of glucocorticoids (181).

11β-HYDROXYSTEROID DEHYDROGENASE This enzyme catalyzes the conversionbetween an active glucocorticoid and inactive glucocorticoid (45) as described ear-lier. Thus, changes in this enzyme could result in differences in circulating or tissueglucocorticoid concentrations. In obese men, an impairment in the conversion of

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 23: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 147

the inactive cortisone to the active cortisol was noted, indicating an impairmentin the type I 11β-hydroxysteroid dehydrogenase and resulting in a drop in plasmacortisol levels (182). A decrease in 11β-hydroxysteroid dehydrogenase mRNA inulcerative colitis has also been demonstrated (183).

GLUCOCORTICOID RECEPTOR Glucocorticoid resistance has been associated withmutation of GR. This has been particularly studied in relation to familial gluco-corticoid resistance, a hereditary condition caused by a mutation in the GR withassociated decreased number of receptors, decreased affinity or stability of thereceptor, and a decrease in translocation of the receptor to the nucleus. To date themolecular defects of four families and one sporadic case have been determined.These include three different point mutations in the ligand-binding domain, onein the hinge region, and a deletion in the ligand-binding domain. [For a review seeKino & Chrousos (161)].

A mutation in the GR may not be the only mechanism by which a change in thisgene could affect the sensitivity to glucocorticoids. A polymorphism in codon 363of GR has been described and associated with an increased sensitivity to glucocor-ticoids (184). On the other hand, five polymorphisms in GR (including the one incodon 363) have been described in a normal population, and they cannot be cor-related with glucocorticoid resistance (185). Furthermore, there are patients withglucocorticoid resistance with no mutation detectable in GR, indicating that otherdefects in the steps in the pathway leading to gene activation by glucocorticoidscould result in glucocorticoid resistance (186). The GR has several phosphoryla-tion sites of which the function is unclear. Mutation of these phosphorylation sitesresults in reduced transactivation capability of GR of a minimal promoter and re-duces the stability of the GR protein (187). Such changes as in the phosphorylationstatus of GR could have profound effects on GR function and be one method bywhich glucocorticoid resistance could occur.

GRβ has been proposed to act as a negative regulator of GR function. Thus,variations in the level of GRα and GRβ could also be associated with apparent glu-cocorticoid resistance, both initial and acquired. Glucocorticoid-resistant asthmahas been associated with a higher expression of GRβ on the peripheral bloodlymphocytes (188, 189). GRβ expression is induced by cytokine treatment (190);thus as inflammatory disease progresses and cytokines are produced, induced GRβ

expression could lead to exacerbation or development of glucocorticoid-resistantdisease states (189). Increased expression of GRβ has been shown in mononuclearblood cells from patients with glucocorticoid-resistant ulcerative colitis comparedto similar cells from patients with glucocorticoid-sensitive disease (191). A pa-tient with generalized glucocorticoid resistance and chronic lymphoid leukemiahas been described as having a decreased GR number and a reduced affinity fordexamethasone. This has been shown to be due not to a mutation in the GR but to aredistribution of GRα to GRβ with a reduced expression of GRα and an increasedexpression of GRβ (192). Finally, a polymorphism in exon 9β of the human GRβgene has been found to be associated with rheumatoid arthritis. This polymorphism

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 24: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

148 WEBSTER ET AL.

increases the stability of GRβ and thus alters glucocorticoid sensitivity (193).Taken together, these studies suggest that a variety of pre- and posttranslationalchanges in both forms of the GR are associated with autoimmune/inflammatorydiseases.

COFACTORS Although to date a mutation in a cofactor has not been found asso-ciated with glucocorticoid resistance, a recent study into the glucocorticoid hy-persensitive state associated with HIV-1 infection is worthy of comment. Thishypersensitive state is specific to the immune system, the brain, musculoskeletalsystem, and fat and liver with maintenance of an intact HPA system. A HIV-1 ac-cessory protein, virion-associated protein (Vpr), is able to act as a transcriptionalactivator to aid the replication of the HIV-1 virus. This protein contains a nuclearreceptor binding motif (LXXLL) that binds directly to GR and cooperatively en-hances transcription via SRC1a and p300/CBP cofactors (161, 194). Recently, twosisters have been described with resistance to multiple steroids, and this may bedue to a cofactor defect (195).

TRANSPORT PROTEINS A group of ABC transmembrane transporters, MDR pro-teins, have been suggested to be involved in the active transport of glucocorticoidsout of cells. Evidence exists that these proteins can transport glucocorticoids (196,197; J. I. Webster, J. Carlstedt-Duke, unpublished data). These proteins are nor-mally expressed in the blood-brain barrier, and knockout mice have been generatedfor the mousemdr1a. In these knockout mice, an increase in the amount of dexam-ethasone present in the brain indicates that this protein is involved in the regulationof brain glucocorticoids (198, 199). These proteins have been identified throughtheir involvement in the multidrug resistance phenotype (200). Recently, it wasshown that patients with inflammatory bowel disease whom medical therapy hadfailed had an increase in the expression of the human MDR1 (ABCB1) (201).This raises the possibility that these proteins may be another component that couldaffect the effectiveness of glucocorticoid therapy by reducing the concentrationof the hormone within the cells and thus reducing the ligand availability for theGR. The novel orphan receptor PXR is involved in the upregulation of MDR1(202). Because PXR itself is activated by glucocorticoids among many other lig-ands (203), this could possibly represent another mechanism by which acquiredglucocorticoid resistance may develop.

OTHER NEUROENDOCRINE FACTORSAFFECTING IMMUNE FUNCTION

While this review has been focused on the HPA axis and glucocorticoid regulationof immunity and autoimmune/inflammatory disease, many other neural and neu-roendocrine pathways connect with and regulate the immune system at multiplelevels. These include the sympathetic nervous system and adrenergic factors andthe peripheral nervous system and neuropeptides released by peripheral nerves.

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 25: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 149

Adrenergic Factors

The adrenergic sympathetic nervous system modulates local immune responses[for a comprehensive review of this topic, see Madden et al. (204)]. Noradren-ergic sympathetic nerve fibers run from the CNS to primary and secondary lym-phoid organs, such as the thymus, spleen, and lymph nodes. These nerve terminalsmake synaptic-like connections with neighboring immune cells releasing nora-drenalin (205). The primary neurotransmitter released from sympathetic nerves,noradrenalin, exerts its effect at the target tissues through its receptor, the adren-ergic receptor. Numerous cells of the immune system including lymphocytes andmacrophages express adrenoreceptors. These are G-protein coupled receptors thatcan be divided into two subgroups—theα- andβ-adrenergic receptors. These canbe further subdivided intoα1-,α2-,β1-,β2-, andβ3-adrenergic receptors. The mostimportant receptor in terms of the immune system is theβ2-adrenergic receptor.Expression ofα- andβ-adrenergic receptors on T and B lymphocytes, neutrophils,mononuclear cells, and NK cells has been described. Activation ofβ2-adrenergicreceptors results in an increase in cyclic AMP concentrations, which can modulatecytokine expression, i.e., decreasing TNF-α and increasing IL-8 (94). It was orig-inally thought that noradrenalin activated theβ2-adrenergic receptor resulting insuppressed lymphocyte function. However, more recent data suggest that it mod-ulates B cell function to protect against or aid progression of immune disease. Ifα-adrenergic receptors are present on immune cells, then activation of these recep-tors will lead to the activation of a different signaling cascade and the activation ofMAP kinases. Activation of this cascade has different effects on cellular activityof immune cells than does activation via theβ2 adrenergic receptor (206–208).Furthermore, there appear to be regional difference in the effects of noradrenalinon immune function. Noradrenalin in the thymus is thought to play a role in mod-ulation of thymocyte proliferation and differentiation, whereas in the spleen andlymph nodes it is thought to be involved in enhancement of the primary antibodyresponse (205).

Sex Hormones

The role of sex hormones, particularly estrogen, in the modulation of the im-mune system is an extensive and important area of research that is not reviewedhere [for further reading, see Jansson & Holmdahl (209)]. It is noteworthy thatthere is a greatly increased risk—in the range of two- to tenfold higher female-to-male ratio—of developing many autoimmune/inflammatory diseases such as SLE,rheumatoid arthritis, and multiple sclerosis in women compared to men. Regulationof the immune system by estrogens is of particular importance during pregnancy.In this case a balance between glucocorticoid and estrogen regulation probablyplays a role in suppression of the maternal immune system to prevent rejectionof the fetus (209). Animal models have provided evidence for the importance ofin vivo modulation of the immune system by the estrogen receptor (210, 211).There are two receptors for estrogens. Estrogen receptorα (ERα) was the first tobe described (212). A second independent gene was identified in 1996 and termed

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 26: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

150 WEBSTER ET AL.

ERβ (213). Knockout mice have shown that both estrogen receptors are importantin a gender-specific manner for thymus development and atrophy (214). In addi-tion, tamoxifen, an estrogen receptor antagonist, has some inhibitory effects oninflammation in LEW/N female rats (215). Sex hormone regulation of the immunesystem also varies with aging; for example, expression of IL-6 is under the controlof sex hormones such as estrogen and testosterone. In postmenopausal women andpostandropausal men, the loss of regulation of IL-6 results in increased concen-trations of IL-6 that is associated with the increased occurrence of inflammatorydiseases with old age such as rheumatoid arthritis, bowel disease, and osteoporosis(84).

CONCLUSION

In summary there are multiple neuro-anatomical, hormonal, and molecular mech-anisms by which the CNS regulates immune function and plays a role in suscepti-bility to and pathogenesis of autoimmune/inflammatory disease. We have focusedhere on the HPA axis and glucocorticoids, the final effector end point of the HPAaxis in regulating immunity. It is clear that even in the case of a single hormone,there are many potential mechanisms at gene, protein, receptor, signaling, and cellfunction levels where dysregulation could lead to disease. Given the many differenthormonal and nerve pathways that regulate immunity, each with its own specificmolecular end points, the potential mechanisms for pathogenesis of autoimmunedisease(s) resulting from disruptions in these interactions is large. Nonetheless, athorough understanding at all levels of the means by which the CNS and immunesystems communicate will provide many new insights into the bidirectional reg-ulation of these systems and the disruptions in these communications that lead todisease. Ultimately this understanding will inform new avenues of therapy.

ACKNOWLEDGMENTS

The authors would like to thank Melanie Vacchio for critically reading the manu-script and for useful discussions.

Visit the Annual Reviews home page at www.annualreviews.org

LITERATURE CITED

1. Hench PS, Kendall EC, Slocumb CH,Polley HF. 1950. Effects of cortisone ac-etate and primary ACTH on rheumatoidarthritis, rheumatic fever and certain otherconditions.Arch. Intern. Med.85:545–666

2. Adcock IM. 2000. Molecular mechanismsof glucocorticosteroid actions.Pulmon.Pharmacol. Ther.13:115–26

3. Barnes PJ, Adcock IM. 1998. Transcrip-tion factors and asthma.Eur. Resp. J.12:221–34

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 27: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 151

4. Watkins LR, Maier SF, Goehler LE.1995. Cytokine-to-brain communication:a review and analysis of alternative mech-anisms.Life Sci.57:1011–26

5. Dantzer R, Bluthe RM, Laye S, Bret-Dibat JL, Parnet P, Kelley KW. 1998.Cytokines and sickness behavior.Ann. NYAcad. Sci.840:586–90

6. Benveniste EN. 1998. Cytokine actionsin the central nervous system.CytokineGrowth Factor Rev.9:259–75

7. Hopkins SJ, Rothwell NJ. 1995. Cyto-kines and the nervous system. I. Expres-sion and recognition.Trends Neurosci.18:83–88

8. Brenneman DE, Schultzberg M, BartfaiT, Gozes I. 1992. Cytokine regulation ofneuronal cell survival.J. Neurochem.58:454–60

9. Banks WA, Kastin AJ, Broadwell RD.1995. Passage of cytokines across theblood-brain barrier.Neuroimmunomodu-lation 2:241–48

10. Dantzer R, Konsman JP, Bluthe RM,Kelley KW. 2000. Neural and humoralpathways of communication from the im-mune system to the brain: parallel orconvergent?Autonomic Neurosci.85:60–65

11. Goehler LE, Gaykema RP, Hansen MK,Anderson K, Maier SF, Watkins LR.2000. Vagal immune-to-brain communi-cation: a visceral chemosensory pathway.Autonomic Neurosci.85:49–59

12. Elmquist JK, Scammell TE, Saper CB.1997. Mechanisms of CNS response tosystemic immune challenge: the febrileresponse.Trends Neurosci.20:565–70

13. Gaillard RC. 1998. Cytokines in the neu-roendocrine system.Int. Rev. Immunol.17:181–216

14. Mulla A, Buckingham JC. 1999. Regula-tion of the hypothalamo-pituitary-adrenalaxis by cytokines.Baillieres Best Prac.Res. Clin. Endocrinol. Metab.13:503–21

15. Argo A, Stanisz AM. 1992. Are lympho-cytes a target for substance P modula-

tion in arthritis?Semin. Arthritis Rheum.21:252–58

16. Crofford LJ, Sano H, Karalis K, WebsterEL, Goldmuntz EA, Chrousos GP, Wil-der RL. 1992. Local secretion of corti-cotrophin-releasing hormone in the jointsof Lewis rats with inflammatory arthritis.J. Clin. Invest.90:2555–64

17. Scott LV, Dinan TG. 1998. Vasopressinand the regulation of hypothalamic-pituitary-adrenal axis function: implica-tions for the pathophysiology of depres-sion.Life Sci.62:1985–98

18. Calogero AE, Gallucci WT, Gold PW,Chrousos GP. 1988. Multiple feedbackregulatory loops upon rat hypothalamiccorticotrophin-releasing hormone secre-tion. Potential clinical implications.J.Clin. Invest.82:767–74

19. DeRijk RH, Petrides J, Deuster P, GoldPW, Sternberg EM. 1996. Changes incorticosteroid sensitivity of peripheralblood lymphocytes after strenuous ex-ercise in humans.J. Clin. Endocrinol.Metab.81:228–35

20. DeRijk R, Sternberg EM. 1997. Corticos-terone resistance and disease.Ann. Med.29:79–82

21. Singh A, Zelazowski EB, Petrides JS,Raybourne RB, Sternberg EM, Gold PW,Deuster PA. 1996. Lymphocyte subset re-sponses to exercise and glucocorticoidsuppression in healthy men.Med. Sci.Sports Exercise28:822–28

22. Zelazowski EB, Singh A, Raybourne RB,Sternberg EM, Gold PW, Deuster PA.1997. Lymphocyte subpopulation expres-sion in women: effect of exercise and cir-cadian rhythm.Med. Sci. Sports Exercise29:467–73

23. Deuster PA, Zelazowski EB, Singh A,Sternberg EM. 1999. Expression of lym-phocyte subsets after exercise and dexam-ethasone in high and low stress respon-ders.Med. Sci. Sports Exerc.31:1799–806

24. Wilder RL, Calandra GB, Garvin AJ,Wright KD, Hansen CT. 1982. Strain and

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 28: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

152 WEBSTER ET AL.

sex variation in the susceptibility to strep-tococcal cell wall-induced polyarthritis inthe rat.Arthritis Rheum.25:1064–72

25. Sternberg EM, Hill JM, Chrousos GP,Kamilaris T, Listwak SJ, Gold PW, Wil-der RL. 1989. Inflammatory mediator-induced hypothalamic-pituitary-adrenalaxis activation is defective in streptococ-cal cell wall arthritis-susceptible Lewisrats.Proc. Natl. Acad. Sci. USA86:2374–78

26. Sternberg EM, Young WS III, BernardiniR, Calogero AE, Chrousos GP, GoldPW, Wilder RL. 1989. A central ner-vous system defect in biosynthesis ofcorticotrophin-releasing hormone is asso-ciated with susceptibility to streptococ-cal cell wall-induced arthritis in Lewisrats.Proc. Natl. Acad. Sci. USA86:4771–75

27. Dhabhar FS, McEwen BS, Spencer RL.1993. Stress response, adrenal steroid re-ceptor levels and corticosteroid-bindingglobulin levels—a comparison betweenSprague-Dawley, Fischer 344 and Lewisrats.Brain Res.616:89–98

28. Moncek F, Kvetnansky R, Jezova D.2001. Differential responses to stressstimuli of Lewis and Fischer rats at thepituitary and adrenocortical level.En-docrinol. Regul.35:35–41

29. Smith CC, Omelijaniuk RJ, Whitfield HJJr, Aksentijevich S, Fellows MQ, Zelazo-wski E, Gold PW, Sternberg EM. 1994.Differential mineralocorticoid (type 1)and glucocorticoid (type 2) receptor ex-pression in Lewis and Fischer rats.Neu-roimmunomodulation1:66–73

30. Dhabhar FS, Miller AH, McEwen BS,Spencer RL. 1995. Differential activationof adrenal steroid receptors in neural andimmune tissues of Sprague Dawley, Fis-cher 344, and Lewis rats.J. Neuroim-munol.56:77–90

31. Edwards CK III, Yunger LM, LorenceRM, Dantzer R, Kelley KW. 1991. Thepituitary gland is required for protectionagainst lethal effects ofSalmonella ty-

phimurium. Proc. Natl. Acad. Sci. USA88:2274–77

32. Ruzek MC, Pearce BD, Miller AH, BironCA. 1999. Endogenous glucocorticoidsprotect against cytokine-mediated lethal-ity during viral infection. J. Immunol.162:3527–33

33. Misiewicz B, Poltorak M, RaybourneRB, Gomez M, Listwak S, SternbergEM. 1997. Intracerebroventricular trans-plantation of embryonic neuronal tissuefrom inflammatory resistant into inflam-matory susceptible rats suppresses spe-cific components of inflammation.Exp.Neurol.146:305–14

34. MacPhee IAM, Antoni FA, Mason DW.1989. Spontaneous recovery of rats fromexperimental allergic encephalomyelitisis dependent on regulation of the immunesystem by endogenous adrenal corticos-teroids.J. Exp. Med.169:431–45

35. Rozlog LA, Kiecolt-Glaser JK, MaruchaPT, Sheridan JF, Glaser R. 1999. Stressand immunity: implications for viral dis-ease and wound healing.J. Periodontol.70:786–92

36. Glaser R, Kiecolt-Glaser JK. 1998.Stress-associated immune modulation:relevance to viral infections and chronicfatigue syndrome.Am. J. Med.105:35S–42S

37. Vedhara K, Cox NKM, Wilcock GK,Perks P, Hunt M, Anderson S, Light-man SL. 1999. Chronic stress in elderlycarers of dementia patients and antibodyresponse to influenza vaccination.Lancet353:627–31

38. Friedl KE, Moore RJ, Hoyt RW, Mar-chitelli LJ, Matinez-Lopez LE, AskewEW. 2000. Endocrine markers of semi-starvation in healthy lean men in a mul-tistressor environment.J. Appl. Physiol.88:1820–30

39. de Kloet ER, Vreugdenhil E, Oitzl MS,Joels M. 1998. Brain corticosteroid re-ceptor balance in health and disease.En-docrine Rev.19:269–301

40. Deleted in proof

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 29: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 153

41. Funder JW. 1997. Glucocorticoid andmineralocorticoid receptors: biology andclinical relevance.Annu. Rev. Med.48:231–40

42. Liu W, Wang J, Sauter NK, Pearce D.1995. Steroid receptor heterodimerizationdemonstrated in vitro and in vivo.Proc.Natl. Acad. Sci. USA92:12480–84

43. Trapp T, Holsboer F. 1996. Heterodimer-ization between mineralocorticoid andglucocorticoid receptors increases thefunctional diversity of corticosteroidaction.Trends Pharmacol. Sci.17:145–49

44. de Kloet ER. 2000. Stress in the brain.Eur.J. Pharmacol.405:187–98

45. Seckl JR, Walker BR. 2001. Minireview:11β-hydroxysteroid dehydrogenase type1—a tissue-specific amplifier of glucocor-ticoid action.Endocrinology142:1371–76

46. Nuclear Receptors Nomenclature Com-mittee. 1999. A unified nomenclature sys-tem for the nuclear receptor superfamily.Cell 97:161–63

47. Evans RM. 1988. The steroid and thy-roid hormone receptor superfamily.Sci-ence240:889–95

48. McKay LI, Cidlowski JA. 1999. Molec-ular control of immune/inflammatory re-sponses: interactions between nuclearfactor-κB and steroid receptor-signalingpathways.Endocrine Rev.20:435–59

49. Enc´ıo IJ, Detera-Wadleigh SD. 1991. Thegenomic structure of the human glucocor-ticoid receptor.J. Biol. Chem.266:7182–88

50. Oakley RH, Jewell CM, Yudt MR, Bofe-tiado DM, Cidlowski JA. 1999. The domi-nant negative activity of the human gluco-corticoid receptorβ isoform. Specificityand mechanisms of action.J. Biol. Chem.274:27857–66

51. Oakley RH, Sar M, Cidlowski JA.1996. The human glucocorticoid receptorβ isoform. Expression, biochemical prop-erties and putative function.J. Biol. Chem.271:9550–59

52. Vottero A, Chrousos GP. 1999. Gluco-

corticoid receptorβ: View I. Trends En-docrinol. Metab.10:333–38

53. Hecht K, Carlstedt-Duke J, Stierna P,Gustafsson J-A, Bronnegard M, Wik-strom A-C. 1997. Evidence that theβ-isoform of the human glucocorticoidreceptor does not act as a physiologi-cally significant repressor.J. Biol. Chem.272:26659–64

54. Brogan IJ, Murray IA, Cerillo G, Need-ham M, White A, Davis JRE. 1999. In-teraction of glucocorticoid receptor iso-forms with transcription factors AP-1 andNF-κB: lack of effect of glucocorticoid re-ceptorβ. Mol. Cell. Endocrinol.157:95–104

55. Carlstedt-Duke J. 1999. Glucocorticoidreceptorβ: View II. Trends Endocrinol.Metab.10:339–42

56. Schule R, Rangarajan P, Kliewer S, Ran-sone LJ, Bolado J, Yang N, Verma IM,Evans RM. 1990. Functional antagonismbetween oncoprotein c-Jun and the gluco-corticoid receptor.Cell 62:1217–26

57. Adcock IM, Brown CR, Shirasaki H,Barnes PJ. 1994. Effects of dexametha-sone on cytokine and phorbol ester stim-ulated c-Fos and c-Jun DNA binding andgene expression in human lung.Eur. Resp.J. 7:2117–23

58. Pearce D, Matsui W, Miner JN, Ya-mamoto KR. 1998. Glucocorticoid re-ceptor transcriptional activity determinedby spacing of receptor and nonreceptorDNA sites. J. Biol. Chem.273:30081–85

59. Pearce D, Yamamoto KR. 1993. Min-eralocorticoid and glucocorticoid recep-tor activities distinguished by nonreceptorfactors at a composite response element.Science259:1161–65

60. Malkoski SP, Dorin RI. 1999. Compositeglucocorticoid regulation at a functionallydefined negative glucocorticoid responseelement of the human corticotrophin-releasing hormone gene.Mol. Endocrinol.13:1629–44

61. Baldwin AS Jr. 1996. The NF-κB and IκB

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 30: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

154 WEBSTER ET AL.

proteins: new discoveries and insights.Annu. Rev. Immunol.14:649–81

62. Ghosh S, May MJ, Kopp EB. 1998. NF-κB and Rel proteins: Evolutionarily con-served mediators of immune responses.Annu. Rev. Immunol.16:225–60

63. van der Burg B, Liden J, Okret S, De-launay F, Wissink S, van der Saag PT,Gustafsson J-A. 1997. Nuclear factor-κB repression in antiinflammation andimmunosuppression by glucocorticoids.Trends Endocinol. Metab.8:152–57

64. Ramdas J, Harmon JM. 1998. Glucocor-ticoid-induced apoptosis and regulationof NF-κB activity in human leukemic Tcells.Endocrinology139:3813–21

65. Ray A, Prefontaine KE. 1994. Physi-cal association and functional antagonismbetween the p65 subunit of transcriptionfactor NF-κB and the glucocorticoid re-ceptor. Proc. Natl. Acad. Sci. USA91:752–56

66. Caldenhoven E, Liden J, Wissink S, Vande Stolpe A, Raaijmakers J, Koender-man L, Okret S, Gustafsson J-A, Vander Saag PT. 1995. Negative cross-talkbetween RelA and the glucocorticoid re-ceptor: a possible mechanism for the an-tiinflammatory action of glucocorticoids.Mol. Endocrinol.9:401–12

67. Scheinman RI, Gualberto A, Jewell CM,Cidlowski JA, Baldwin AS Jr. 1995.Characterization of mechanisms involvedin transrepression of NF-κB by activatedglucocorticoid receptors.Mol. Cell. Biol.15:943–53

68. Kleinert H, Euchenhofer C, Ihrig-BiedertI, Forstermann U. 1996. Glucocortico-ids inhibit the induction of nitric oxidesynthase II by down-regulating cytokine-induced activity of transcription factor nu-clear factor-κB. Mol. Pharmacol.49:15–21

69. De Bosscher K, Schmitz ML, VandenBerghe W, Plaisance S, Fiers W, Hae-geman G. 1997. Glucocorticoid-mediatedrepression of nuclear factor-κB-dep-endent transcription involved direct inter-

ference with transactivation.Proc. Natl.Acad. Sci. USA94:13504–9

70. Ray KP, Farrow S, Daly M, TalabotF, Searle N. 1997. Induction of the E-selectin promoter by interleukin-1 and tu-mour necrosis factorα, and inhibition byglucocorticoids.Biochem. J.328:707–15

71. Liden J, Rafter I, Truss M, Gustafsson J-A, Okret S. 2000. Glucocorticoid effectson NF-κB binding in the transcription ofthe ICAM-1 gene.Biochem. Biophys. Res.Commun.273:1008–14

72. Wissink S, van Heerde EC, van der BurgB, van der Saag PT. 1998. A dual mecha-nism mediates repression of NF-κB activ-ity by glucocorticoids.Mol. Endocrinol.12:355–63

73. Liden J, Delaunay F, Rafter I, GustafssonJ-A, Okret S. 1997. A new function forthe C-terminal zinc finger of the glucocor-ticoid receptor. Repression of RelA trans-activation. J. Biol. Chem.272:21467–72

74. Wissink S, van de Stolpe A, CaldenhovenE, Koenderman L, van der Saag PT.1997. NF-kB/Rel family members regu-lating the ICAM-1 promoter in monocyticTHP-1 cells. Immunobiology 198:50–64

75. Sheppard K-A, Phelps KM, WilliamsAJ, Thanos D, Glass K, Rosenfeld MG,Gerritsen ME, Collins T. 1998. Nuc-lear integration of glucocorticoid recep-tor and nuclear factor-κB signaling byCREB-binding protein and steroid re-ceptor coactivator-1.J. Biol. Chem.273:29291–94

76. McKay LI, Cidlowski JA. 2000. CBP(CREB binding protein) integrates NF-κB(nuclear factor-κB) and glucocorticoid re-ceptor physical interaction and antago-nism.Mol. Endocrinol.14:1222–34

77. Doucas V, Shi Y, Miyamoto S, West A,Verma I, Evans RM. 2000. Cytoplasmiccatalytic subunit of protein kinase A me-diates cross-repression by NF-κB and theglucocorticoid receptor.Proc. Natl. Acad.Sci. USA97:11893–98

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 31: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 155

78. Barnes PJ. 1998. Anti-inflammatory ac-tions of glucocorticoids: molecular mech-anisms.Clin. Sci.94:557–72

79. Lee SW, Tsou A-P, Chan H, Thomas J,Petrie K, Eugui EM, Allison AC. 1988.Glucocorticoids selectively inhibit thetranscription of the interleukin 1β geneand decrease the stability of interleukin1βmRNA.Proc. Natl. Acad. Sci. USA85:1204–8

80. Groujon E, Lay´e S, Parnet P, DantzerR. 1997. Regulation of cytokine gene ex-pression in the central nervous systemby glucocorticoids: mechanisms and fun-ctional consequences.Psychoneuroen-docrinology22:S75–S80

81. Schmidt M, Pauels H-G, L¨ugering N,Lugering A, Domschke W, KucharzikT. 1999. Glucocorticoids induce apopto-sis in human monocytes: Potential role ofIL-1β. J. Immunol.163:3484–90

82. Kunicka JE, Talle MA, Denhardt GH,Brown M, Prince LA, Goldstein G.1993. Immunosuppression by glucocorti-coids: Inhibition of production of multiplelymphokines by in vivo administration ofdexamethasone.Cell. Immunol.149:39–49

83. Colotta F, Re F, Muzio M, Bertini R,Polentarutti N, Sironi M, Giri JG, DowerSK, Sims JE, Mantovani A. 1993. Inter-leukin-1 type II receptor: A decoy targetfor IL-1 that is regulated by IL-4.Science261:472–75

84. Vanden Berghe W, Vermeulen L, DeWilde G, De Bosscher K, Boone E, Hae-geman G. 2000. Signal transductionby tumor necrosis factor and gene regu-lation of the inflammatory cytokineinterleukin-6. Biochem. Pharmacol.60:1185–95

85. Wang J, Zhu Z, Nolfo R, Elias JA. 1999.Dexamethasone regulation of lung epithe-lial cell and fibroblast interleukin-11 pro-duction.Am. J. Physiol. Lung Cell. Mol.Physiol.276:L175–L85

86. Steer JH, Ma DTS, Dusci L, GarasG, Pederson KE, Joyce DA. 1998. Al-

tered leucocyte trafficking and suppressedtumour necrosis factor -α release fromperipheral blood monocytes after intra-articular glucocorticoid treatment.Ann.Rheu. Dis.57:732–37

87. Steer JH, Kroeger KM, Abraham LJ,Joyce DA. 2000. Glucocorticoids sup-press tumor necrosis factor-α expressionby human monocytic THP-1 cells by sup-pressing transactivation through adjacentNF-κB and c-Jun-activating transcriptionfactor-2 binding sites in the promoter.J.Biol. Chem.275:18432–40

88. Joyce DA, Steer JH, Abraham LJ. 1997.Glucocorticoid modulation of humanmonocyte/macrophage function: Controlof TNF-α secretion.Inflamm. Res.46:447–51

89. Moynihan JA, Callahan TA, Kelley SP,Campbell LM. 1998. Adrenal hormonemodulation of type 1 and type 2 cy-tokine production by spleen cells: Dex-amethasone and dehydroepiandrosteronesuppress interleukin-2, interleukin-4, andinterferon-γ production in vitro.Cell. Im-munol.184:58–64

90. Verhoef CM, van Roon JAG, Vianen ME,Lafeber FPJG, Bijlsma JWJ. 1999. Theimmune suppressive effect of dexametha-sone in rheumatoid arthritis is accompa-nied by upregulation of interleukin 10 andby differential changes in interferonγ andinterleukin 4 production.Ann. Rheu. Dis.58:49–54

91. Chang MM-J, Juarez M, Hyde DM, WuR. 2001. Mechanism of dexamethasone-mediated interleukin-8 gene suppressionin cultured airway epithelial cells.Am.J. Physiol. Lung Cell. Mol. Physiol.280:L107–L15

92. Wu C-Y, Wang K, McDyer JF, SederRA. 1998. Prostaglandin E2 and dexam-ethasone inhibit IL-12 receptor expres-sion and IL-12 responsiveness.J. Im-munol.161:2723–30

93. Franchimont D, Galon J, Gadina M, Vis-conti R, Zhou Y-J, Aringer M, FruchtDM, Chrousos GP, O’Shea JJ. 2000.

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 32: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

156 WEBSTER ET AL.

Inhibition of Th1 immune response byglucocorticoids: dexamethasone selec-tively inhibits IL-12-induced Stat4 phos-phorylation in T lymphocytes.J. Im-munol.164:1768–74

94. Elenkov IJ, Chrousos GP. 1999. Stresshormones, Th1/Th2 patterns, pro/anti-inflammatory cytokines and susceptibil-ity to disease.Trends Endocrinol. Metab.10:359–68

95. Bianchi M, Meng C, Ivashkiv LB. 2000.Inhibition of IL-2-induced Jak-STAT sig-naling by glucocorticoids.Proc. Natl.Acad. Sci. USA97:9573–78

96. Adkins KK, Levan TD, Miesfeld RL,Bloom JW. 1998. Glucocorticoid regula-tion of GM-CSF: evidence for transcrip-tional mechanisms in airway epithelialcells. Am. J. Physiol. Lung Cell. Mol.Physiol.275:L372–L78

97. Hodge S, Hodge G, Flower R, Han P.1999. Methyl-prednisolone up-regulatesmonocyte interleukin-10 production instimulated whole blood.Scand. J. Im-munol.49:548–53

97a. Visser J, van Boxel-Dezaire A, MethorstD, Brunt T, de Kloet ER, Nagelker-ken L. 1998. Differential regulation ofinterleukin-10 (IL-10) and IL-12 by glu-cocorticoids in vitro.Blood91:4255–64

98. Yoshikawa H, Nakajima Y, Tasaka K.1999. Glucocorticoid suppresses au-tocrine survival of mast cells by inhibitingIL-4 production and ICAM-1 expression.J. Immunol.162:6162–70

99. Sewell WA, Scurr LL, Orphanides H,Kinder S, Ludowyke RI. 1998. Inductionof interleukin-4 and interleukin-5 expres-sion in mast cells is inhibited by glucocor-ticoids.Clin. Diagn. Lab. Immunol.5:18–23

100. Daynes RA, Araneo BA. 1989. Contrast-ing effects of glucocorticoids on the ca-pacity of T cells to produce the growthfactors interleukin 2 and interleukin 4.Eur. J. Immunol.19:2319–25

101. Daynes RA, Araneo BA, Dowell TA,Huang K, Dudley D. 1990. Regula-

tion of murine lymphokine productionin vivo. III. The lymphoid tissue mi-croenvironment exerts regulatory influ-ences over T helper cell function.J. Exp.Med.171:979–96

102. Cronstein BN, Kimmel SC, Levin RI,Martiniuk F, Weissmann G. 1992. Amechanism for the antiinflammatory ef-fects of corticosteroids: the glucocorti-coid receptor regulates leukocyte adhe-sion to endothelial cells and expression ofendothelial-leukocyte adhesion molecule1 and intercellular adhesion molecule 1.Proc. Natl. Acad. Sci. USA89:9991–95

103. Atsuta J, Plitt J, Bochner BS, SchleimerRP. 1999. Inhibition of VCAM-1 expres-sion in human bronchial epithelial cellsby glucocorticoids.Am. J. Resp. Cell Mol.Biol. 20:643–50

104. Nakagawa M, Bondy GP, Waisman D,Minshall D, Hogg JC, van Eeden SF.1999. The effect of glucocorticoids onthe expression of L-selectin on polymor-phonuclear leukocyte.Blood93:2730–37

105. Ohtsuka T, Kubota A, Hirano T, Watan-abe K, Yoshida H, Tsurufuji M, IizukaY, Konishi K, Tsurufuji S. 1996. Glu-cocorticoid-mediated gene suppression ofrat cytokine-induced neutrophil chemoat-tractant CINC/gro, a member of theinterleukin-8 family, through impairmentof NF-κB activation. J. Biol. Chem.271:1651–59

106. Richards DF, Fernandez M, Caulfield J,Hawrylowicz CM. 2000. Glucocorticoidsdrive human CD8+ T cell differentiationtowards a phenotype with high IL-10 andreduced IL-4, IL-5 and IL-13 production.Eur. J. Immunol.30:2344–54

107. Mori A, Kaminuma O, Suko M, InoueS, Ohmura T, Hoshino A, Asakura Y,Miyazawa K, Yokota T, Okumura Y, ItoK, Okudaira H. 1997. Two distinctpathways of interleukin-5 synthesis inallergen-specific human T-cell clones aresuppressed by glucocorticoids.Blood89:2891–2900

108. Pype JL, Dupont LJ, Menten P, Van

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 33: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 157

Coillie E, Opdenakker G, Van Damme J,Chung KF, Demedts MG, Verleden GM.1999. Expression of monocyte chemotac-tic protein (MCP)-1, MCP-2, and MCP-3 by human airway smooth-muscle cells.Am. J. Resp. Cell Mol. Biol.21:528–36

109. Wingett D, Forcier K, Nielson CP.1996. Glucocorticoid-mediated inhibitionof RANTES expression in human T lym-phocytes.FEBS Lett.398:308–11

110. Poon M, Liu B, Taubman MB. 1999.Identification of a novel dexamethasone-sensitive RNA-destabilizing region onrat monocyte chemoattractant protein 1mRNA. Mol. Cell. Biol.19:6471–78

111. Lilly CM, Nakamura H, Kesselman H,Nagler-Anderson C, Asano K, Garcia-Zepeda EA, Rothenberg ME, Drazen JM,Luster AD. 1997. Expression of eotaxinby human lung epithelial cells. Inductionby cytokines and inhibition by glucocor-ticoids.J. Clin. Invest.99:1767–73

112. Yamamoto K, Arakawa T, Ueda N, Ya-mamoto S. 1995. Transcriptional rolesof nuclear factorκB and nuclear factor-interleukin-6 in the tumor necrosis factorα-dependent induction of cylooxygenase-2 in MC3T3-E1 cells.J. Biol. Chem.270:31315–20

113. Perkins DJ, Kniss DA. 1997. Tumornecrosis factor-α promotes sustainedcyclooxygenase-2 expression: attenua-tion by dexamethasone and NSAIDs.Prostaglandins54:727–43

114. Newton R, Kuitert LM, Slater DM, Ad-cock IM, Barnes PJ. 1997. Cytokine in-duction of cytosolic phospholipase A2 andcyclooxygenase-2 mRNA is suppressedby glucocorticoids in human epithelialcells.Life Sci.60:67–78

115. Dolan-O’Keefe M, Nick HS. 1999. In-hibition of cytoplasmic phospholipase A2

expression by glucocorticoid in rat in-testinal epithelial cells.Gastroenterology116:855–64

116. de Vera ME, Taylor BS, Wang Q, ShapiroRA, Billiar TR, Geller DA. 1997. Dexam-ethasone suppresses iNOS gene expres-

sion by upregulating I-κBα and inhibit-ing NF-κB. Am. J. Physiol.273:G1290–G96

117. Buckingham JC, Flower RJ. 1997. Lipo-cortin 1: a second messenger of glu-cocorticoid action in the hypothalamo-pituitary-adrenocortical axis.Mol. Med.Today3:296–302

118. Collins S, Caron MG, Lefkowitz RJ.1988. Beta-adrenergic receptors in ham-ster smooth muscle cells are transcription-ally regulated by glucocorticoids.J. Biol.Chem.263:9067–70

119. Mak JC, Nishikawa M, Barnes PJ.1995. Glucocorticoids increase beta 2-adrenergic receptor transcription in hu-man lung.Am. J. Physiol.268:L41–L46

120. Ihara H, Nakanishi S. 1990. Selective in-hibition of expression of the substanceP receptor mRNA in pancreatic acinarAR42J cells by glucocorticoids.J. Biol.Chem.265:22441–45

121. Katsunuma T, Mak JC, Barnes PJ.1998. Glucocorticoids reduce tachykininNK2 receptor expression in bovine tra-cheal smooth muscle.Eur. J. Pharmacol.344:99–106

122. Russo-Marie F. 1992. Macrophages andthe glucocorticoids.J. Neuroimmunol.40:281–86

123. O’Connor TM, O’Halloran DJ, Shana-han F. 2000. The stress response andthe hypothalamic-pituitary-adrenal axis:from molecule to melancholia.Q. J. Med.93:323–33

124. Goulding NJ, Euzger HS, Butt SK, Per-retti M. 1998. Novel pathways for gluco-corticoid effects on neutrophils in chronicinflammation. Inflamm. Res.47:S158–S65

125. Nelms K, Keegan AD, Zamorano J, RyanJJ, Paul WE. 1999. The IL-4 receptor:signaling mechanisms and biologic func-tions.Annu. Rev. Immunol.17:701–38

126. Franchimont D, Louis E, Dewe W,Martens H, Vrindts-Gevaert Y, de GrooteD, Belaiche J, Greenen V. 1998. Ef-fects of dexamethasone on the profile of

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 34: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

158 WEBSTER ET AL.

cytokine secretion in human whole bloodcell cultures.Regl. Pept.73:59–65

127. Agarwal SK, Marshall GD Jr. 2001. Dex-amethasone promotes type 2 cytokine pro-duction primarily through inhibition oftype 1 cytokines.J. Interferon CytokineRes.21:147–55

128. Tamada K, Harada M, Abe K, Li T,Nomoto K. 1998. IL-4-producing NK1.1+

T cells are resistant to glucocorticoid-induced apoptosis: Implications for theTh1/Th2 balance.J. Immunol.161:1239–47

129. Elenkov IJ, Hoffman J, Wilder RL. 1997.Does differential neuroendocrine controlof cytokine production govern the expres-sion of autoimmune diseases in pregnancyand the postpartum period?Mol. Med. To-day3:379–83

130. Schleimer RP, Bochner BS. 1994. Theeffects of glucocorticoids on humaneosinophils.J. Allergy Clin. Immunol.94:1202–13

131. Hirai K, Miyamasu M, Takaishi T, Mor-ita Y. 1997. Regulation of the function ofeosinophils and basophils.Crit. Rev. Im-munol.17:325–52

132. Cohan VL, Undem BJ, Fox CC, Adkin-son NFJ, Lichtenstein LM, SchleimerRP. 1989. Dexamethasone does not in-hibit the release of mediators from humanmast cells residing in airway, intestine,or skin. Am. Rev. Respir. Dis.140:951–54

133. Laitinen LA, Laitinen ATH. 1992. A com-parative study of the effects of an in-haled corticosteroid, budesonide, and abeta 2-agonist, terbutaline, on airway in-flammation in newly diagnosed asthma: arandomized, double-blind, parallel-groupcontrolled trial.J. Allergy Clin. Immunol.90:32–42

134. Nelson DJ, McWilliam AS, Haining S,Holt PG. 1995. Modulation of airway in-traepithelial dendritic cells following ex-posure to steroids.Am. J. Resp. Crit. CareMed.151:475–81

135. Holm AF, Fokkens WJ, Godthelp T, Mul-

der PG, Vroom TM, Rijntes E. 1995. Ef-fect of 3 months’ nasal steroid therapy onnasal T cells and Langerhans cells in pa-tients suffering from allergic rhinitis.Al-lergy50:402–9

136. Boschetto P, Rogers DF, Fabbri LM,Barnes PJ. 1991. Corticosteroid inhibitionof airway microvascular leakage.Am. Rev.Respir. Dis.143:605–9

137. Adcock IM, Gilbey T, Gelder CM,Chung KF, Barnes PJ. 1996. Glucocor-ticoid receptor localization in normal andasthmatic lung.Am. J. Resp. Crit. CareMed.154:771–82

138. Kai H, Yoshitake K, Hisatsune A, KidoT, Isohama Y, Takahama K, Miyata T.1996. Dexamethasone suppresses mucusproduction and MUC-2 and MUC-5ACgene expression by NCI-H292 cells.Am.J. Physiol.271: L484–L488

139. Shimura S, Sasaki T, Ikeda K, Yam-aguchi K, Sasaki H, Takishima T. 1990.Direct inhibitory action of glucocorticoidon glycoconjugates secretion from airwaysubmucosal glands.Am. Rev. Respir. Dis.141:1044–49

140. King KL, Cidlowski JA. 1998. Cell cy-cle regulation and apoptosis.Annu. Rev.Physiol.60:601–17

141. Planey SL, Litwack G. 2000. Glucocor-ticoid-induced apoptosis in lympho-cytes.Biochem. Biophys. Res. Commun.279:307–12

142. Samuelsson MKR, Pazirandeh A, Da-vani B, Okret S. 1999. P57kip2, a glucocor-ticoid-induced inhibitor of cell cycle pro-gression in HeLa cells.Mol. Endocrinol.13:1811–22

143. Schmidt M, L¨ugering N, Lugering A,Pauels H-G, Schulze-Osthoff K, Doms-chke W, Kucharzik T. 2001. Role ofthe CD95/CD95 ligand system in glu-cocorticoid-induced monocyte apoptosis.J. Immunol.166:1344–51

144. Ashwell JD, Lu FWM, Vacchio MS.2000. Glucocorticoids in T cell develop-ment and function.Annu. Rev. Immunol.18:309–45

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 35: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 159

145. Pazirandeh A, Xue Y, Rafter I, Sj¨o-vall J, Jondal M, Okret S. 1999. Para-crine glucocorticoid activity producedby mouse thymic epithelial cells.FASEBJ. 13:893–901

146. Vacchio MS, Ashwell JD, King LB.1998. A positive role for thymus-derivedsteroids in formation of the T-cell reper-toire.Ann. NY Acad. Sci.840:317–27

147. Lechner O, Wiegers GJ, Oliveira-dos-Santos AJ, Dietrich H, Recheis H, Wa-terman M, Boyd R, Wick G. 2000.Glucocorticoid production in the murinethymus.Eur. J. Immunol.30:337–46

148. Vacchio MS, Ashwell JD. 1997. Thy-mus-derived glucocorticoids regulateantigen-specific positive selection.J.Exp. Med.185:2033–38

149. Vacchio MS, Lee JJY, Ashwell JD.1999. Thymus-derived glucocorticoidsset the thresholds for thymocyte selec-tion by inhibiting TCR-mediated thymo-cyte activation.J. Immunol.163:1327–33

150. Michelson D, Stone L, Galliven E, Ma-giakou MA, Chrousos GP, SternbergEM, Gold PW. 1994. Multiple scle-rosis is associated with alterationsin hypothalamic-pituitary-adrenal axisfunction. J. Clin. Endocrinol. Metab.79:848–53

151. Morale MC, Batticane N, Gallo F, Bar-den N, Marchetti B. 1995. Disruption ofhypothalamic-pituitary-adrenocorticalsystem in transgenic mice expressingtype II glucocorticoid receptor antisenseribonucleic acid permanently impairsT cell function: Effects on T cell traf-ficking and T cell responsiveness duringpostnatal development.Endocrinology136:3949–60

152. Reichardt HM, Kaestner KH, Tucker-mann J, Kretz O, Wessely O, Bock R,Gass P, Schmid W, Herrlich P, AngelP, Schutz G. 1998. DNA binding of theglucocorticoid receptor is not essentialfor survival.Cell 93:531–41

153. Godfrey DI, Purton JF, Boyd RL,

Cole TL. 2000. Stress-free T-cell devel-opment: Glucocorticoids are not obliga-tory. Immunol. Today21:606–11

154. Purton JF, Boyd RL, Cole TJ, God-frey DI. 2000. Intrathymic T cell devel-opment and selection proceeds normallyin the absence of glucocorticoid receptorsignaling.Immunity13:179–86

155. Wick G, Sgonc R, Lechner O. 1998.Neuroendocrine-immune disturbancesin animal models with spontaneous au-toimmune diseases.Ann. NY Acad. Sci.840:591–98

156. Hu Y, Dietrich H, Herold M, HeinrichPC, Wick G. 1993. Disturbed immuno-endocrine communication via the hypo-thalamo-pituitary-adrenal axis in au-toimmune disease.Int. Arch. AllergyImmunol.102:232–41

157. Lechner O, Hu Y, Jafarian-Tehrani M,Dietrich H, Schwartz S, Herold M,Haour F, Wick G. 1996. Disturbedimmunoendocrine communication viathe hypothalamo-pituitary-adrenal axisin murine lupus.Brain Behav. Immun.10:337–50

158. Jafarian-Tehrani M, Sternberg EM.1999. Animal models of neuroimmuneinteraction in inflammatory diseases.J.Neuroimmunol.100:13–20

158a. Wakeland EK, Wandstrat AE, Liu K,Morel L. 1999. Genetic dissection of sys-temic lupus erythematosus.Curr. Opin.Immunol.11:701–7

158b. Remmers EF, Longman RE, Du Y,O’Hare A, Cannon GW, Griffiths MM,Wilder RL. 1996. A genome scan lo-calizes five non-MHC loci controllingcollagen-induced arthritis in rates.Nat.Genet.14:82–85

158c. Listwak S, Barrientos RM, Koike G,Ghosh S, Gomez M, Misiewica B,Sternberg EM. 1999. Identification of anovel inflammation-protective locus inthe Fischer rat.Mamm. Genome10:362–65

159. Petrides JS, Gold PW, Mueller GP,Singh A, Stratakis C, Chrousos GP,

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 36: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

160 WEBSTER ET AL.

Deuster PA. 1997. Marked differencesin functioning of the hypothalamic-pituitary-adrenal axis between groups ofmen.J. Appl. Physiol.82:1979–88

160. Huizenga NATM, Koper JW, de LangeP, Pols HAP, Stolk RP, Grobbee DE,de Jong FH, Lamberts SWJ. 1998. In-terperson variability but intraperson sta-bility of baseline plasma cortisol con-centrations, and its relation to feedbacksensitivity of the hypothalamo-pituitary-adrenal axis to a low dose of dexametha-sone in elderly individuals.J. Clin. En-docrinol. Metab.83:47–54

161. Kino T, Chrousos GP. 2001. Gluco-corticoid and mineralocorticoid resis-tance/hypersensitivity syndromes.J. En-docrinol.169:437–45

162. Schmidt ED, Janszen AW, WouterloodFG, Tilders FJ. 1995. Interleukin-1-in-duced long-lasting changes in hypotha-lamic corticotrophin-releasing hormone(CRH)–neurons and hyperresponsive-ness of the hypothalamus-pituitary-adrenal axis.J. Neruosci.15:7417–26

163. Chowdry HS, Larsen PJ, Harbuz MS,Jessop DS, Aguilera G, Eckland DJ,Lightman SL. 1995. Evidence for argi-nine vasopressin as the primary activatorof the HPA axis during adjuvant-inducedarthritis.Br. J. Pharmacol.116:2417–24

164. Cash JM, Crofford LJ, Gallucci WT,Sternberg EM, Gold PW, ChrousosGP, Wilder RL. 1992. Pituitary-adrenalaxis responsiveness to ovine corti-cotrophin releasing hormone in patientswith rheumatoid arthritis treated withlow dose prednisolone.J. Rheumatol.19:1692–96

165. Chikanza IC, Petrou P, Kingsley G,Chrousos GP, Panayi GS. 1992. De-fective hypothalamic response to im-mune and inflammatory stimuli in pa-tients with rheumatoid arthritis.ArthritisRheum.35:1281–88

166. Cutolo M, Foppiani L, Perete C,Ballarino P, Sulli A, Villaggio B, Se-riolo B, Giusti M, Accardo S. 1999.

Hypothalamic-pituitary-adrenocorticalaxis function in premenopausal womenwith rheumatoid arthritis not treatedwith glucocorticoids.J. Rheumatol.26:282–88

167. Gutierrez MA, Garcia ME, RodriguezJA, Mardonez G, Jacobelli S, RiveroS. 1999. Hypothalamic-pituitary-adre-nal axis in patients with active rheuma-toid arthritis: a controlled study us-ing insulin hypoglycemia stress testand prolactin stimulation.J. Rheumatol.26:277–81

168. Gutierrez MA, Garcia ME, RodriguezJA, Rivero S, Jacobelli S. 1998. Hypo-thalamic-pituitary-adrenal axis functionand prolactin secretion in systemic lupuserythematosus.Lupus7:404–8

169. Johnson EO, Vlachoyiannopoulos PG,Skopouli FN, Tzioufas AG, Mout-sopoulos HM. 1998. Hypofunction ofthe stress axis in Sjogren’s syndrome.J.Rheumatol.25:1508–14

170. Buske-Kirschbaum A, Jobst S, Psych D,Wustman A, Kirschbaum C, Rauh W,Hellhammer D. 1997. Attenuated freecortisol response to psychosocial stressin children with atopic dermatitis.Psy-chosom. Med.59:419–26

171. Neeck G, Crofford LJ. 2000. Neuroen-docrine perturbations in fibromyalgiaand chronic fatigue syndrome.Rheum.Dis. Clin. North Am.26:989–1002

172. Demitrack MA, Dale JK, Straus SE,Laue L, Listwak SJ, Kruesi MJ, Chro-usos GP, Gold PW. 1991. Evidence forimpaired activation of the hypothalamic-pituitary-adrenal axis in patients withchronic fatigue syndrome.J. Clin. En-docrinol. Metab.73:1224–34

173. Crofford LJ, Pillemer SR, KalogerasKT, Cash JM, Michelson D, Kling MA,Sternberg EM, Gold PW, ChrousosGP, Wilder RL. 1994. Hypothalamic-pituitary-adrenal axis perturbations inpatients with fibromyalgia.ArthritisRheum.37:1583–92

174. Tsigos C. 1999. Isolated glucocorticoid

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 37: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 161

deficiency and ACTH receptor mutation.Arch. Med. Res.30:475–80

175. Tsigos C, Tsiotra P, Garibaldi LR,Stravridis JC, Chrousos GP, Raptis SA.2000. Mutation of the ACTH receptorgene in a new family with isolated gluco-corticoid deficiency.Mol. Genet. Metab.71:646–50

176. Wu S-M, Stratakis CA, Chan CHY,Hallermeier KM, Bourdony CJ, RennertOM, Chan W-Y. 1998. Genetic hetero-geneity of adrenocorticotropin (ACTH)resistance syndromes: identification of anovel mutation of the ACTH receptorgene in hereditary glucocorticoid defi-ciency.Mol. Genet. Metab.64:256–65

177. Hiroi N, Yakushiji F, Shimojo M,Watanabe S, Sugano S, Yamaguchi N,Miyachi Y. 1998. Human ACTH hyper-sensitivity syndrome associated with ab-normalities of the ACTH receptor gene.Clin. Endorinol.48:129–34

178. Burke CW. 1985. Adrenocortical in-sufficiency. Clin. Endocrinol. Metab.14:947–76

179. Kageyama Y. 2000. A case of iso-lated ACTH deficiency who developedautoimmune-mediated hypothyroidismand impaired water diuresis during glu-cocorticoid replacement therapy.En-docr. J.47:667–74

180. Pugeat MM, Chrousos GP, NisulaBC, Loriaux DL, Brandon D, LipsettMB. 1984. Plasma cortisol transportand primate evolution.Endocrinology115:357–61

181. Mingrone G, DeGaetano A, Pugeat M,Capristo E, Greco AV, Gasbarrini G.1999. The steroid resistance of Crohn’sdisease.J. Invest. Med.47:319–25

182. Rask E, Olsson T, Soderberg S, AndrewR, Livingstone DE, Johnson O, WalkerBR. 2001. Tissue-specific dysregulationof cortisol metabolism in human obesity.J. Clin. Endocrinol. Metab.86:1418–21

183. Takahashi KI, Fukushima K, SasanoH, Sasaki I, Matsuno S, KrozowskiZS, Nagura H. 1999. Type II 11beta-

hydroxysteroid dehydrogenase expres-sion in human colonic epithelial cells ofinflammatory bowel disease.Dig. Dis.Sci.44:2516–22

184. Huizenga NATM, Koper JW, de LangeP, Pols HAP, Stolk RP, Burger H,Grobbee DE, Brinkmann AO, de JongFH, Lamberts SWJ. 1998. A poly-morphism in the glucocorticoid recep-tor gene may be associated with anincreased sensitivity to glucocorticoidsin vivo. J. Clin. Endocrinol. Metab.83:144–51

185. Koper JW, Stolk RP, de Lange P,Huizenga NATM, Molijn G-J, PolsHAP, Grobbee DE, Karl M, de JongFH, Brinkmann AO, Lamberts SWJ.1997. Lack of association between fivepolymorphisms in the human glucocor-ticoid receptor gene and glucocorticoidresistance.Hum. Genet.99:663–68

186. Huizenga NATM, de Lange P, KoperJW, de Herder WW, Abs R, KasterenJHLMV, de Jong FH, Lamberts SWJ.2000. Five patients with biochemicaland/or clinical generalized glucocorti-coid resistance without alterations in theglucocorticoid receptor gene.J. Clin. En-docrinol. Metab.85:2076–81

187. Webster JC, Jewell CM, Bodwells JE,Munck A, Sar M, Cidlowski JA. 1997.Mouse glucocorticoid receptor phospho-rylation status influences multiple func-tions of the receptor protein.J. Biol.Chem.272:9287–93

188. Hamid QA, Wenzel SE, Hauk PJ, Tsi-copoulos A, Wallaert B, Lafitte J-J,Chrousos GP, Szefler SJ, Leung DYM.1999. Increased glucocorticoid recep-tor β in airway cells of glucocorticoid-insensitive asthma.Am. J. Resp. Crit.Care Med.159:1600–4

189. Leung DYM, Hamid Q, Vottero A, Sze-fler SJ, Surs W, Minshall E, ChrousosGP, Klemm DJ. 1997. Associationof glucocorticoid insensitivity with in-creased expression of glucocorticoid re-ceptorβ. J. Exp. Med.186:1567–74

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 38: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

162 WEBSTER ET AL.

190. Webster JC, Oakley RH, Jewell CM,Cidlowski JA. 2001. Proinflammatorycytokines regulate human glucocorticoidreceptor gene expression and lead to theaccumulation of the dominant negativeβ isoform: a mechanism for the gener-ation of glucocorticoid resistance.Proc.Natl. Acad. Sci. USA98:6865–70

191. Honda M, Orii F, Ayabe T, Imai S, Ash-ida T, Obara T, Kohgo Y. 2000. Ex-pression of glucocorticoid receptorβ inlymphocytes of patients with glucocor-ticoid-resistant ulcerative colitis.Gas-troenterology118:859–66

192. Shahidi H, Vottero A, Stratakis C, Tay-mans SE, Karl M, Longui CA, Chro-usos GP, Daughaday WH, GregorySA, Plate JMD. 1999. Imbalanced ex-pression of the glucocorticoid recep-tor isoforms in cultured lymphocytesfrom a patient with systemic glucocorti-coid resistance and chronic lymphocyticleukemia.Biochem. Biophys. Res. Com-mun.254:559–65

193. DeRijk R, Schaaf MJM, Turner G,Datson NA, Vreugdenhil E, CidlowskiJ, de Kloet ER, Emery P, SternbergEM, Detera-Wadleigh SD. 2001. A hu-man glucocorticoid receptor gene variantthat increases the stability of the gluco-corticoid receptorβ-isoform mRNA isassociated with rheumatoid arthritis.J.Rheumatol.In press

194. Kino T, Gragerov A, Kopp JB, StauberRH, Pavlakis GN, Chrousos GP. 1999.The HIV-1 virion-associated protein Vpris a coactivator of the human glucocorti-coid receptor.J. Exp. Med.189:51–61

195. New MI, Nimkarn S, Brandon DD,Cunningham-Rundles S, Wilson RC,Newfield RS, Vandermeulen J, BarronN, Russo C, Loriaux DL, O’Malley B.1999. Resistance to several steroids intwo sisters.J. Clin. Endocrinol. Metab.84:4454–64

196. Kralli A, Yamamoto KR. 1996. AnFK506-sensitive transporter selectivelydecreases intracellular levels and po-

tency of steroid hormones.J. Biol. Chem.271:17152–56

197. Medh RD, Lay RH, Schmidt TJ. 1998.Agonist-specific modulation of gluco-corticoid receptor-mediated transcrip-tion by immunosuppressants.Mol. Cell.Endocrinol.138:11–23

198. Meijer OC, de Lange EC, Breimer DD,de Boer AG, Workel JO, de KloetER. 1998. Penetration of dexamethasoneinto brain glucocorticoid targets is en-hanced in Mdr1A P-glycoprotein knock-out mice.Endocrinology139:1789–93

199. Schinkel AH, Wagenaar E, van DeemterL, Mol CA, Borst P. 1995. Absenceof the Mdr1a P-glycoprotein in mice af-fects tissue distribution and pharmacoki-netics of dexamethasone, digoxin, andcyclosporin A.J. Clin. Invest.96:1698–705

200. Gottesman MM, Pastan I. 1993. Bio-chemistry of multidrug resistance medi-ated by the multidrug transporter.Annu.Rev. Biochem.62:385–427

201. Farrell RJ, Murphy A, Long A, Don-nelly S, Cherikuri A, O’Toole D, Mah-mud N, Keeling PW, Weir DG, Kelle-her D. 2000. High multidrug resistance(P-glycoprotein 170) expression in in-flammatory bowel disease patients whofail medical therapy.Gastroenterology118:279–88

202. Geick A, Eichelbaum M, Burk O. 2001.Nuclear receptor response elements me-diate induction of intestinal MDR1 by ri-fampicin.J. Biol. Chem.276:14581–87

203. Moore JT, Kliewer S. 2000. Use of thenuclear receptor PXR to predict drug in-teractions.Toxicology153:1–10

204. Madden KS, Sanders VM, FeltenDL. 1995. Catecholamine influences andsympathetic neural modulation of im-mune responsiveness.Annu. Rev. Phar-macol. Toxicol.35:417–48

205. Felten DL, Felten SY. 1988. Sympathe-tic noradrenergic innervation of immuneorgans. Brain Behav. Immun.2:293–300

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 39: Neuroendocrine Regulationof Immunity

5 Feb 2002 7:56 AR AR152-06.tex AR152-06.SGM LaTeX2e(2001/05/10)P1: GJC

NEUROENDOCRINE REGULATION OF IMMUNITY 163

206. Hori T, Katafuchi T, Take S, ShimizuN, Niijima A. 1995. The autonomic ner-vous system as a communication channelbetween the brain and the immune sys-tem. Neuroimmunomodulation2:203–15

207. Heijnen CJ, Kavelaars A. 1999. The im-portance of being receptive.J. Neuroim-munol.100:197–202

208. Kohm AP, Sanders VM. 2000. Nore-pinephrine: a messenger from the brainto the immune system.Immunol. Today21:539–42

209. Jansson L, Holmdahl R. 1998. Estrogen-mediated immunosuppression in au-toimmune diseases.Inflamm. Res.47:290–301

210. Kincade PW, Medina KL, Payne KJ,Rossi MI, Tudor KS, Yamashita Y,Kouro T. 2000. Early B-lymphocyteprecursors and their regulation by sexsteroids.Immunol. Rev.175:128–37

211. Medina KL, Strasser A, Kincade PW.2000. Estrogen influences the differenti-ation, proliferation, and survival of early

B-lineage precursors.Blood 95:2059–67

212. Jensen EV, Greene GL, Closs LE, De-Sombre ER, Nadji M. 1982. Receptorsreconsidered: a 20-year perspective. InRecent Progress in Hormone Research,ed. RO Greep, pp. 1–34. New York: Aca-demic

213. Kuiper GG, Enmark E, Pelto-HuikkoM, Nilsson S, Gustafsson J-A. 1996.Cloning of a novel receptor expressed inrat prostate and ovary.Proc. Natl. Acad.Sci. USA93:5925–30

214. Erlandsson MC, Ohlsson C, GustafssonJ-A, Carlsten H. 2001. Role of oestro-gens receptors alpha and beta in immuneorgan development and in oestrogen-mediated effects on thymus.Immunol-ogy103:17–25

215. Misiewicz B, Griebler C, Gomez M,Raybourne R, Zelazowski E, Gold PW,Sternberg EM. 1996. The estrogen an-tagonist tamoxifen inhibits carrageenaninduced inflammation in LEW/N femalerats.Life Sci.58:281–86

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 40: Neuroendocrine Regulationof Immunity

P1: FRK

February 1, 2002 12:47 Annual Reviews AR152-FM

Annual Review of ImmunologyVolume 20, 2002

CONTENTS

FRONTISPIECE, Charles A. Janeway, Jr. xiv

A TRIP THROUGH MY LIFE WITH AN IMMUNOLOGICAL THEME,Charles A. Janeway, Jr. 1

THE B7 FAMILY OF LIGANDS AND ITS RECEPTORS: NEW PATHWAYSFOR COSTIMULATION AND INHIBITION OF IMMUNE RESPONSES,Beatriz M. Carreno and Mary Collins 29

MAP KINASES IN THE IMMUNE RESPONSE, Chen Dong, Roger J. Davis,and Richard A. Flavell 55

PROSPECTS FOR VACCINE PROTECTION AGAINST HIV-1 INFECTIONAND AIDS, Norman L. Letvin, Dan H. Barouch, and David C. Montefiori 73

T CELL RESPONSE IN EXPERIMENTAL AUTOIMMUNEENCEPHALOMYELITIS (EAE): ROLE OF SELF AND CROSS-REACTIVEANTIGENS IN SHAPING, TUNING, AND REGULATING THEAUTOPATHOGENIC T CELL REPERTOIRE, Vijay K. Kuchroo,Ana C. Anderson, Hanspeter Waldner, Markus Munder, Estelle Bettelli,and Lindsay B. Nicholson 101

NEUROENDOCRINE REGULATION OF IMMUNITY, Jeanette I. Webster,Leonardo Tonelli, and Esther M. Sternberg 125

MOLECULAR MECHANISM OF CLASS SWITCH RECOMBINATION:LINKAGE WITH SOMATIC HYPERMUTATION, Tasuku Honjo, KazuoKinoshita, and Masamichi Muramatsu 165

INNATE IMMUNE RECOGNITION, Charles A. Janeway, Jr. and RuslanMedzhitov 197

KIR: DIVERSE, RAPIDLY EVOLVING RECEPTORS OF INNATE ANDADAPTIVE IMMUNITY, Carlos Vilches and Peter Parham 217

ORIGINS AND FUNCTIONS OF B-1 CELLS WITH NOTES ON THE ROLEOF CD5, Robert Berland and Henry H. Wortis 253

E PROTEIN FUNCTION IN LYMPHOCYTE DEVELOPMENT, MelanieW. Quong, William J. Romanow, and Cornelis Murre 301

LYMPHOCYTE-MEDIATED CYTOTOXICITY, John H. Russell andTimothy J. Ley 323

SIGNAL TRANSDUCTION MEDIATED BY THE T CELL ANTIGEN

x

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.

Page 41: Neuroendocrine Regulationof Immunity

P1: FRK

February 1, 2002 12:47 Annual Reviews AR152-FM

CONTENTS xi

RECEPTOR: THE ROLE OF ADAPTER PROTEINS, Lawrence E. Samelson 371

INTERACTION OF HEAT SHOCK PROTEINS WITH PEPTIDES ANDANTIGEN PRESENTING CELLS: CHAPERONING OF THE INNATE ANDADAPTIVE IMMUNE RESPONSES, Pramod Srivastava 395

CHROMATIN STRUCTURE AND GENE REGULATION IN THE IMMUNESYSTEM, Stephen T. Smale and Amanda G. Fisher 427

PRODUCING NATURE’S GENE-CHIPS: THE GENERATION OF PEPTIDESFOR DISPLAY BY MHC CLASS I MOLECULES, Nilabh Shastri, SusanSchwab, and Thomas Serwold 463

THE IMMUNOLOGY OF MUCOSAL MODELS OF INFLAMMATION, WarrenStrober, Ivan J. Fuss, and Richard S. Blumberg 495

T CELL MEMORY, Jonathan Sprent and Charles D. Surh 551

GENETIC DISSECTION OF IMMUNITY TO MYCOBACTERIA: THE HUMANMODEL, Jean-Laurent Casanova and Laurent Abel 581

ANTIGEN PRESENTATION AND T CELL STIMULATION BY DENDRITICCELLS, Pierre Guermonprez, Jenny Valladeau, Laurence Zitvogel, ClotildeThery, and Sebastian Amigorena 621

NEGATIVE REGULATION OF IMMUNORECEPTOR SIGNALING, AndreVeillette, Sylvain Latour, and Dominique Davidson 669

CPG MOTIFS IN BACTERIAL DNA AND THEIR IMMUNE EFFECTS,Arthur M. Krieg 709

PROTEIN KINASE Cθ IN T CELL ACTIVATION, Noah Isakov and AmnonAltman 761

RANK-L AND RANK: T CELLS, BONE LOSS, AND MAMMALIANEVOLUTION, Lars E. Theill, William J. Boyle, and Josef M. Penninger 795

PHAGOCYTOSIS OF MICROBES: COMPLEXITY IN ACTION, DavidM. Underhill and Adrian Ozinsky 825

STRUCTURE AND FUNCTION OF NATURAL KILLER CELL RECEPTORS:MULTIPLE MOLECULAR SOLUTIONS TO SELF, NONSELFDISCRIMINATION, Kannan Natarajan, Nazzareno Dimasi, Jian Wang,Roy A. Mariuzza, and David H. Margulies 853

INDEXESSubject Index 887Cumulative Index of Contributing Authors, Volumes 1–20 915Cumulative Index of Chapter Titles, Volumes 1–20 925

ERRATAAn online log of corrections to Annual Review of Immunologychapters (if any, 1997 to the present) may be foundat http://immunol.annualreviews.org/

Ann

u. R

ev. I

mm

unol

. 200

2.20

:125

-163

. Dow

nloa

ded

from

arj

ourn

als.

annu

alre

view

s.or

gby

Uni

vers

ity o

f B

ern

on 1

2/05

/05.

For

per

sona

l use

onl

y.


Recommended