Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2015) Volume 6, Issue 5

Sympathetic Nervous System Regulation of Th Cells in Autoimmunity: Beyond Th1 and Th2 Cell Balance

Dianne Lorton1* and Denise L Bellinger2
1College of Arts and Sciences, Kent State University, Kent OH 44304, USA
2Department of Pathology and Human Anatomy, Loma Linda University, School of Medicine, Loma Linda, California, 92350, USA
*Corresponding Author: Dianne Lorton, PhD, College of Arts and Sciences, Kent State University, 850 University Esplanade, Bowman Hall 124A, Kent, OH, 44242, USA, Tel: 330-672-2255, Fax: 330-672-3720 Email:

Abstract

Mechanisms that cause autoimmune disease are complex and include interactions with genetic, environmental, immunologic, and neural-endocrine factors. These events are often separated by many years, suggesting that disease onset requires a triggering event that if understood, could be targeted therapeutically. How tolerance is broken and disease onset is initiated remain enigmatic. Psychological stressors are implicated in the development and progression of autoimmune diseases. First, severe life stressors are strongly associated with disease onset in up to 80% of patients. Second, the major stress pathways, the sympathetic nervous system (SNS) and hypothalamicpituitary- adrenal (HPA) axis function become pathological in autoimmune diseases. Finally, there is a common “trifecta” of dysregulated immune functions, elevated SNS activity, low parasympathetic and low HPA-axis responsiveness across most autoimmune diseases. Understanding the changes in bidirectional cross-talk between these systems leading to this “trifecta” is key to fully understanding autoimmune diseases. These neuro-endocrine systems normally function to regulate immune responses and restore immune system homeostasis after immune challenges. Here, we focus on our current understanding of the imbalance of the CD4+ T cell subtypes thought to drive autoimmunity and how dysregulation of the cross-talk between the SNS and the immune system impacts CD4+ Th cell subtype balance in the autoimmune disease, rheumatoid arthritis (RA). Our lab has shown that elevated SNS tone and altered nerve to β2-adrenergic receptor (β2-AR) signaling occurs in lymphocytes in immune organs where arthritogenic CD4+ T helper cells develop. In an animal model of RA, these receptors no longer signal via cAMP, the canonical signaling pathway for β2-ARs. Instead, β2-AR signaling is shifted towards signaling pathways expected to promote the generation of arthritogenic CD4+ T cells. These findings indicate that dysfunctions in SNS to immune system communication are pathological events that could trigger the onset of RA and, by extension, other autoimmune diseases.

Keywords: Rheumatoid arthritis; Inflammation; T helper cells; β2-Adrenergic receptor signaling; Disease mechanisms; Stress; Neuralimmune

Introduction

Rheumatoid arthritis (RA) is an autoimmune disease that afflicts~1.5 million Americans [1-3] – mostly middle-aged or older, and mostly women, but also children and young adults [4]. RA causes chronic inflammation and synovial hyperplasia in the joints that destroys articular cartilage, and leads to bone loss and joint deformities. In addition to the joint pathology, systemic pathology often occurs as part of the disease course [5]. Systemic pathology may involve various tissues and organ systems, including the skin (rheumatoid nodules), cardiovascular system (vasculitis), lungs (interstitial lung disease), kidneys, hematologic system (Felty’s syndrome), eyes and nervous system [5-8].

While the etiology of RA is not fully known, the clinical association of the major histocompatibility complex (MHC) class II allele, HLADRB1, with RA, and the presence of dysfunctional CD4+ T cells in the joints of RA patients provide compelling evidence for a central role of CD4+ T cells in the pathogenesis of this disease [9,10]. Studies using animal models of RA have further solidified the role CD4+ T cells play in disease induction and progression [11]. These studies have provided a more specific understanding of the CD4+ T cell subsets, the key cytokines they produce [12,13] and/or their signaling molecules [14-16] that are critically involved in disease initiation and progression of RA. Collectively, both patient and animal studies indicate a dysregulation of CD4+ T cell differentiation and expansion that lead to an imbalance in CD4+ T helper (Th) cell subtypes, specifically Th1, Th2, Th17, and T regulatory (Treg) cells, key components in RA pathology. However, the mechanisms that lead to this imbalance in Th cell subsets in RA are not clear.

One participant that regulates the balance of CD4+ Th cells is the sympathetic nervous system (SNS) [17-19]. This “super” system functions to maintain homeostasis in essentially every organ system of the body. A successful immune response requires the coordinated functions of many systems of the body (e.g., cardiovascular and energy metabolism) [20-23]. The SNS coordinates these organ systems so that the body can respond to a threat immediately. It subsequently returns the functions of the involved organ systems back to normal after the threat is eliminated.

Immune cells release cytokines that serve as messengers to the brain, and provide immune system functional information that is utilized by the SNS to coordinate all the systems required for an appropriate immune response [18,19] (Figure 1). Conversely, sympathetic nerves reside in the parenchyma of lymphoid organs (LOs) and release norepinephrine (NE) in response to increases in circulating and local cytokines [24,25]. NE released from sympathetic nerves activates either α- or β-adrenergic receptors (ARs) that are expressed by immune cells.

clinical-cellular-immunology-Sympathetic-immune

Figure 1: Sympathetic-immune communication relevant to rheumatoid arthritis. Neural circuits for sympathetic regulation of immune functions in secondary lymphoid organs and tissue immunity (e.g., synovial tissue in an articulating joint). The brain receives immune system information in the form of circulating cytokines and chemokines, and from activated sensory neurons. The hypothalamus responds by sending the appropriate signals to areas of the brainstem that regulate the firing rates of sympathetic neurons (not shown). Neurons in these brainstem areas send their axons to the preganglionic sympathetic neurons located in the intermediolateral cell column of the thoracic and lumbar spinal cord. These sympathetic neurons are the first neuron of a two neuron chain. Preganglionic neurons send their axons to post-ganglionic neurons located in sympathetic ganglia. Axons of post-ganglionic neurons extend into the parenchyma of secondary lymphoid tissues (spleen and lymph nodes) to regulate immune functions. The SNS also sends postganglionic nerves to most organs and tissues of the body to modulate their essential functions, as well as, to regulate inflammatory and cell-mediated immune responses that may occur in each of these tissues due to infection or tissue damage. Of relevance to RA, the synovial tissue of articulating joints is innervated by sympathetic nerves, and is thought to play a role in inflammatory arthritis.

The β2-AR is the receptor subtype almost exclusively expressed in T cells [26]. β2-AR stimulation activates a cascade of signaling messengers, including cAMP and protein kinase A (PKA). PKA phosphorylates numerous cellular proteins that ultimately modulate T cell functions [18,27] (Figure 2). By activating this signal transduction pathway, the SNS regulates the differentiation of naïve CD4+ Th cells, and augments or suppresses the functions of effector CD4+ Th cells. In RA and other autoimmune disorders, abnormalities in SNS functions contribute to the symptom burden linked to immune dysfunction – possibly triggering, but clearly mediating disease pathology.

clinical-cellular-immunology-immune-organs

Figure 2: β2-AR-mediated signal transduction in immune organs. Norepinephrine (NE) is released from sympathetic nerves. NE released from sympathetic nerves binds to β2-ARs expressed in innate and adaptive immune cells to regulate immune cell functions. Binding of NE to β2-ARs activates the G protein, Gs. Gs protein regulates the activity of adenylate cyclase (AC). Adenylate cyclase catalyzes dephosphorylation of ATP to cyclic AMP (cAMP). Cyclic AMP activates protein kinase A (PKA), G protein-coupled receptor kinases (GRK), and exchange protein activated by adenylyl cyclase (EPAC). PKA is a kinase that regulates the activity of several proteins by their phosphorylation. PKA phosphorylation of target proteins is responsible for many of the β2-AR-induced cellular responses. Activation of GRK ultimately determines the later functions and fate of β2-ARs, including the induction of alternative intracellular signaling, and receptor desensitization, internalization, recycling and/or trafficking to the lysosomes for degradation. Activation of EPAC leads to activation of the B-Raf/mitogen-activated protein kinase signaling pathway. Downstream effects of PKA, GRK and EPAC regulate diverse cellular processes, including gene transcription by transcription factors. Additionally, cAMP can bind to their binding sites on ion channels that express cAMP binding sites. Binding of cAMP to these cyclic nucleotides-gated ion channels then alter movement of ions across the channel to exert change in cell functions.

Abnormal functions of the SNS include increased sympathetic nerve firing rates [28-34], changes in receptor functions [17,35-41], and dying back of sympathetic nerves in affected joints [42-49] (Table 1 and Figure 3). Similar defects in SNS functions are observed in animal models of RA and also show SNS neuropathy and dysfunction in LOs [50-53] that is currently not feasible to assess in RA patients. Still, the extent that SNS dysfunction contributes to the imbalance in CD4+ Th cell subtypes observed in RA is not fully understood. Here, we will review the current understanding of the contribution of stress pathways, particularly the SNS to autoimmunity , CD4+ Th cells in regulating, inducing and promoting inflammatory joint disease and the potential impact of altered SNS regulation of these cells on disease processes.

clinical-cellular-immunology-Sympathetic-neurotransmission

Figure 3: Sympathetic neurotransmission in the spleen from rodent models of RA. 1, 2) Increases in the firing rate of sympathetic neurons that supply the spleen. Preganglionic sympathetic neurons stimulated by descending axons from central regulatory nuclei (e.g., the rostroventral lateral medulla) drive hyperactivity. 2, 3) The greater reactivity of the preganglionic neurons induces an increase firing rate in postganglionic sympathetic neurons (e.g., superior mesenteric-celiac ganglion) that send their axons to the spleen. Sympathetic nerves that enter the spleen (and lymph nodes) reside in T cells regions and at sites of antigen presentation and processing. 4) Increased firing rates of sympathetic nerves induce greater release of norepinephrine (NE), increasing the interaction of NE with adrenergic receptors (ARs) expressed in antigen presenting cells (APC) and Th cells. Ligand binding to ARs can cause shifts in Th cell subset balance and functions. 5) Increased nerve activity over time destroys nerves (referred to as autodestruction or “dying back”). Subsequently, sympathetic nerves respond with a sprouting response in which the nerves return, but with a different pattern of distribution in immune compartments, a process termed remodeling. The high frequency of nerve firing also increases NE concentrations/availability that induces decreases β2-AR number and sensitivity and affects AR-mediated signal pathway activation and transduction. These events also occur in the arthritic joints (not shown).

1 ↑ NE release and availability
2 LO-specific change in NE content
3 Sympathetic nerve neuropathy with subsequent nerve remodeling in splenic white pulp
4 Aberrant innervation of the red pulp, a site where activated immune cells (e.g., plasma cells) reside
5 Loss of neurotrophic factors in secondary lymphoid tissues
6 β2-AR down-regulation and desensitization (peripheral blood mononuclear cells (PBMCs), spleen cells, and synovial immune lymphocytes)
7 β2-AR down-regulation and desensitization (PBMCs, spleen cells and synovial immune immune lymphocytes)
8 Shift in β2-AR signaling pathways activated (lymph nodes that drain the arthritic joints)
β2-AR: β2-Adrenergic Receptor; LO: Lymphoid Organ; NE: Norepinephrine; PMBCs: Peripheral Blood Mononuclear Cells; SNS: Sympathetic Nervous System

Table 1: Changes in SNS-Immune System Transmission.

The Link between Stress and Autoimmune Diseases

For decades, physical and psychological stress have been implicated by many animal and human studies to induce the onset of many autoimmune diseases. The majority of patients (up to 80%) report severe or chronic emotional distress and upheaval preceding the onset of autoimmunity (like RA) [54]. Stress is well known to aggravate disease activity (Figure 4A) [55-60]. These observations coupled with evidence of dysfunction in the major stress pathways, the autonomic nervous system (both sympathetic and parasympathetic components) [28-32] and the hypothalamic-pituitary-adrenal (HPA) axis dysfunction [61-69], provide compelling support that the nervous system is important in the onset of clinical disease. Most of these studies indicate an intriguing pattern of high SNS, and low parasympathetic nervous system activity combined with a hypoactive HPA axis [33,54,60,70] (Figure 4B). Observations that arthritis does not develop on the paralyzed side in hemiplegic patients with RA [71-75] further supports a causal role of the nervous system. Similarly, in animal models of RA (i.e., adjuvantinduced arthritis (AA)), the nervous system is required for arthritic spread to the contralateral side [76-78].

clinical-cellular-immunology-Imbalance-neural

Figure 4: Imbalance of neural, hormonal and immune regulation in autoimmunity. A) The common patterns of neural-Immune dysregulation in autoimmune diseases, including RA, are illustrated. The stress systemcomposed of the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic and parasympathetic nervous systems (SNS and PaSNS, respectively) regulate immune responses, promoting immune functions in responses to danger signals and pathogens, and inhibiting immune responses as these threats are removed and homeostasis is restored. In autoimmunity, homeostasis is not restored, and an imbalance in these cross-regulatory systems develops. B) Changes in the ability of the autonomic nervous system to appropriately control body functions, such as those listed on the right side of the panel for RA patients, are often observed in patients with autoimmune diseases. These findings indicate ANS dysregulation in these diseases. The observed pattern of dysregulation generally observed for each major regulatory system is indicated by the direction of the arrows. The SNS becomes hyperreactive, whereas the PaSNS becomes hyporeactive. Similarly, dampened responsiveness of glucocorticoid receptors to glucocorticoids in patients with RA and other autoimmune disorders indicates a hypo-functional HPA axis. Increased immune system activation (production of proinflammatory cytokines and auto-antibodies and increased activity of autoreactive T cells) may, in part, result for from lost inhibitory regulation by these neural and neuroendocrine pathways that are required to restore immune system homeostasis.

The SNS is well-known to regulate both inflammatory and cellmediated immune responses, initially promoting these responses upon immune challenge; and later suppressing immune responses as the pathogen is cleared. In this manner the SNS modulates immune responses, as well as, coordinates the ensuing responses of other systems (e.g., cardiovascular, energy metabolism). This is required to mount an effective immune response and subsequently to restore homeostasis once the threat is eliminated. The chronic high SNS activity observed in patients with autoimmune diseases suggests the normal crosscommunication required to promote, then suppress cellular immunity to restore homeostasis is impaired. Discordant cross-communication between stress pathways and immune reactivity is likely causal for many stress-linked diseases (e.g., hypertension, cardiovascular disease). Disruption in any part of the bidirectional communication pathway can lead to disease development and progression [79]. The extent to which high SNS activity contributes/causes pathology in autoimmunity is unclear. Presumably, the high SNS activity contributes significantly to the immune dysregulation observed in these diseases. We [17,27,80] and others [71,81] provide substantial evidence to support the hypothesis that high SNS activity is a primary mediator of the dysregulated cell-mediated immunity in RA. However, the notion that immune dysregulation imposed by altered neural regulation could trigger the onset of clinical disease has received much less attention. A better understanding of when these stress pathways become dysregulated relative to each other, as well as relative to clinical measures of pre- and post-disease, are necessary to address these problems.

SNS regulation of the immune system is complex. It is not yet entirely clear how sympathetic nerves in secondary immune organs direct antigen-presenting cell (APC) functions and CD4+ Th cell differentiation and expansion [82-84]. Moreover, it is not clear what role the SNS plays in regulating auto-reactive CD4+ Th cells or the regulatory T (Treg) cells, cells that drive autoimmunity and maintain peripheral tolerance, respectively [85]. The presence of circulating autoantibodies can precede the clinical onset of disease by decades, indicating that tolerance is lost well before disease onset [85]. The extent that the SNS is involved in maintaining a new homeostatic “set-point” during preclinical disease is unclear. Finally, the role of the SNS in triggering disease onset remains unknown [85]. Still it is clear that the SNS is a major regulator of the differentiation and clonal expansion of CD4+ Th cells and the balance between the different CD4+ Th cell subtypes. Therefore, we hypothesize that changes in the SNS can indeed induce a loss of peripheral tolerance to induce clinical- onset of autoimmunity. We review new findings that support this role of the SNS in perpetuating immune dysregulation and loss of tolerance to promote and possibly trigger the onset of RA.

Aberrations in SNS Neurotransmission in Immune Organs: RA Rodent Models

Dysregulation of the 3-way cross-talk between the SNS, dendritic cells (DCs) and Th cells occurs in the pathogenesis of RA [81,86-88]. Its complexity is reflected by the disparate findings of drugs that target sympathetic nerves or their receptors on disease outcome of experimental arthritis (EA) [89-94]. Opposing SNS effects on disease outcome depend on the timing of SNS activation relative to the disease phase. The opposing effects support a transition phase between SNS-mediated disease promoting or ameliorating effects in early asymptomatic and late symptomatic phases of EA, respectively [76,89,91,94,95] (Figure 5). Dynamic and dramatic changes in the concentration of NE [51,89,91], sympathetic innervation [96,97] and neurotransmission [49,97-101] in secondary LOs and affected joints [49], further complicate the role of the SNS in RA development and progression. Still, noradrenergic (NA) nerves do differentially regulate immune responses in immune compartments relevant to disease development (e.g., draining lymph nodes (DLNs), spleen) and after disease onset (e.g., DLNs, spleen, skin, inflamed joint) [17,76,102]. Dissecting out LO-specific regulatory roles of NA nerves in Th cell functioning during different phases of disease is important for understanding the etiology and pathophysiology of RA, and may be translational to other types of autoimmune disorders. In this manuscript, we focus on the consequences of manipulating sympathetic signaling via treatment with a β-AR agonist on Th cell function during the effector phase of AA in disease-relevant immune organs. Identifying key events in neural-immune signaling that affect the handling of adjuvants by APCs and Th cell differentiation that triggers the breach in tolerance will provide new insights into the disease pathogenesis.

clinical-cellular-immunology-inflammatory-arthritis

Figure 5: Effect of high sympathetic reactivity or β2-AR treatment on inflammatory arthritis in rodent models. In both the collagen-induced and adjuvant-induced animal models of RA, treatments that mimic increasing sympathetic nerve activity (e.g., administration of β2-AR agonists) have differential effects on disease severity depending upon the timing of the treatment relative to disease onset. Treatment with β2-AR agonists at the time of the disease-inducing challenge to disease onset increases the severity of disease at the time when disease becomes apparent and as the disease progresses from severe to chronic stages. In contrast, when β2-AR agonists are administered beginning at disease onset, disease severity, including extent of joint inflammation and progressive joint destruction, is significantly reduced. (Tx: treatment).

Th Cell Subsets and the Th Balance in Autoimmunity

Since the SNS regulates mechanisms that drive Th cell differentiation toward different phenotypes, we will briefly overview humoral and transcriptional regulation of Th cell differentiation. Both Th and Treg cells recognize peptides presented by APCs; however, each plays distinctly different roles in host defense under various conditions of immune activation [103]. Naïve Th cells (Th0) differentiate into a variety of Th effector cells under the direction of APCs. The most relevant for autoimmunity include Th1, Th2, Th17, and Treg cells. These Th cell subsets will be the focus of this section.

CD4+ Th1 and Th2 cells

The effector Th cell subsets are defined by unique developmental programs that control their differentiation and the specific cytokine profiles they produce. Based on the Th1-Th2 cell paradigm, the cytokine profile for each subtype counter-regulates differentiation of the other subsets as well as self-reinforcing mechanisms that promote and stabilize their own maturation [104]. For example, with antigen stimulation (viral/tumor antigens), interferon-γ (IFN-γ) and interleukin (IL-12) promote the differentiation of naïve T (Th0) cells to Th1 cells (Figure 6). This response occurs by activating a Th1 cell-specific transcriptional network regulated by the transcription factor, T-bet, that results from IFN-γ and IL-12 induction of STAT1 and STAT4, respectively [104-107]. In contrast, IL-4 directs CD4+ Th0 cells to differentiate into Th2 cells by inducing the transcriptional cascade regulated by the transcription factor, GATA3, in response to IL-4-induced STAT6 [108]. Development of each of these Th cell subsets is reinforced by Th1 or Th2 cell production of IFN-γ or IL-4, respectively (Figure 7). These feedback mechanisms further push subsequent differentiation of Th0 cells toward Th1 or Th2 cells, (IFN-γ or IL-4, respectively), and counter-inhibit effects on Th2 cell development (IFN-γ) and Th1 cell differentiation (IL-4).

clinical-cellular-immunology-antigen-presenting-cells

Figure 6: Th0 cell differentiation. CD4+ T-helper (Th) cell differentiation from naïve CD4+ Th cells activated by antigen-presenting cells (APCs). In response to different pathogens or abnormal host cells, APCs present pathogen/tissue specific antigens to Th0 cells that then induce a biased differentiation into functional Th cells with specific phenotypes. The dominant Th cell subtype that develops is dependent upon the cytokine milieu produced by the APCs and naïve/activated Th cells. Differentiation of each Th cell subset results from the induction of uniquely subset-specific transcriptional programs, as indicated and described in the text. The different Th cell subtypes that differentiate from naïve Th0 cells, their hallmark transcription factor that is expressed and production a specific cytokine profiles characteristic for each subtype is indicated.

clinical-cellular-immunology-counter-regulation

Figure 7: Th cell subtype counter-regulation of the differentiation of the other Th cell subsets from activated naïve Th cells. Counter-regulation serves to allow the development of subset-specific immune responses necessary to eliminate the pathogen/tumor/antigen detected by the APCs and subsequently presented to Th0 cells. Antigen presentation to naïve Th0 cells drives their differentiation towards the most appropriate adaptive immune response. Red lines indicate inhibitory influences on Th cell differentiation and green lines illustrate lineage promoting effects. Transcription factor, downstream signaling factors, and cytokines that mediate Th cell subset regulation are indicated. (Green arrows, excitatory; red arrows, inhibitory).

CD4+ Th17 and Treg cells

Similarly, the more recently discovered Th17 cells are defined by their production of IL-17A and IL-17F. Th0 cell differentiation into Th17 cells requires transforming growth factor-β (TGF-β) and induction of the transcriptional program RORγt [109] (Figure 6). Consistent with the cross-inhibitory roles shown for Th1 and Th2 cells, IFN-γ and IL-4 are potent inhibitors of Th17 cell development and IL-17 impedes Th1 cell development [110] (Figure 7). Interestingly, another recently discovered Th cell subtype, inducible T regulatory (iTreg) cells, also requires TGF-β, as well as IL-4 and IL-10, for their differentiation from naïve Th0 cells [111]. Inducible Treg cells differ from natural Treg (nTreg) cells in that iTreg cells acquire CD25 expression outside of the thymus, typically induced by inflammation or disease processes, like autoimmunity. Th0 cells differentiate into a Th17 cell phenotype in the presence of TGF-β and IL-6 (produced by activated DC), or IL-23 (produced by IL-6- stimulated T cells) [112,113]. Like Th1- and Th2-cell driving cytokines, the cytokines required for Th17 cell differentiation form an autocrine feedback loop that further promote the differentiation and maturation of Th0 cells into Th17 cells [109,111] (Figure 7). Thus, the presence of pro-inflammatory signals from the innate immune system promotes the differentiation of naïve Th0 cells to Th17 cells [111].

In the absence of IL-6, TGF-β promotes Th0 cell differentiation towards Treg cell development by activating the FoxP3-mediated transcriptional program (Figure 6). In a non-inflammatory environment, antigen-induced activation of Th0 cells in a TGF-β-rich environment enhances iTreg cell development. Reciprocal regulation of Treg cells and Th17 cells also exists at the transcriptional level, as FoxP3 protein physically interferes with the association between RORγt and the IL-17 promoter [114-116] (Figure 7). IL-6 blocks FoxP3 interference with RORγt, initiating Th17 cell differentiation. Thus, IL-6 is pro-inflammatory by its promotion of Th17 cell differentiation and inhibition of Treg cell differentiation. The dual function of TGF-β in driving both Th17 and iTreg cell differentiation challenges the Th1- Th2 cell paradigm that distinct inductive cytokines are uniquely linked to each Th cell lineage. Indeed, induction of Th17 and Treg cell differentiation by the distinct transcriptional factors, RORγt and FoxP3, respectively, both depend on TGF-β signaling [114].

Sympathetic Regulation of Th Cell Balance

SNS regulation of Th1 and Th2 cell differentiation

The SNS regulates the differentiation of Th cells, including autoreactive cells, through the activation of α- and β2-ARs expressed on their cell surfaces [35,36,49,95]. Sympathetic nerves are present adjacent to target immune cells in secondary LOs [42,97] and the immune cell infiltrates in the tertiary lymph nodules that occur in the affected joints [117]. In secondary LOs, NE released from sympathetic nerves activates β2-ARs expressed in T cells. This receptor activation inhibits IL-2 production, which subsequently suppresses lymphocyte proliferation required for clonal expansion (reviewed in [18]). Activation of β2-ARs on Th0/Th1 cells also inhibits cellular and promotes humoral immunity by regulating the phenotypic differentiation of CD4+ Th cells in response to challenge with T cell-dependent antigens [18]. β2-AR stimulation inhibits IFN-γ production by receptor coupling to Gs protein, increased intracellular cAMP production and subsequent phosphorylation of PKA. Reduced IFN-γ limits Th0 cell differentiation towards the Th1 phenotype, and dampens IFN-γ-mediated inhibition of IL-4 production, promoting IL-4 synthesis by Th2 cells (Figure 8A). In this manner, the SNS provides a negative feedback mechanism to restore immune system homeostasis after antigen challenges that activate cellular immunity [18]. Additionally, β2-ARs preferentially inhibit IL-12 and increase IL-10 production by APCs, which subsequently promotes Th2 and inhibits Th1 differentiation [17,102] (Figure 8B).

clinical-cellular-immunology-Sympathetic-regulation

Figure 8: Sympathetic regulation of Th1 and Th2 cell balance. A, B) CD4+Th0 and Th1 cells express β2-ARs, but CD4+Th2 cells do not. APCs also express β2-ARs. These receptors provide a direct mechanism by which the SNS regulates Th cell differentiation and Th1 effector cell function. NE released from sympathetic nerves binds with β2-ARs expressed on the cell surface of Th0 and APCs mediate SNS regulation of Th1 and Th2 cell differentiation. Under normal conditions, the SNS drives the differentiation of Th2 cells by inhibiting the production of interferon-γ and IL-2 by Th0 cells, and also by reducing IL-12 and increasing IL-10 production by APCs. Ligand-receptor binding on differentiated Th1 cells modulates effector functions, most often reported to be suppressive. In contrast, Th2 cells lack β2-ARs, and therefore, the effects of the SNS on Th2 cells are indirect. (Green arrows, excitatory; red arrows, inhibitory).

SNS regulation of Th17 and Treg cell differentiation

Sympathetic regulation of the differentiation and expansion of Th17 and Treg cells is not as well studied as it is for Th1/Th2 cells. It is known that IL-2 inhibits Th17 cell differentiation from naïve Th0 cells [118], and promotes the clonal expansion of differentiated Th17 cells [119-122]. Since β2-AR stimulation of T cells suppresses IL-2 production, sympathetic activation is expected to promote Th17 cell differentiation (Figure 9A, see ①). In contrast, β2-AR-mediated IL-2 suppression is expected to inhibit clonal expansion of fully differentiated Th17 cells (Figure 9A, see ②). Moreover, under conditions that promote Th1 cell differentiation, the SNS is also expected to augment Th17 cell differentiation due to a β2-AR-mediated suppression of IFN-γ and IL- 12 production by Th1 cells [123] (Figure 9A see ③). Similarly, β2-AR suppresses IFN-γ and IL-12, both of which are potent inhibitors of Th17 development.

clinical-cellular-immunology-cytokine-profiles

Figure 9: Effects of β2-ARs stimulation on cytokine profiles and Th 1 and Th17 cell differentiation. The SNS regulates the differentiation of Th0 cells to Th17 and Treg cells, in part by modulating levels of IL-2. A) NE released from sympathetic nerves bind β2-ARs expressed in naïve Th cells. Activation of Th0 cell β2-ARs reduces the production of IL-2. IL-2 exerts an inhibitory effect on Th17 cell differentiation from naïve Th0. However, IL-2 does promote the clonal expansion of Th17 once these cells develop. By suppressing IL-2 production, the SNS is expected to promote Th17 cell differentiation, but inhibit clonal expansion of the Th17 cells once differentiated. B) Activation of β2-ARs in APCs and Th0 cells reduces their production of IL-2. Since IL-2 promotes the differentiation of Treg cells, and is required for their suppressor functions, SNS suppression of IL-2 production is anticipated to inhibit Treg cell differentiation and their suppressor functions. C) Stimulation of β2-ARs on dendritic cells (DC) that have been co-stimulated with a TLR2 and a NOD-like receptor agonist skews the immune response away from a Th1-type response but towards a Th17 response by increasing DC production of IL-6 and reducing DC production of IL-12, without changes in IL-23. (Green arrows, excitatory; red arrows, inhibitory).

In contrast to Th17 cells, IL-2 promotes the differentiation of Treg cells, and is required for their suppressor functions [120-122]. Thus, SNS-induced IL-2 suppression will likely inhibit Treg cell differentiation and their suppressor functions (Figure 9B). It is not clear how the SNS influences IL-23-promoting expansion of Th17 cells [124]. The predicted effects of the SNS via β2-AR stimulation under conditions that promote Th17 or Treg cell differentiation and clonal expansion need to be confirmed.

No studies have directly examined the effects of the SNS on Th17 cell or Treg cell differentiation under conditions that would promote the differentiation of either of these Th cell subsets. While several studies indicate that Treg cells express β2-ARs [125-127], there are no studies that indicate Th17 cells express β2-ARs. However, the SNS can influence Th17 cell differentiation through indirect effects via ARs expressed on APCs, Th0, or Th1 cells (Figure 9C). ARs modulate cytokine production by DCs and affect their ability to prime Th cells [128]. The fate of naïve CD4+ Th cell responses is dependent upon cytokines produced by DCs that sense pathogen-associated molecular patterns (PAMPs) expressed in microbes and local microenvironmental factors. DC production of IL-10, IL-12, IL-23 or TGF-β/IL6 controls the differentiation of naïve CD4+ Th cells into Th2, Th1, Th17 or Treg cells, respectively [35,124,129-136].

NE released from sympathetic nerves regulates macrophage and DC production of both IL-12 and IL-23 [137] to potentially influence Th cell differentiation towards Th1 or Th17 cells (Figure 9C). Manni and coworkers [138] find that β2-ARs stimulation shifts the balance from a Th1 to a Th17 cell response after DC priming with a toll-like receptor (TLR)2 agonist and/or a nucleotide-binding oligomerization domain receptor (NOD) agonist either in vitro or in vivo. The shift in Th1/Th17 cell balance is blocked in the presence of a β2-AR antagonist. Stimulation of β2-ARs in activated DCs increases in vitro production of IL-6, and inhibits IL-12 without affecting IL-23 secretion, a cytokine pattern favoring Th17 cell differentiation (Figure 9C), since both IL-6 and IL-23 are central mediators for the generation of Th17 cells [111,139]. Manni et al. cultured DLN cells from mice immunized with DCs stimulated in vitro with muramyl dipeptide and keyhole limpet hemocyanin in the presence of the β2-AR agonist, salbutamol. They report an increase in IL-17 and reduction in IFN-γ. These findings are consistent with a shift in Th cells towards the Th17 cell subtype. This study did not examine effects of β2-AR agonists on the generation of Treg cells directly. However, the findings that β2-AR activation increases IL-6, a cytokine that inhibits Treg cell differentiation, would be expected to suppress Treg cell development in favor of Th17 cells (Figure 10). Suppression of IL-2 by β2-AR stimulation would be anticipated to further suppress Treg cell differentiation in favor of Th17 cell development. Further, modulatory effects of the SNS on Treg cell functions are provided below.

clinical-cellular-immunology-Treg-cell-balance

Figure 10: Sympathetic regulation of Th17 and Treg cell balance. Th0 and Treg cells express β2-ARs; however, whether Th17 express this receptor has not been determined. APCs also express β2-ARs. These receptors provide a direct mechanism by which the SNS regulates Th cell differentiation and Treg effector cell function. NE released from sympathetic nerves binds with β2-ARs expressed on the cell surface of Th0 and APCs mediate SNS regulation of Th17 and Treg cell differentiation. Under normal conditions, the SNS drives the differentiation of Th17 cells by increasing the production of IL-6 and TGF-β by APCs. In contrast, activation of β2-ARs in Th0 cells inhibits Treg cell differentiation by reducing IL-2. Further, downstream signaling pathways of β2-AR in Treg cells, inhibit effects of TGF-β by suppressing TGF-β-induced activation of ERK- and JNK pathways. (Green arrows, excitatory; red arrows, inhibitory).

Similarly, Kim and Jones [133] show that DCs treated with epinephrine and lipopolysaccharide enhance IL-17 and IL-4 but not IFN-γ production by CD4+ T cells in vitro, a cytokine profile indicative of shift towards Th17 /Th2 cells and reduced Th1 cells. Notably, these findings indicate that the SNS via β2-ARs expressed on APCs are involved in defense against extracellular bacteria and in diseases in which the pathology is driven towards Th17 cell polarization. These findings have both basic and clinical relevance for immune responses against extracellular bacteria and fungi, and the pathogenesis of autoimmune and inflammatory disorders. Thus, stimulation of β2-ARs may enhance IL-17-type immune responses contrary to the concept that β2-AR agonists suppress cellular immunity and promote humoral immunity. These findings need to be confirmed and expanded.

SNS and Treg cell functions

Elevating cAMP is one mechanism by which G protein-coupled receptors (GPCRs), like β-ARs, can promote Treg cell suppression of proliferation and responder Th cell functions [125-127]. NE, the major neurotransmitter of sympathetic nerves, induces cAMP production upon ligation with β2-ARs, the major AR subtype expressed on T cells. Very little is known about sympathetic regulation of Treg cells or the influence of NE on their functions, including β-AR-induced intracellular cAMP concentrations. However, the data that is available indicate that sympathetic nerves can positively regulate suppressive activity of Treg cells via β2-AR-mediated mechanisms. It is unknown whether regulation of cAMP in Treg cells by β2-AR stimulations is responsible for greater Treg cell activity.

SNS regulation of Treg cell number and function in secondary immune organs is indicated from a study by Bhowmick et al. [140] . They demonstrate that 6-hydroxydopamine-induced sympathectomy increases murine splenic and lymph node CD4+FoxP3+ Treg cell number by a TGF-β-dependent mechanism. Moreover, this research shows that the increase in Treg cell numbers coincides with an inhibition of the induction of experimental autoimmune encephalomyelitis. Interestingly, agents that elevate cAMP, including the β-AR agonist, isoproterenol, suppress TGF-β-mediated Treg cell differentiation by inhibiting the activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) [141] (Figure 11). Cyclic AMPmediated inhibition of TGF-β occurs in a transcription-independent manner for ERK and a transcription-dependent manner for JNK. Therefore, the SNS can regulate Treg cell differentiation by activating β2- ARs, which increases intracellular levels of cAMP in Treg cells to reduce TGF-β-induced FoxP3 expression. Thus, the SNS is well positioned to regulate the balance and functions of T cells by multiple mechanisms. However, pathology in the SNS has not been explored in the imbalance between Treg and autoreactive T effector cells in autoimmune diseases. Still, these findings implicate the importance of SNS dysfunction in understanding the loss of tolerance, and in triggering the onset of clinically apparent autoimmune diseases.

clinical-cellular-immunology-adaptive-Tregs

Figure 11: Regulatory role of cAMP in TGF-β induced expression of FoxP3 and differentiation of adaptive Tregs. Cyclic AMP suppresses TGF-β-induced expression of FoxP3 by at least two mechanisms. TGF-β activates ERK and JNK, but not p38 signaling pathways. ERK and JNK induce transcription factors that induce transcription of Foxp3 genes to induce expression of FoxP3. Cyclic AMP blocks TGF-β-induced activation of ERK in a PKA-dependent and CREB independent manner. In contrast, cAMP inhibits JNK- induced FoxP3 expression through a transcription-dependent manner. Thus, by these two mechanisms cAMP suppresses TGF-β-mediated differentiation of adaptive Treg cells. (Green arrows, excitatory; red arrows, inhibitory).

CD4+FoxP3+ Treg cells use multiple mechanisms to suppress immune responses (Figures 11 and 12), maintain self-tolerance, and efficiently regulate innate [122,142,143] and adaptive immunity [144,145]. Cyclic AMP signaling plays a significant role in this process. Compared with conventional CD4+ T cells, Treg cells express high levels of cAMP, which can be transferred into activated conventional T cells via gap junctions [146,147] (Figure 12). Similarly, co-culturing Treg cells with activated murine DCs causes an increase in intracellular cAMP in Treg cells that suppresses DC function [148] (Figure 12). In Treg cells, high cAMP is induced by suppressing transcription of the microRNA, miR-142-3p, which increases the activity of adenylate cyclase (AC) 9 to phosphorylate ATP to cAMP (Figure 12, left side). FoxP3 down-regulates miR-142-p3 to keep the AC9/cAMP pathway active. In contrast, this microRNA limits the concentration of cAMP in other Th cell subsets by down-regulating AC9 transcription (Figure 12, right side). These regulatory features of Treg cells indicate that cAMP is crucial for naturally occurring Treg-mediated suppression.

clinical-cellular-immunology-AMP-mediated-regulation

Figure 12: Cyclic AMP-mediated regulation of Th cells by Treg cells. Treg cells express high levels of cAMP compared with other Th cell subsets. Cyclic AMP generally suppresses Th cell functions. One mechanism in which Treg cells suppress effector Th cells is by the transfer of cAMP to other Th cell subsets via gap junctions. The mRNA, miR-142-3p, regulates the upper limits of cAMP expression in Treg cells (cell on the left) by targeting adenylate cyclase (AC) [9]. Greater transcription of miR-142-3p inhibits AC9 activity. FoxP3 downregulates miR-142-3p transcription, which disinhibits AC9 activity to increase cAMP levels in Treg cells. In contrast, miR-142-3p inhibits AC9 expression in other T cell subsets (shown on the right), which reduces cAMP production. (Green arrows, excitatory; red arrows, inhibitory).

Like naïve T cells, Treg cells express transcripts for β1-AR, β2-AR and α2-AR, but not α1-ARs. The predominant AR expressed in Treg cells is the β2-AR. However, in comparison to naïve T cells, β2-ARs expression is 5-fold lower in Treg cells [125]. β2-ARs expressed in FoxP3+ Treg cells are functional, as they increase intracellular cAMP levels and induce PKA-dependent cAMP response element-binding protein (CREB) phosphorylation (Figure 13), and these responses are abrogated by β2- AR blockade [125]. This is consistent with PKA mediating many of the downstream actions of cAMP through nuclear transcription, and PKA as a regulator of TCR signaling through C-terminal Src kinase (Csk) or by regulating gene transcription [149-153] (Figure 13). The activating transcription factor (ATF)/CREB family bind to CRE (cAMP-response elements) in the promoter region of many genes to mediate cAMP/PKA signal transduction. These CREB modulatory proteins are encoded by three genes, Creb, Crem and Atf-1. CREB, CREM and ATF-1 suppress or activate gene expression to control T cell functions (e.g., proliferations/ differentiation, cytokine/chemokine production, apoptosis and cell homeostasis).

clinical-cellular-immunology-receptors-stimulates

Figure 13: Cyclic AMP-mediated regulation of Th cells by Treg cells. Treg cells predominantly express β2-ARs on their cell surface (top center). Ligand activation of these receptors stimulates cAMP production, which induces a cascade of PKA-dependent cAMP response element-binding protein (CREB) phosphorylation and subsequent transcription of ICER and CREM. CREM inhibits PDE activity, which prevents the breakdown of cAMP, and therefore promotes higher intracellular cAMP in Treg cells (upper right side). Elevated cAMP also increases EPAC activity which acts to increase Rap 1 and Rap 2 GTPase activity. Ras-proximate-1/2 (Rap1/2) is implicated in promoting Treg cell regulation of Th cells. FoxP3, a phenotypic marker of Treg cells, regulate intracellular cAMP concentrations by at least two mechanisms. FoxP3 inhibits PDE activity, promoting greater intracellular levels of cAMP. FoxP3 also downregulates miR-142-3p (upper left side) to keep the AC/9/cAMP pathway active, a role that is opposite to that played by this microRNA in other Th cell subsets. The cAMP generated in Treg cells and transferred to Th cells via gap junctions (center of figure) can exert both suppressive and enhancing actions on Th cells. These suppressive and enhancing effects are illustrated in the lower left and right of the figure, respectively. Greater cAMP concentrations inhibit proinflammatory cytokine production by suppressing NF-κB and Th cell differentiation by blocking IL-2 and TGF-β production, via reduced NFAT/AP-1 binding to DNA binding sites (lower right). Lower influx of calcium is induced by cAMP, which negatively regulates NFAT protein kinase. Increased cAMP concentrations in Th cells induce PKA-dependent CTLA-4 upregulation, a negative regulator of contactdependent T cell receptor (TCR) activation (lower right), preventing binding of CD28 (signal 2) necessary for T cell activation. EPAC transduces cellular actions of cAMP in Th cells that promote anergy via the activation of Rap1 (lower right). (Green arrows, excitatory; red arrows, inhibitory).

Crem encodes many activators or suppressor isoforms, including ICER (inducible cAMP early repressor). ICER is a splice variant of CRE modulator (CREM) that negatively regulates CRE-mediated transcription, including the IL-2 gene. Treg cells suppress IL-2 transcription through ICER induction in T effector cells; ICER levels correlate with cAMP levels (Figure 13; feedback shown by red arrows). ICER signals autoregulatory feedback loops of transcription that govern the downregulation of early response genes critical for immune responses [149-153]. Thus, the PKA–CREM/ICER signaling axis is considered a central pathway for cAMP-mediated T-cell suppression and Treg cell function. ICER can suppress proliferation, production of pro-inflammatory cytokines (IL-2, tumor necrosis factor-α (TNF-α), IFN-γ), anergy, and expression of FasL [154-156]. In Treg cells, ICER is a mediator of cAMP-induced suppression of Il2 and Nfat gene transcription in T effector cells [146-158]. ICER binds to multiple nuclear factor of activated T cell (NFATc1)/ activation protein 1 (AP-1) sites within the Il2 promoter, and correlates with reduced numbers of IL- 2-expressing CD4+ T cells [149,150]. NFAT and ICER form inhibitory ternary complexes on several composite NFAT/AP-1 DNA binding sites. These ternary complexes repress cytokines in addition to IL-2, including TNF-α, IL-4, IL-13, and granulocyte macrophage colonystimulating factor (GM-CSF) [159]. Thus, NFAT/ICER complexes mediate transcriptional attenuation of many NFATc1-driven cytokine promoters in conventional CD4+ T cells [160].

In contrast, Treg cells cannot induce NFATc1 at the transcriptional levels. They express low cytoplasmic levels that do not translocate efficiently to the nucleus after CD3/CD28 stimulation [161]. Under these conditions, calcium flux is reduced, along with lower calcineurin activation and greater glycogen synthase kinase-3β, which negatively regulates NFAT protein kinase [161]. Thus, the suppression mechanisms in Tregs differ markedly from those generating NFAT/AP-1 and other NFAT complexes that activate cytokine promoters in conventional CD4+ T cells. Direct contact between Tregs and CD4+ T cells increases intracellular cAMP in Treg cells. Increased cAMP induces greater expression and nuclear localization of ICER/CREM, represses NFATc1 and IL-2 production, and interferes with their activity in T cell activation. IL-2 is an autocrine growth factor that increases the number and suppressive activity of Tregs.

Treg cells express lower levels of phosphodiesterase (Pde) genes than effector T cells. Pde) gene expression and protein increase when T cells are incubate with a β-AR agonist [147,153,162]. However, in Treg cells, FoxP3 prevents the degradation of cAMP generated from GPCR activation by repressing gene expression of Pde3A and Pde4A10. Consequently, Pde3A and Pde4A10 repression promote higher cAMP levels that inhibit conventional T cell functions and amplify Treg cell function (Figure 13). Therefore, β2-AR stimulation of Treg cells drives their suppressor function and maintains tolerance and dampens Th cell immunity by increasing intracellular cAMP. Houslay and Baillie [163] report β-arrestin recruitment of PDE4 desensitizes PKA-mediated suppression of β2-AR signaling via cAMP, thus PDE4 promotes receptor signaling via cAMP.

Additionally, T effector cell suppression can be mediated by another cAMP mediator, EPAC (exchange protein elevated by cAMP) (Figure 13). EPAC is directly activated by cAMP and its activation induces Ras GTPase, Ras-proximate (Rap)1 and Rap2, implicated in enhancing negative regulation of Th cells by Treg cells [153,164-166]. Epac expression promotes the Treg anergic phenotype after anti-CD3 antibody stimulation, and in part, mediates cAMP-induced T effector cell suppression [167,168]. Naïve and activated Treg cells express higher levels of EPAC than T effector cells [153,164], suggesting a greater role for EPAC in Treg cells. Rap1 is activated by cytotoxic T lymphocyte antigen-4 (CTLA-4) engagement on T cells and mediates the subsequent inhibitory signal on IL-2 production [169,170]; EPAC-induced Rap1 may contribute to this response. CTLA-4 is a CD28-family receptor expressed on Tregs and other activated effector T cells. It negatively regulates T cell function and proliferation [171]. Mice deficient in CTLA-4 develop lethal lymphoproliferative disease [172,173].

Interestingly, β2-AR stimulation with NE or a β2-AR agonist promotes the conversion of naïve T cells to FoxP3+ T cells (iTreg cells). The number of FoxP3 iTreg cells increases two-fold when anti-CD3- stimulated naïve T cells are co-cultured with β2-ARs [125]. These effects are cAMP-, PKA- and EPAC-dependent.

Moreover, β2-AR signaling augments the in vitro suppressive function of Treg cells. Cultured at a Treg-naïve T cell ratio of 1:0.25, β2-AR activation promotes Treg-mediated reduction in anti-CD3- stimulated CD4+ T cell proliferation and IL-2 mRNA levels [125]. Lower TCR-induced IL-2 transcription is linked to higher levels of ICER in Treg cells [146]. ICER expression depends on cAMP, PKA, and pCREB [147], intracellular mediators induced by β2-AR activation that suppress Treg function (Figure 13). The effect of β2-AR stimulation on T cell proliferation and IL-2 production is blocked by β2-AR antagonist treatment, but not a specific α2a-AR antagonist [125]. Treg cells from β2-AR-/- mice display proliferative responses and IL-2 levels similar to that from unstimulated Tregs cells from wild type controls [125]. Collectively, these findings suggest that β2-AR-induced cAMP signals ICER-mediated reduction in IL-2 production and cell proliferation in activated CD4+ T cells.

Like other CD4+ T cells [174], β2-AR stimulation of Treg cells upregulates CTLA-4 in a PKA-dependent manner (Figure 13) [125]. CTLA- 4 is an important negative regulator of contact-dependent suppressive functions of Treg cells and of T cell activation [175-177]. CTLA-4 binds to CD80 or CD86 expressed on APCs with greater affinity than CD28, thereby blocking costimulatory signals, and subsequent cell cycle progression and IL-2 production [178]. A similar suppressive effect on proliferation and IL-2 is mediated by cAMP (Figure 13) [146,179]. CTLA-4 expression is regulated by adaptive immune signaling at the level of transcription [175,180]). Blocking the binding of CTLA- 4 to CD80 or CD86 prevents the accumulation of ICER, and rescues production of IL-2 [157,158,181]. Recent studies indicate that cAMP induces CTLA-4 in human CD4+ T lymphocytes [174,182]. Thus, these finding provide a possible mechanism for β2-AR-mediated suppression of anti-CD3-stimulated proliferation and IL-2 secretion. Collectively, these findings support many mechanisms through which the SNS can regulate the suppressive function of Treg cells on conventional T cells via β2-AR-cAMP signal transduction. Therefore, dysregulation of the SNS in rheumatic diseases could be a major player in triggering RA and regulating its disease course. In the next two sections, we discuss newly discovered complexities in the terminal differentiation of Th cell subsets. We believe this discussion is relevant to their regulation by the SNS. They raise important questions about whether Th effector cells are truly terminally differentiated and elude to possibly new Th cell subsets under SNS regulation.

Terminal Differentiation and Plasticity of Th17 and Treg Cells

During the host’s defense against different pathogens, antagonism between differentiating CD4+ effector T cells are thought to sustain each lineage-specific response. However, recent findings of late-stage plasticity and lineage convergence, particularly with Th17 and iTreg cells, complicate our understanding of their mutually antagonistic and reciprocal nature. Th17 and iTreg cells challenge the Th1-Th2 cell paradigm that Th cells terminally commit to a specific lineage once a key transcription factor becomes dominant. Several recent studies indicate that both Th17 and Treg cells display developmental plasticity. Th cells committed to the Th17 lineage can shift to Th1-like cells in response to IL-12, indicating the ability to shift their phenotypic fate late in their differentiation. CD4+T cells can produce both IFN-γ and IL-17, especially at sites of inflammation. This cytokine profile is reported to occur in re-stimulated Th17 cells. Co-expression of both IL-23R and IL-12Rβ2 enables Th17 to respond to either IL-23 or IL- 12 [183] (Figure 14). If re-stimulated with TGF-β alone, these cells regain high production of IL-17A and IL-17F. Interestingly, upon repeated stimulation of Th17 cells with TGF-β, a subset of Th17 cells co-produces both IL-17 and IFN-γ, despite the absence of IL-12 or IL- 23 signaling. Further, re-stimulation of Th17 cells with IL-12 induces rapid transition to a Th1-like phenotype [183-185]. In contrast, restimulation with IL-23 induces IFN-γ-producing Th17 cells that fail to produce IL-17A. Plasticity is a property of Th17 cells that does not extend to IL-17+ memory T cells. They maintain the Th17 cell phenotype regardless of being cultured with IL-12. Thus, IL7+ memory T cells achieve phenotype stability; the mechanisms by which they do so are unclear. Shifts in Th17 cell phenotype towards IFN-γ-producing cells have clinical relevance in models of RA, colitis [183], antigen-specific models of ocular inflammation [186], and models of diabetes [187,188]. In the latter case, pancreatic islet injury depends on the production of IFN-γ, indicating that IFN-γ production by Th17 cell is pathogenic in some autoimmune models.

clinical-cellular-immunology-inflammatory-environment

Figure 14: Late Th17 cell plasticity. Naïve Th cells that have been activated by antigen in the presence of TGF-β and IL-6 differentiate into Th17 cells via activation of the RORγt transcription program. IL-6 induces Th17 cells to produce IL-12, which subsequently up regulates RORγt expression. In an inflammatory environment, re-stimulation with TGF-β causes Th17 cells to regain their high production of IL-17A. However, re-stimulation with IL-12 leads to the expression of IL-23R in addition to low levels of IL-12Rβ2. In the repeated presence of relatively high TGF-β (compared to IL-23 or IL-12), Th 17 cells derived from Th0 cells produce high levels of IL-17A and IL-17F. If IL-23 is the dominant cytokine, then T-bet expression is increase and these cells further differentiate to IL-17A and IFN-γ producing Th17/Th1 cells. Interestingly, either repeated stimulation with TGF-β in the absence of IL-12 or IL-23 or a dominant presence of IL-12 can induce shift the transcriptional program towards higher activation of T-bet, resulting in Th17 cells that produce both IL-17A and IFN-γ. In contrast, Th17 cells restimulated in an inflammatory environment with IL- 23 induce a differentiation shift towards non-classic Th1 cells that produce IFN-γ due to activation of the T-bet transcription program. All of these Th17 subtypes express CD161, a traditional marker of Th17 cells. Thus, Th17 cells are responsive to both IL-23 and IL-12. Th17 cells exhibit high late plasticity, depending upon the balance of TGF-β, IL-23 and IL-12 in the milieu.

An increasing number of studies indicate iTreg cells also display substantial late developmental plasticity under conditions of elevated inflammatory cytokines. Both natural and iTreg cells have been reported to produce IFN-γ and IL-17 [189-192]. Under in vitro conditions, stimulation of mature Treg cells with IL-6 can induce IL-17, decrease FoxP3 and initiate RORγt expression. These findings suggest that mature Treg cells can further differentiate into Th17-like cells in an inflammatory environment [190,193] (Figure 15). In contrast, reciprocal conversion of Th17 cells into Treg-like cells has not been described. This may indicate that Tregs-to-Th17 cell and Th17-to-Th1 cell conversion are irreversible. Regarding Th17 and Treg cells, most findings that support their plasticity derive from in vitro studies. The extreme polarizing conditions that enable clearer phenotypes and more homogeneous T cell populations could exaggerate the potential for plasticity of these cell types. Some in vivo studies confirm the potential for their plasticity under more physiologic conditions. However, further studies are needed to determine whether similar Th cell plasticity can occur in vivo, and if so, its implications for health and disease. Phenotypic plasticity of Th17 and Treg cells has important implications in autoimmune diseases, including RA. With respect to the SNS, no studies have examined β2-ARs regulation of Th17- to-Th1 or Treg-to-Th17 phenotypic plasticity.

clinical-cellular-immunology-Late-Treg-cell

Figure 15: Late Treg cell plasticity. Multiple reports indicate that inducible Treg cells can readily switch to IL-17-producing cells upon stimulation by inflammatory cytokines. Tregs stimulated with IL-6 and IL-21 can be induced to express IL-17. Under these conditions expression of FoxP3 is down-regulated. These findings indicate that mature Treg cells can be subverted to Th17-like cells after commitment to the FoxP3 transcriptional program. There is, at present, no evidence that the reciprocal conversion of Th17 cells towards a Treg cell phenotype occurs.

Th Cell Balance, Tolerance, and RA

RA is thought to arise from a loss of immunologic tolerance to self-antigens that result in an aberrant immune response towards selfproteins. Tolerance is maintained and autoimmunity avoided by central and peripheral tolerance mechanisms [194,195]. Central tolerance is a complex developmental process. In this process, high-affinity self-reactive T and B cells are eliminated by negative selection in the thymus and bone marrow. Self-reactive T and B cells with low affinity towards self-antigens survive negative selection and join the mature T cell repertoire. Peripheral tolerance exerted by the suppressive activity of Treg cells, normally keep potentially self-reactive immune cells that escape negative selection in check. Thymic-derived nTreg cells (CD4+CD25hiFoxP3+) [196,197] and naïve CD4+ Th cell-derived iTreg cells [198,199] prevent autoimmune diseases in humans [200] and mice [201]. Treg cells maintain self-tolerance by suppressing effector T cells in the periphery [121-202]. An imbalance between CD4+ effector T cells that target self-proteins and CD4+ Treg cells is proposed to precipitate onset of autoimmune diseases. For example, activated self-reactive Th17 cells increase, while Treg cell numbers and/or their functions decline in RA patients with active disease compared with healthy controls [203,204].

Treg cells critically maintain peripheral tolerance by suppressing antigen-specific immune responses of activated self-reactive Th cells [205,206]. These cells make up about 10% of the peripheral CD4+ T cells. They constitutively express CD25 (IL-2Rα), FoxP3, CTLA- 4, and glucocorticoid-induced TNF-related protein (GITR) [207-209] – signaling proteins that serve important functional roles in Treg cells. For example, simply blocking CTLA-4 abrogates Treg cell function in vivo [210,211]. CTLA-4 blockade prevents the cell-to-cell contact between Treg and Th effector cells that is required for Tregmediated immunosuppression of T effector cells [212,213]. In contrast, GITR activation stimulates T effector cell functions and inhibits Treg suppressor activity, which potentiates immune responses against pathogens [208,214,215]. Further, studies indicate that GITR activation can break Treg cell anergy and stimulate proliferation of Treg cells [207-209].

The mechanisms responsible for lost tolerance in autoimmune diseases like RA are not entirely clear. Available evidence supports that peripheral tolerance is broken due to an imbalance between effector CD4+ Th cells and CD4+ Treg cells [216]. However, the initiating factors, and whether the imbalance includes Th1 and/or Th17 effector cells, are still debated. Early studies indicate Th1 and Th2 cell imbalance is skewed toward Th1 reactivity in animal models of RA, like collagen-induced arthritis (CIA) and AA [217,218]. For example, IFN-γ-expressing CD4+ T cells home to the inflamed arthritic joints. However, CD4+ T cells fail to release high levels of IFN-γ in the synovium of patients with RA [219-222]. In fact, neutralizing Th1 cells and/or their key cytokine, IFN-γ, yield paradoxical results on disease outcome, depending upon the animal model and timing of the intervention. Neutralizing IFN-γ activity in the pre-onset or early stages of CIA was protective, whereas, late administration exacerbated disease [223,224]. Genetic manipulation of IFN-γ in autoimmune arthritis models reveals equally paradoxical results of increased or decreased disease susceptibility/ severity depending on the model [225-229].

Recent discovery of Th17 and iTreg cells and the tolerance function of Treg cells have shifted the focus of research towards understanding Th17-Treg cell functional imbalance. However, it is likely that this is an over simplification of the disease process. Th1 cells are important in the initial disease stages and/or for disease induction as they are predominant in the pre-arthritic stage in animal models. Deleting Th1 cells prevent disease onset and their ability to adoptively transfer the disease to naïve animals in some animal models of RA [230-233]. Moreover, it seems that Th17 cell involvement in the pathogenesis depends on CD4+ Th1 cells [234].

Extensive Th1-Th17 cell cross-regulation supports dysregulation of both subpopulations, as well as, Th17 cells that express a Th1-like phenotype, in the pathogenesis of RA. Controversy remains over Th1- Th17 cell cross-regulation and late plasticity in Treg cell differentiation, but data continue to support an important role of Th1 cells in the etiology of RA. Th1 and Th17 lineages are more intertwined and complex than initially appreciated; the integrated counter-regulatory mechanisms, cooperativity and interdependency of these lineages supports this notion. The role of Th1 in autoimmune disease cannot be discounted, particularly prior to and at disease onset, and clearly warrants further investigation. Thus, it appears that Th17, Th17/Th1, and classic Th1 cells are all involved in joint pathology, with each having variable importance at different disease stages. Understanding how Th cell interactions induce and moderate clinically-apparent disease is critical for developing therapeutics that prevent disease in individuals at risk for RA.

Does the SNS Play a Role in the Loss of Tolerance in RA?

The SNS’s primary function is the fine tuning of biological responses to ensure whole body homeostasis, which includes immune system functions. During an immune challenge, the SNS coordinates systemic responses (e.g., neural, immune, cardiovascular, renal, and metabolic, etc.) required for an appropriate immune response and subsequent return to homeostasis once the challenge is eliminated. One mechanism by which the SNS regulates immune function is by modulating the production of key cytokines that regulate the differentiation of CD4+ Th subpopulations. In this manner, the SNS has a role in determining the appropriate phenotypic balance of Th effector cell subtypes to effectively handle pathogens. In the absence of overt pathogens, the role of the SNS in maintaining tolerance to prevent autoimmunity is less clear. Treatments that destroy sympathetic nerves in immune organs do not induce autoimmunity. However, animal models of autoimmune diseases, like MS or RA, indicate that enhanced SNS signaling can exacerbate disease outcome during the preclinical phase, but reduce disease severity after disease onset [51,91,94,95]. Moreover, severe life stressors often precede RA induction, suggesting that increased sympathetic activity may be a trigger for disease onset [54,56,235,236].

Identifying factors that precipitate disease onset is highly relevant clinically. Patients at risk for autoimmune diseases can be identified by circulating autoantibodies (e.g., rheumatoid factor anti-citrulline antibodies) that precede onset of clinically evident disease by as much as 10 or more years [56,237-243]. Early expression of self-reactive CD4+ T cells and B cells that cooperatively produce these autoantibodies [244] precede onset of clinically obvious disease. Loss of tolerance in the pre-disease state provides an opportunity for identifying at risk patients for preventative treatment, if the onset-triggering event(s) is(are) identified.

Under conditions of acute psychological stress, sympathetic activation can induce effector Th cell/Treg cell imbalance that favors immune reactivity. Two studies in which healthy volunteers were recruited lend support to this notion. In one study [245], acute stress increases antigen-experienced effector T cells. In the other study [127] stress reduces circulating Treg cells, implying that the stressinduced increases in effector T cells are linked to a loss of Treg cell number and/or function [127,246]. Interestingly, greater expression of β-ARs in Th1 [246] and Treg [127] cells accompany these stressinduced effects on T cell subtype number/functions. The increase in circulating Th1 cells is consistent with the elevated IFN-γ production that occurs after the acute stress. These reports pertain to acute stress. Others postulate that chronic/severe psychological stress constantly drives elevated peripheral effector T cells, and promotes inflammatory and autoimmune conditions [247]. These stress-induced effects on immunity may provide a biological explanation for psychological stress exerting an influence on autoimmune diseases [60,248,249]. However, the mechanisms by which chronic stress drive self-reactive effector T cells and suppress Treg cell inhibition of autoreactive T cells are not clear.

The SNS suppresses the production of IFN-γ in Th1 cells by activating β2-ARs expressed on their cell surface. β2-ARs are GPCRs that classically regulate cellular functions by activating the cAMP-PKA signaling pathway (canonical pathway). This signaling pathway inhibits mitogen-activated protein kinase (MAPK) signaling pathways that drive IFN-γ production. β2-AR-reduced IFN-γ production disinhibits its cross-regulatory drive on Th2 cells. Potential effects of β2-AR-induced IFN-γ suppression on Th17 differentiation has not been assessed directly. However, lowering IFN-γ is expected to promote Th17 cell differentiation under Th17 cytokine-promoting conditions. Thus, the SNS could shift the immune response towards a humoral (Th2) or Th17 (cell-mediated) response. The SNS also regulates clonal expansion of Th cells through its ability to suppress the production of IL-2, a cytokine that expands effector Th cell clones. IL-2 is critically required for Treg cell suppression of Th effector cells, providing an indirect mechanism for SNS-regulated Treg cell inhibition of Th cell functions. Through these mechanisms, SNS is positioned as an important neural regulator of immunity, consistent with a homeostatic role across the course of an immune response.

Is the SNS Functionally Intact to Signal Th cells in Autoimmune Arthritis?

The activity of the SNS is increased in patients with RA. Thus, sympathetic nerve firing rates, release of NE, and transmitter engagement of ARs are elevated in sympathetic target organs. High NE concentrations or chronic β2-AR stimulation can induce β2-AR down-regulation. β2-AR down-regulation impairs receptor coupling to the G protein, Gs, that is required to induce cAMP production (receptor desensitization). Increased SNS activity observed in RA patients may explain the decreased expression and desensitization of β2-ARs on peripheral blood lymphocytes observed in these patients. This observation is further coupled with an impaired ability of catecholamines to stimulate β2-AR-induced cAMP and to regulate immune functions [37,38,40,250]. These findings further underscore the concept of a dysfunctional SNS in these patients. For obvious reasons, the function of β2-ARs has only been examined in peripheral blood leukocytes. The peripheral blood is not innervated by the SNS, thus, elevated circulating NE in the blood is the result of spillover from all tissues that are innervated by the SNS. Also, immune cells present in the blood do not exert their immune functions in this tissue, but are being transported to tissues where they exert these functions. Thus, immune cells obtained from the blood are unlikely to be the most relevant place to assess SNS neurotransmission.

Examining β2-AR functions in lymphocytes in immune organs that participate in disease process are of greater relevance. Arthritogenic Th1 cells develop in the DLNs of arthritic limbs and the spleen and are capable of transferring disease in animal models of RA. We are using the AA model of RA in Lewis rats to determine the functional status of SNS neurotransmission and their impact on the production of IFN-γ, a key cytokine required for the development of Th1 cells in these relevant immune organs. Using the AA model, we’ve shown that in vivo β2-AR stimulation induces greater ex vivo IFN-γ production in lymphocytes from lymph nodes that drain arthritic joints [17,35]. This finding supports the SNS as a driver of Th1 (or Th17 cells with a Th1 phenotype): Treg cell imbalance. Augmented IFN-γ production is specific to lymph node cells that drain the arthritic limbs, as IFN-γ production remains unchanged in splenocytes and peripheral blood mononuclear cells (PBMCs), and reduced (the classical response) in mesenteric lymph nodes cells after β2-agonist treatment. The inability of β2-ARs to alter IFN-γ in the spleen indicates a loss in the ability of the SNS to regulate immune homeostasis in this target tissue, and is consistent with our findings that β2-ARs are down-regulated and desensitized in splenic lymphocytes [17,27,35]. Thus, the mechanism for regulating Th cell subtype balance is lost in the spleens of arthritic rats. Collectively, our data support that (1) β2- AR stimulation can promote production of pathogenic T cells in lymph nodes that drain disease-relevant tissues; (2) SNS regulation of Th cell subtype balance is lost in the spleen; (3) β2-ARs function differently in lymphocytes from lymph nodes that drain the affected limbs compared with lymphocytes from mesenteric lymph nodes or the spleen; and (4) aberrant β2-AR-mediation of Th cell functions is specific to diseaserelevant immune organs (Figure 16).

clinical-cellular-immunology-secondary-immune

Figure 16: Experimental design and results of assessing β2-AR-mediated changes in IFN-γ production by immune cells from secondary immune organs of arthritic rats. Lewis rats were treated with complete Freund’s adjuvant to induce arthritis on experimental day 0. Disease onset occurs consistently at days 11-12 in the adjuvant-induced arthritis model. Joint inflammation peaks at day 21 and then wanes by day 28; however, the joint destruction progresses steadily between days 12-28. On days 12 through 28, the β2-AR agonist, terbutaline, or vehicle was administered by intraperitoneal injection. Secondary lymphoid organs (lymph nodes that drain the arthritic hind limbs (DLNs), spleen and mesenteric lymph nodes (MLNs)) were collected and cell harvested to assess IFN-γ production in vitro. After 24 hours in culture (without additional stimulation), the media was collected and IFN-γ levels were assessed using ELISA methods. In vivo treatment with the β2-AR agonist resulted in an ex vivo increase in DLN cell production of IFN-γ. In contrast, MLN cells produced less IFN-γ. IFN-γ production by spleen cells was unchanged by β2-AR agonist treatment.

Molecular mechanisms for β2-AR-mediated increases in IFN-γ [17,26,251,252] remain a mystery, as β2-ARs typically activate the cAMP-PKA pathway, a pathway well known to suppress the production of IFN-γ. Our findings in the DLNs are not easily explained by signaling via the canonical cAMP-PKA pathway. There are a few studies in nonimmune cells that may suggest some possible cAMP/PKA-mediated routes under specific conditions (reviewed in [253]), these are pathways that have not been clearly defined. We believe it is more likely these findings can be explained by the recently described ability of the β2-AR to “switch” signaling towards MAPK pathways (reviewed in [27]). This is an important mystery to solve, as IFN-γ is a key regulator of Th cell differentiation, clonal expansion, and a cross-regulator of Th2, Th17, and Treg cell differentiation, with important implications for autoimmune pathophysiology. The Nobel Prize winning discovery in 2012 that GPCRs, which include β2-ARs, can “switch” signaling pathways from the classical cAMP pathway to MAPK signaling pathways [254,255] is a timely discovery that may solve this puzzle.

β2-ARs are dynamically regulated, in large part, by receptor ligand availability and receptor activation-induced PKA [256] (Figure 17). Similarly, G protein receptor kinases (GRKs) and β-arrestins also are critical regulators of β2-AR signaling [256]. PKA and GRKs regulate β2-AR function by site-specific phosphorylation of β2-ARs (pβ2-AR) at different serines [257-259]. High NE or agonist concentrations can cause β2-ARs to become non-responsive to additional stimulation, a condition referred to as receptor desensitization. Receptor desensitization is induced by PKA-mediated pβ2-AR (pβ2-ARPKA), and at higher agonist concentrations, by PKA- and GRK-induced pβ2-AR [260-262]. The pβ2-ARPKA alters receptor conformation, impairing Gs and enhancing Gi protein coupling that transiently activates the mitogen-activated protein kinases MAPKs, and extracellular signalregulated kinase (ERK 1/2) [262]. In contrast, pβ2-AR by GRK (pβ2- ARGRK), specifically GRK2, induces further receptor uncoupling with Gs protein, which prevents cAMP production, and recruits β-arrestin-1 to the receptor [262-264]. Binding of β-arrestin-1 to the receptor induces receptor internalization. Once the receptor is internalized, it is dephosphorylated and either recycled to the plasma membrane or degraded [265,266]. Receptor degradation reduces intracellular and/or cell surface expression of β2-ARs, a condition referred to as receptor down-regulation. High SNS nerve activity resulting in high or sustained NE availability, as reported under chronic inflammatory conditions can induce β2-AR desensitization and down-regulation.

clinical-cellular-immunology-non-classical-signaling

Figure 17: A-B. Classical and non-classical signaling of β2-ARs. A) Agonist binding to β2-ARs causes the dissociation of Gs subunits (Gsα and βγ). The Gsα subunit activates AC, which catalyzes the conversion of ATP to cAMP and the subsequent cAMP-induced activation of PKA. PKA activates transcription factors that subsequently regulate gene transcription to alter immune cell function. Receptor function is terminated by GRK2 phosphorylation of the receptor, which causes an uncoupling of Gs to AC. GRK2 recruits β-arrestin-1 to the receptor. Receptors phosphorylated by PKA and GRK2 are internalized via functions of β-arrestin-1. The internalized receptor is then either dephosphorylated and recycled to the membrane or transported to lysosomes for degradation. B) Under conditions of high agonist concentration, β2-ARs can be phosphorylated by GRK5/6 rather than GRK2. Phosphorylation of β2-AR by GRKs 5 or 6 desensitizes receptor signaling via cAMP-PKA by uncoupling the receptor from Gs. Phosphorylation of the receptor by GRK5/6 also induces β2-AR internalization by recruiting β-arrestin-2 to the receptor. Once bound to the receptor, β-arrestin-2 acts as a scaffold for the sustained activation of the MAPK, ERK 1/2. Beta-arrestin-2 activation of ERK1/2 MAPK, in turn, increases the translocation of transcription factors into the nucleus to influence gene transcription and thus, alter cell functions.

Under conditions of high concentrations of β2-AR agonists, the receptor also can be phosphorylated by PKA- and GRK5/6 (rather than GRK2) [267]. In contrast to GRK2, GRK5/6 recruits β-arrestin-2 to the receptor. Unlike β-arrestin-1, β-arrestin-2 serves as a scaffold/adaptor protein for MAPK activation [268,269]. In this manner, high β2-AR agonist concentrations can induce a G protein-independent sustained activation of the MAPK, ERK [255,270,271]. Studies demonstrating activation of this pathway have mostly been demonstrated in cell lines. We have not found any studies that have determined the extent to which this non-classical signaling pathway for β2-ARs can be activated in cells of the immune system. However, our work supports this non-classical signaling pathway for β2-ARs activation in lymphocytes present in DLNs from arthritic joints [27,35]. In contrast, β2-ARs are down-regulated and desensitized in splenocytes, although arthritogenic T cells are derived from both sites. This suggests that microenvironmental factors may be important determinants in directing β2-AR signal transduction in different lymphoid organs.

We are actively testing the hypothesis that disparate effects of β2- AR agonists on IFN-γ production in splenocytes and DLN lymphocytes from AA rats is due to differences in receptor expression, differential phosphorylation, and signaling pathways activated by receptor stimulation (Figure 18). In splenocytes harvested at peak disease (day 21), β2-ARs fail to induce an increase in intracellular cAMP production [35]. This is coupled with a decrease in affinity and number of cell surface β2-ARs. These findings are consistent with receptor desensitization and down-regulation and with the inability of β2-AR agonists to reduce IFN-γ production in splenocytes from arthritic rats.

clinical-cellular-immunology-adjuvant-induced-arthritis

Figure 18: Proposed β2-AR signaling pathways responsible for differential receptor regulation of IFN-γ production in immune cells from different secondary lymphoid organs in adjuvant-induced arthritis. A) In the DLN, where a β2-AR agonist increased IFN-γ production, receptor phosphorylation patterns support phosphorylation of the receptor by PKA and GRK5/6. This phosphorylation pattern induces receptor uncoupling from Gs and recruitment of β-arrestin-2 to the receptor, which shifts signaling towards the ERK1/2 pathway. This shift in receptor signaling then promotes IFN-γ production. B) In spleen cells, the inability of a β-AR agonist to induce an increase in cAMP coupled with the pattern of receptor phosphorylation we observed is consistent with phosphorylation by PKA and GRK2. GRK2 phosphorylation uncouples the receptor from Gs and recruits β-arrestin 1 resulting in receptor desensitization. C. In contrast, MLN cell the ability of the β2-AR agonist to suppress IFN-γ production in AA rats indicates receptors in these cells signal through the classical cAMP-PKA pathway.

Due to the limited number of cells that can be harvested from the DLN, cAMP assessments and receptor binding studies were not completed in this study. However, assessments of total β2-AR expression (cell surface and intracellular) and receptors phosphorylated by PKA and GRKs using western blot analysis were completed for both spleen and DLN cells. Strikingly, different patterns of receptor phosphorylation and changes in receptor density were observed in splenocytes and DLN cells from rats with AA, despite both being sites shown to generate arthritogenic T cells [35].

In splenocytes, total expression of β2-ARs (cell surface and intracellular) increases during peak disease (day 21), but declines during chronic severe disease (day 28). Although total expression of β2-ARs increases during peak disease, a reduction in cell surface receptor expression is observed with receptor binding assays on day 21 along with the decrease in total receptor expression on day 28. These findings suggest that β2-ARs are internalized and traffick to lysosomes for degradation. We also find that receptor phosphorylation by PKA is reduced during severe and chronic disease stages. Receptor phosphorylation by GRK does not differ at severe disease, but increases during chronic disease. The lack of PKA-induced phosphorylation of the receptors is consistent with the receptor desensitization observed on day 21. The continued decrease in PKA-mediated receptor phosphorylation during chronic disease implies that β2-ARs continue to be unresponsive. The pattern of reduced PKA-mediated phosphorylation coupled with the increase phosphorylation of the receptor by GRK, the inability of the receptor to increase intracellular cAMP, and reduced total β2-AR expression suggests that the receptor is phosphorylated by GRK2, and subsequently targeted for lysosomal degradation. Thus, these findings are consistent with impairment of the SNS to reduce IFN-γ production in the spleen due to β2-AR desensitization and down-regulation. While the data are intriguing, further studies are needed to confirm our findings and test our hypotheses more directly.

In contrast to the spleen, the total number of β2-ARs in DLN cells does not differ between arthritic and non-arthritic rats during peak or severe disease stages [35]. Further, we find a striking two-fold increase in PKA-mediated phosphorylation of β2-ARs during severe disease (day 21) followed by a significant decrease in PKA-mediated receptor phosphorylation during chronic disease (day 28) compared with nonarthritic control animals. Phosphorylation of β2-ARs by GRK significantly increases during both the severe and chronic disease stages. These findings coupled with the observed β2-AR-induced increase in IFN-γ in DLN cells from arthritic rats is consistent with receptor phosphorylation by PKA and GRK5/6, inducing a shift in signaling from the classical cAMP-PKA pathway for this receptor towards ERK1/2.

A similar shift in β2-AR signaling away from cAMP toward ERK1/2 has been demonstrated for other cells types, but not yet shown in lymphocytes or other immune cells. Thus, in AA, different GRKs (i.e., GRK2 versus GRK5/6) may be responsible for the disparate phosphorylation of β2-ARs by GRKs in spleen and DLN cells [255,259,272]. This hypothesis is consistent with the β2-ARinduced increase in IFN-γ production in DLN without affecting IFN-γ secretion in spleen cells. Our data is also consistent with reports that phosphorylation of β2-ARs by GRK5/6 shifts signaling into the ERK 1/2 cascade in HEK-293 cells treated with high physiological concentration of β-agonists [256,261,273]. Similarly, high SNS activity occurs in AA and RA. Elevated pro-inflammatory cytokines in inflammatory arthritis may also contribute to the phosphorylation of β2-ARs by GRK, as IFN-γ and IL-6, or TNF-α, IL-1, and IL-12 induce different expression of GRKs and β2-AR phosphorylation in PBMCs from healthy controls [274-276]. The SNS is crucial for maintaining homeostasis of the immune system, and particularly for regulating the balance of Th cell subtypes. Elucidating the mechanisms responsible for differential phosphorylation of β2-ARs by PKA and GRKs, and their consequences for immune function in AA and other animal models of RA, are needed to understand disease-specific SNS-immune cross-regulation in RA. These studies are necessary to determine if β2-ARs undergo downregulation and desensitization or a shift in signaling towards ERK1/2, and if so, in which immune organs these events happen, when do they occur relative to different disease stages, and what are the consequences for disease pathology. Additional studies are also required to elucidate the mechanisms by which receptor-second messenger signaling shifts occur in DLN cells in the AA rat model of RA.

We propose that one mechanism for “signal switching” is by the activation of at least one type of pattern recognition receptor (PRR). PRRs are transmembrane receptors that detect microbial ligands of which TLRs are one family of these receptors. After TLRs detect microbial ligands they activate inflammatory signaling pathways [277-280]. In the AA model, complete Freund’s adjuvant (CFA) is used to induce arthritis. It is composed of heat-killed mycobacterial cell wall that is emulsified in mineral oil. We have used the individual components of CFA to determine the role of each of its components in the altered β2-AR signaling observed in this model. Rats were challenged with CFA, the cell wall in saline or mineral oil alone. As previously described, receptor phosphorylation by GRK is increased during severe disease in the DLNs of arthritic rats challenged with CFA. This finding is also observed in rats treated with the mycobacterial cell wall suspended in saline. In contrast, in rats treated with mineral oil, receptor phosphorylation by GRK is not increased [35]. These findings indicate a role for the mycobacterial cell wall component of CFA in driving phosphorylation of the β2-ARs by GRKs. The mycobacterial component of CFA activates several TLRs (e.g., TLR2 and TLR4) [281,282]. Thus, our findings may implicate TLRs that are activated by the mycobacterial cell wall component in the regulation of β2-AR phosphorylation by GRKs.

TLRs are critical regulators of the innate and adaptive immune response to mycobacterial cell wall components. The cell wall of Mycobacteria contain components which are recognized by TLR2 in combination with TLR1 and 6, TLR9 [283,284], and possibly TLR4 [285-288]. The mycobacterial cell wall contains a number of pro-inflammatory TLR2 ligands, including lipoproteins, mycolylarabinogalactanpeptidoglycan complex (the cell wall core structure), lipids, and lipoarabinomannan [289-293]. TLR2 works as a heterodimer with TLR1 or TLR6 in a complex with CD36 to recognize lipopeptides with different structures [294-298]. Further, TLR9 cooperates with TLR2 as a mechanism by which macrophages and DCs recognize mycobacterial cell wall TLR ligands. Thus, components of the mycobacterial cell wall bind to TLRs, and activate signaling pathways in innate immune cells and in T cells. Activation of TLRs leads to pro-inflammatory cytokine production, promotes maturation of APCs, antigen presentation, and T cell activation, which are now considered to be major factors in the development of autoimmunity. In addition to the better known TLR-induced maturation of APCs and pro-inflammatory cytokine/ chemokine production, recent studies indicate TLRs are expressed in CD4+ and CD8+ T cells. In the presence of TCR activation, stimulation of TLRs expressed in T cells can function as a co-stimulatory signal to activate effector T cells and Treg cells [298]. This co-stimulation lowers the strength of the T cell signal that is required for proliferation and survival. These findings indicate an additional complexity in the mechanisms by which tolerance is broken and immune responses against self-antigens can be promoted.

Additionally, our findings that rats challenged with either CFA or mycobacterial cell wall alone promote phosphorylation of β2-ARs by GRKs in the DLNs, suggest that the cell wall component of CFA also alters the ability of the SNS to restore immune system homeostasis, and thus, the susceptibility to autoimmunity. While no studies have directly determined the mechanism by which mycobacteria cell wall components can alter β2-AR signaling in either APCs or T cells, activation of TLRs in APCs induces the nuclear factor kappa-lightchain- enhancer of activated B cells (NF-κB) signaling pathway (Figure 19). Further, activation of TCRs turns on many intracellular signal pathways, including MAPKs (p38, ERK, and JNK), PKC, and pathways that increase intracellular calcium (reviewed in [27]). These pathways activate the intracellular transcription factors, activating transcription factor 2 (ATF-2), nuclear factor of activated T cells (NFAT), activator protein 1 (AP-1), and, notably, NF-κB. Interestingly, in cardiac myocytes, recent studies indicate a possible NF-κB binding site in the promoter region of GRK5 [299]. If a similar NF-κB binding site resides in the promoter region of the GRK5 gene in APCs and/or T cells, this could provide a mechanism for TLR induced promotion of GRK5-mediated phosphorylation of β2-ARs and subsequent β-arrestin-2-mediated shift in β2-AR signaling from cAMP/PKA to ERK1/2 activation (Figure 19). If this is true for CD4+ Th0, Th1 or Th1-like Th17 cells, it would provide a mechanism that explains the observed increase in IFN-γ production in DLNs during severe arthritis when Th cells are treated with a β2-AR agonist after CFA challenge. Obviously, future studies are needed to test this hypothesis.

clinical-cellular-immunology-lymph-node-cells

Figure 19: Proposed mechanism for “switching” β2-AR signaling in draining lymph node cells. Toll-like receptors activated by mycobacterial cell wall lipoprotein and activation of T cell receptors (TCR) in T cells activate the NF-κB signaling pathway. NF-κB is well known to promote gene transcription of IFN-γ. More recently, an NF-κB site was discovered in the promoter region of the GRK 5 and GRK 6 genes. We propose that production of NF-κB, as a result of TLRs and TCR activation, increases production of GRK 5/6. GRK5/6 along with PKA then phosphorylates β2-ARs, which results in a shift in β2-AR signaling from cAMP-PKA towards the ERK1/2 pathway. This shift in signaling by the β2- AR then increases production of IFN-γ.

Of significance for RA, the ERK 1/2 signaling pathway increases IFN- production by CD4+ Th cells from rats with AA; these cells are arthritogenic and drive disease processes [243,300,301]. It is unclear whether stimulation of β2-ARs in T cells can increase IFN- by activating ERK 1/2 under conditions of chronically elevated NE release. If so, then it would provide a mechanism to explain the increased IFN-γ production [302-308] after β2-AR stimulation in activated splenic T cells [26] shortly after the onset of CIA [91,309] and DLN T cells on day 28 after CFA challenge in AA rats [17]. So far our findings are consistent with our hypotheses, but further work is required to definitively prove that changes in receptor signaling explain difference in IFN-γ production in DLN cells and splenocytes. These findings support that the imbalance between Th cell subtypes in autoimmune arthritis may result from faulty receptor regulation (desensitization, spleen) and non-canonical (DLN) β2-AR signaling at sites where autoreactive T cells differentiate.

Both faulty and non-canonical β2-AR signaling can be induced by high agonist concentrations or chronically elevated SNS activity [224]. Chronic hyperreactivity of the SNS, at least partially explains the striking association in ~80% of patients with a chronic or severe life stressor prior to onset of clinical disease. However, it is unclear whether chronically high SNS activity induces the faulty and/or non-canonical β2-AR signaling, or if faulty β2-AR signaling is responsible for severe or chronic stress-triggered onset of autoimmune disease . Furthermore, autonomic dysregulation reported in RA, juvenile chronic arthritis, and arthritic animals (reviewed in [310]) supports elevated sympathetic nerve activity. While receptor functions in lymph node and spleen cells are not available, the finding of altered β2-AR function in PBMCs of RA patients and AA rats are consistent with an increase in SNS activity. β2-AR numbers are reduced and their functional responses blunted in PBMCs collected from RA patients compared with normal agematched controls [37,38,40,99]. Diminished expression and activity of GRKs in PBMCs also occur in RA patients [39] and in animal models of RA [14]. In RA patients, GRK-2 and GRK-6 levels in PBMCs are reduced, whereas, GRK-5 levels are not altered. Given that the receptorligand concentration and GRKs regulate β-AR functions, these findings indicate that sympathetic signaling in immune cells is dysregulated in RA and animal models of RA in a manner that contribute to disease pathology. Therefore, chronically elevated sympathetic tone in patients with RA [33,101,311,312] may contribute to conditions that lead to a shift from GRK2- to GRK5-mediated phosphorylation of β2-ARs that result in β2-AR-induced ERK 1/2 signaling and the promotion of arthritogenic CD4+ Th cells.

Summary and Conclusion

Mechanisms that cause autoimmune disease are complex, and defects in both the immune system and SNS are involved. Tolerance is often broken many years before the onset of disease occurs, but the mechanisms that conspire to break tolerance and trigger disease remain enigmatic. Psychological stressors acting through defective neural and endocrine pathways are implicated; these pathways critically regulate and maintain immune system homeostasis. Therefore, the nervous system is a main player in directing immune mechanisms in the disease process. Convincing evidence indicates an imbalance in the differentiation of specific CD4+ Th cells subsets as a key mechanism for breaking tolerance and disease onset. Differentiation of naïve CD4+ Th cells and their clonal expansion and effector functions are regulated by the SNS. Norepinephrine released from activated sympathetic nerves signal via β2-ARs expressed in all CD4+ Th cell subsets. The induced rise in intracellular cAMP concentrations that result from β2-AR stimulation directs cellular activities required for Th cell subset differentiation. Typically, β2-AR-cAMP-PKA signaling inhibits cellmediated immunity and promotes humoral immunity and tolerance in healthy individuals.

Still, we have many gaps in understanding SNS regulation, particularly Th17 and Treg cell development/differentiation and effector functioning, as they are relatively newly discovered T cell subsets. Moreover, there is no information regarding SNS regulation of post-differentiational plasticity in Th17 and Treg cells that further convert these populations into unique phenotypes in RA and other autoimmune diseases. Our research indicates SNS hyperactivity in immune organs and β2-AR signal switching in Th cells conspire to promote development and differentiation of autoreactive Th cells in inflammatory arthritis.

Interestingly, the pathophysiological mechanisms are uniquely secondary immune organ-dependent. In the spleen, β2-AR neurotransmission in lymphocytes is dramatically suppressed by receptor down-regulation and desensitization due to high sympathetic nerve activity. This indicates a loss of a negative feedback loop that is required to maintain and/or restore Th cell balance in the spleen, with the likely consequence of Th cell subset differentiation into arthritisinducing T cells . In contrast, in the DLNs, our data supports a shift in lymphocyte β2-AR signaling towards signaling pathways expected to promote the generation of arthritis-inducing CD4+ Th cells. The mechanisms underlying cellular adaptation in β2-AR signal switching are not understood. Our results linking chronically heightened sympathetic nerve activity with faulty and/or non-canonical signal transduction in immune cells merits further investigation as a mechanism for autoimmunity. Additionally, altered β2-AR signaling in immune cells may involve PRRs, such as TLRs. Mycobacterial components of disease-inducing adjuvant activate subclasses of these receptors, and research by other investigators report that β2-AR ligands can affect TLR signal transduction. Collectively, these data are intriguing, and underscore the importance of further research that directly tests their role in the pathophysiology of RA.

The functional significance of β2-AR signal switching in immune cells is its downstream derailment from immune inhibition of inflammation and cellular immunity, and redirection to β2-arrestin- MAPK-mediated inflammation and cell-mediated immunity that drive disease. Such a shift in signaling prevents the innate function of the SNS – restoring and maintaining immune system homeostasis. Moreover, β2-AR signal switching in immune cells may explain many of the controversial findings whereby β2-AR agonists are reported to both inhibited and enhance specific types of immune functions. In these cases, more attention should be directed to both the context and the timing of the immune response under study.

Acknowledgment

We would like to thank Dr. Charles Malemud for the kind invitation to submit this manuscript to the Journal of Clinical and Cellular Immunology, and Christine Molinaro for carefully proofreading the paper.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brooks PM (2006) The burden of musculoskeletal disease-a global perspective.Clin Rheumatol 25: 778-781.
  2. Silman Al, Hochberg MC (2001) Epidemiology of the Rheumatic Diseases (2ndedn.) Oxford University Press, New York.
  3. Helmick CG, Felson DT, Lawrence RC, Gabriel S, Hirsch R, et al. (2008) Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part I. Arthritis Rheum 58:15-25.
  4. Lawrence RC, Helmick CG, Arnett FC, Deyo RA, Felson DT, et al. (1998) Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States.Arthritis Rheum 41: 778-799.
  5. Prete M, Racanelli V, Digiglio L, Vacca A, Dammacco F, et al. (2011) Extra-articular manifestations of rheumatoid arthritis: An update.Autoimmun Rev 11: 123-131.
  6. Cutolo M, Kitas GD, van Riel PL (2014) Burden of disease in treated rheumatoid arthritis patients: going beyond the joint. Semin Arthritis Rheum 43: 479-488.
  7. Turesson C (2013) Extra-articular rheumatoid arthritis. Curr Opin Rheumatol 25: 360-366.
  8. Gullick NJ, Scott DL (2011) Co-morbidities in established rheumatoid arthritis.Best Pract Res Clin Rheumatol 25: 469-483.
  9. Taneja V (2014) Cytokines pre-determined by genetic factors are involved in pathogenesis of rheumatoid arthritis. Cytokine 75: 216-221.
  10. Hill JA, Southwood S, Sette A, Jevnikar AM, Bell DA, et al. (2003) Cutting edge: the conversion of arginine to citrulline allows for a high-affinity peptide interaction with the rheumatoid arthritis-associated HLA-DRB1*0401 MHC class II molecule.J Immunol 171: 538-541.
  11. Houri JM, O'Sullivan FX (1995) Animal models in rheumatoid arthritis.Curr Opin Rheumatol 7: 201-205.
  12. Toh ML, Miossec P (2007) The role of T cells in rheumatoid arthritis: new subsets and new targets.Curr Opin Rheumatol 19: 284-288.
  13. Alzabin S, Williams RO (2011) Effector T cells in rheumatoid arthritis: lessons from animal models.FEBS Lett 585: 3649-3659.
  14. Noack M, Miossec P (2014) Th17 and regulatory T cell balance in autoimmune and inflammatory diseases.Autoimmun Rev 13: 668-677.
  15. Diogo D, Okada Y, Plenge RM (2014) Genome-wide association studies to advance our understanding of critical cell types and pathways in rheumatoid arthritis: recent findings and challenges.Curr Opin Rheumatol 26: 85-92.
  16. Dodeller F, Schulze-Koops H (2006) The p38 mitogen-activated protein kinase signaling cascade in CD4 T cells.Arthritis Res Ther 8: 205.
  17. Lubahn CL, Lorton D, Schaller JA, Sweeney SJ, Bellinger DL (2014) Targeting a- and ß-Adrenergic Receptors Differentially Shifts Th, Th, and Inflammatory cytokine profiles in immune organs to attenuate adjuvant arthritis. Front Immunol 5:346.
  18. Bellinger DL, Lorton D (2014) Autonomic regulation of cellular immune function.Auton Neurosci 182: 15-41.
  19. Elenkov IJ, Chrousos GP (2002) Stress hormones, proinflammatory and antiinflammatory cytokines, and autoimmunity.Ann N Y Acad Sci 966: 290-303.
  20. Nieuwdorp M, Gilijamse PW, Pai N, Kaplan LM (2014) Role of the microbiome in energy regulation and metabolism.Gastroenterology 146: 1525-1533.
  21. Okun E, Griffioen KJ, Rothman S, Wan R, Cong WN, et al. (2014) Toll-like receptors 2 and 4 modulate autonomic control of heart rate and energy metabolism.Brain Behav Immun 36: 90-100.
  22. Fullerton MD, Steinberg GR, Schertzer JD (2013) Immunometabolism of AMPK in insulin resistance and atherosclerosis.Mol Cell Endocrinol 366: 224-234.
  23. Rath E, Haller D (2011) Inflammation and cellular stress: a mechanistic link between immune-mediated and metabolically driven pathologies.Eur J Nutr 50: 219-233.
  24. Hori T, Katafuchi T, Take S, Shimizu N, Niijima A (1995) The autonomic nervous system as a communication channel between the brain and the immune system.Neuroimmunomodulation 2: 203-215.
  25. Stevens-Felten SY, Bellinger DL (1997) Noradrenergic and peptidergic innervation of lymphoid organs.Chem Immunol 69: 99-131.
  26. Swanson MA, Lee WT, Sanders VM (2001) IFN-gamma production by Th1 cells generated from naive CD4+ T cells exposed to norepinephrine.J Immunol 166: 232-240.
  27. Lorton D, Bellinger DL (2015) Molecular Mechanisms Underlying ß-adrenergic receptor-mediated cross-talk between sympathetic neurons and immune cells. Int J Mol Sci 16: 5635-5665.
  28. Adlan AM, Lip GY, Paton JF, Kitas GD, Fisher JP (2014) Autonomic function and rheumatoid arthritis: a systematic review.Semin Arthritis Rheum 44: 283-304.
  29. Syngle A, Verma I, Garg N, Krishan P (2013) Autonomic dysfunction in psoriatic arthritis.Clin Rheumatol 32: 1059-1064.
  30. Ng WF, Stangroom AJ, Davidson A, Wilton K, Mitchell S, et al. (2012) Primary Sjogrens syndrome is associated with impaired autonomic response to orthostasis and sympathetic failure.QJM 105: 1191-1199.
  31. Janse van Rensburg DC, Ker JA, Grant CC, Fletcher L (2012) Autonomic impairment in rheumatoid arthritis.Int J Rheum Dis 15: 419-426.
  32. Vlcek M, Rovensky J, Eisenhofer G, Radikova Z, Penesova A, et al. (2012) Autonomic nervous system function in rheumatoid arthritis.Cell Mol Neurobiol 32: 897-901.
  33. Stojanovich L (2009) Autonomic dysfunction in autoimmune rheumatic disease.Autoimmun Rev 8: 569-572.
  34. Mandl T, Granberg V, Apelqvist J, Wollmer P, Manthorpe R, et al. (2008) Autonomic nervous symptoms in primary Sjogren's syndrome.Rheumatology (Oxford) 47: 914-919.
  35. Lorton D, Bellinger DL, Schaller JA, Shewmaker E, Osredkar T, et al. (2013) Altered sympathetic-to-immune cell signaling via ß2-adrenergic receptors in adjuvant arthritis.Clin Dev Immunol 2013: 764395.
  36. Mishima K, Otani H, Tanabe T, Kawasaki H, Oshiro A, et al. (2001) Molecular mechanisms for alpha2-adrenoceptor-mediated regulation of synoviocyte populations.Jpn J Pharmacol 85: 214-226.
  37. Wahle M, Kölker S, Krause A, Burmester GR, Baerwald CG (2001) Impaired catecholaminergic signalling of B lymphocytes in patients with chronic rheumatic diseases.Ann Rheum Dis 60: 505-510.
  38. Wahle M, Krause A, Kölker S, von Wichert P, Baerwald CG (1999) Intracellular cAMP and beta 2-adrenergic receptors on CD19+ lymphocytes in patients with rheumatoid arthritis.Ann N Y Acad Sci 876: 309-311.
  39. Lombardi MS, Kavelaars A, Schedlowski M, Bijlsma JW, Okihara KL, et al. (1999) Decreased expression and activity of G-protein-coupled receptor kinases in peripheral blood mononuclear cells of patients with rheumatoid arthritis.FASEB J 13: 715-725.
  40. Baerwald CG, Wahle M, Ulrichs T, Jonas D, von Bierbrauer A, et al. (1999) Reduced catecholamine response of lymphocytes from patients with rheumatoid arthritis.Immunobiology 200: 77-91.
  41. Zoukos Y, Leonard JP, Thomaides T, Thompson AJ, Cuzner ML (1992) beta-Adrenergic receptor density and function of peripheral blood mononuclear cells are increased in multiple sclerosis: a regulatory role for cortisol and interleukin-1.Ann Neurol 31: 657-662.
  42. Lorton D, Lubahn C, Klein N, Schaller J, Bellinger DL (1999) Dual role for noradrenergic innervation of lymphoid tissue and arthritic joints in adjuvant-induced arthritis.Brain Behav Immun 13: 315-334.
  43. da Silva JA, Fonseca JE, Graça L, Moita L, Carmo-Fonseca M (1996) Reinnervation of post-arthritic joints in the rat.Clin Exp Rheumatol 14: 43-51.
  44. Mapp PI, Walsh DA, Garrett NE, Kidd BL, Cruwys SC, et al. (1994) Effect of three animal models of inflammation on nerve fibres in the synovium.Ann Rheum Dis 53: 240-246.
  45. Pongratz G, Straub RH (2013) Role of peripheral nerve fibres in acute and chronic inflammation in arthritis.Nat Rev Rheumatol 9: 117-126.
  46. Dirmeier M, Capellino S, Schubert T, Angele P, Anders S, et al. (2008) Lower density of synovial nerve fibres positive for calcitonin gene-related peptide relative to substance P in rheumatoid arthritis but not in osteoarthritis. Rheumatology (Oxford) 47: 36-40.
  47. Weidler C, Holzer C, Harbuz M, Hofbauer R, Angele P, et al. (2005) Low density of sympathetic nerve fibres and increased density of brain derived neurotrophic factor positive cells in RA synovium.Ann Rheum Dis 64: 13-20.
  48. Miller LE, Weidler C, Falk W, Angele P, Schaumburger J, et al. (2004) Increased prevalence of semaphorin 3C, a repellent of sympathetic nerve fibers, in the synovial tissue of patients with rheumatoid arthritis.Arthritis Rheum 50: 1156-1163.
  49. Miller LE, Jüsten HP, Schölmerich J, Straub RH (2000) The loss of sympathetic nerve fibers in the synovial tissue of patients with rheumatoid arthritis is accompanied by increased norepinephrine release from synovial macrophages. FASEB J 14: 2097-2107.
  50. Bellinger DL, Millar BA, Perez S, Carter J, Wood C, et al. (2008) Sympathetic modulation of immunity: relevance to disease.Cell Immunol 252: 27-56.
  51. Lorton D, Lubahn C, Felten SY, Bellinger D (1997) Norepinephrine content in primary and secondary lymphoid organs is altered in rats with adjuvant-induced arthritis.Mech Ageing Dev 94: 145-163.
  52. Felten DL, Felten SY, Bellinger DL, Lorton D (1992) Noradrenergic and peptidergic innervation of secondary lymphoid organs: role in experimental rheumatoid arthritis.Eur J Clin Invest 22 Suppl 1: 37-41.
  53. Bellinger DL, Lorton D, Felten SY, Felten DL (1992) Innervation of lymphoid organs and implications in development, aging, and autoimmunity.Int J Immunopharmacol 14: 329-344.
  54. Stojanovich L, Marisavljevich D (2008) Stress as a trigger of autoimmune disease.Autoimmun Rev 7: 209-213.
  55. Bugajska J, Brzosko M, Jedryka-Góral A, Gluszko P, Zolnierczyk-Zreda D, et al. (2010) Psychological stress in rheumatoid arthritis patients: a comparative Polish-German study: summary of the current conceptualization of the role of stress in rheumatoid arthritis. Autoimmun Rev 9: 211-215.
  56. Cutolo M, Straub RH (2006) Stress as a risk factor in the pathogenesis of rheumatoid arthritis.Neuroimmunomodulation 13: 277-282.
  57. Dube SR, Fairweather D, Pearson WS, Felitti VJ, Anda RF, et al. (2009) Cumulative childhood stress and autoimmune diseases in adults.Psychosom Med 71: 243-250.
  58. Herrmann M, Schölmerich J, Straub RH (2000) Stress and rheumatic diseases. Rheum Dis Clin North Am 26: 737-76, viii.
  59. Shih M, Hootman JM, Strine TW, Chapman DP, Brady TJ (2006) Serious psychological distress in U.S. adults with arthritis.J Gen Intern Med 21: 1160-1166.
  60. Straub RH, Dhabhar FS, Bijlsma JW, Cutolo M (2005) How psychological stress via hormones and nerve fibers may exacerbate rheumatoid arthritis.Arthritis Rheum 52: 16-26.
  61. Evers AW, Verhoeven EW, Van Middendorp H, Sweep FC, Kraaimaat FW, et al. (2014) Does stress affect the joints? Daily stressors, stress vulnerability, immune and HPA axis activity, and short-term disease and symptom fluctuations in rheumatoid arthritis. Ann Rheum Dis 73:1683-1718.
  62. Jänig W, Green PG (2014) Acute inflammation in the joint: its control by the sympathetic nervous system and by neuroendocrine systems.Auton Neurosci 182: 42-54.
  63. Imrich R, Rovenský J (2010) Hypothalamic-pituitary-adrenal axis in rheumatoid arthritis.Rheum Dis Clin North Am 36: 721-727.
  64. Tzioufas AG, Tsonis J, Moutsopoulos HM (2008) Neuroendocrine dysfunction in Sjogren's syndrome.Neuroimmunomodulation 15: 37-45.
  65. Johnson EO, Kostandi M, Moutsopoulos HM (2006) Hypothalamic-pituitary-adrenal axis function in Sjögren's syndrome: mechanisms of neuroendocrine and immune system homeostasis.Ann N Y Acad Sci 1088: 41-51.
  66. Kurtais Y, Tur BS, Elhan AH, Erdogan MF, Yalçin P (2006) Hypothalamic-pituitary-adrenal hormonal responses to exercise stress test in patients with rheumatoid arthritis compared to healthy controls.J Rheumatol 33: 1530-1537.
  67. Geenen R, Van Middendorp H, Bijlsma JW (2006) The impact of stressors on health status and hypothalamic-pituitary-adrenal axis and autonomic nervous system responsiveness in rheumatoid arthritis.Ann N Y Acad Sci 1069: 77-97.
  68. Harbuz MS, Richards LJ, Chover-Gonzalez AJ, Marti-Sistac O, Jessop DS (2006) Stress in autoimmune disease models.Ann N Y Acad Sci 1069: 51-61.
  69. Eijsbouts AM, van den Hoogen FH, Laan RF, Hermus AR, Sweep CG, et al. (2005) Hypothalamic-pituitary-adrenal axis activity in patients with rheumatoid arthritis.Clin Exp Rheumatol 23: 658-664.
  70. Geenen R, Godaert GL, Jacobs JW, Peters ML, Bijlsma JW (1996) Diminished autonomic nervous system responsiveness in rheumatoid arthritis of recent onset.J Rheumatol 23: 258-264.
  71. THOMPSON M, BYWATERS EG (1962) Unilateral rheumatoid arthritis following hemiplegia.Ann Rheum Dis 21: 370-377.
  72. Yaghmai I, Rooholamini SM, Faunce HF (1977) Unilateral rheumatoid arthritis: protective effect of neurologic deficits.AJR Am J Roentgenol 128: 299-301.
  73. Hamilton S (1983) Unilateral rheumatoid arthritis in hemiplegia.J Can Assoc Radiol 34: 49-50.
  74. Ueno Y, Sawada K, Imura H (1983) Protective effect of neural lesion on rheumatoid arthritis.Arthritis Rheum 26: 118.
  75. Smith RD (1979) Effect of hemiparesis on rheumatoid arthritis.Arthritis Rheum 22: 1419-1420.
  76. Levine JD, Dardick SJ, Roizen MF, Helms C, Basbaum AI (1986) Contribution of sensory afferents and sympathetic efferents to joint injury in experimental arthritis.J Neurosci 6: 3423-3429.
  77. Lorton D, Lubahn C, Engan C, Schaller J, Felten DL, et al. (2000) Local application of capsaicin into the draining lymph nodes attenuates expression of adjuvant-induced arthritis.Neuroimmunomodulation 7: 115-125.
  78. Stangenberg L, Burzyn D, Binstadt BA, Weissleder R, Mahmood U, et al. (2014) Denervation protects limbs from inflammatory arthritis via an impact on the microvasculature.Proc Natl Acad Sci U S A 111: 11419-11424.
  79. Kin NW, Sanders VM (2006) It takes nerve to tell T and B cells what to do.J Leukoc Biol 79: 1093-1104.
  80. Lorton D, Lubahn C, Bellinger DL (2003) Potential use of drugs that target neural-immune pathways in the treatment of rheumatoid arthritis and other autoimmune diseases.Curr Drug Targets Inflamm Allergy 2: 1-30.
  81. Wahle M, Hanefeld G, Brunn S, Straub RH, Wagner U, et al. (2006) Failure of catecholamines to shift T-cell cytokine responses toward a Th2 profile in patients with rheumatoid arthritis.Arthritis Res Ther 8: R138.
  82. Nance DM, Sanders VM (2007) Autonomic innervation and regulation of the immune system (1987-2007).Brain Behav Immun 21: 736-745.
  83. Decaris E, Guingamp C, Chat M, Philippe L, Grillasca JP, et al. (1999) Evidence for neurogenic transmission inducing degenerative cartilage damage distant from local inflammation.Arthritis Rheum 42: 1951-1960.
  84. Ramer-Quinn DS, Swanson MA, Lee WT, Sanders VM (2000) Cytokine production by naive and primary effector CD4+ T cells exposed to norepinephrine.Brain Behav Immun 14: 239-255.
  85. Goronzy JJ, Weyand CM (2009) Developments in the scientific understanding of rheumatoid arthritis.Arthritis Res Ther 11: 249.
  86. Maestroni GJ1 (2004) Modulation of skin norepinephrine turnover by allergen sensitization: impact on contact hypersensitivity and T helper priming.J Invest Dermatol 122: 119-124.
  87. Lorton D, Lubahn CL, Zautra AJ, Bellinger DL (2008) Proinflammatory cytokines and sickness behavior in rheumatic diseases.Curr Pharm Des 14: 1242-1260.
  88. del Rey A, Wolff C, Wildmann J, Randolf A, Hahnel A, et al. (2008) Disrupted brain-immune system-joint communication during experimental arthritis.Arthritis Rheum 58: 3090-3099.
  89. Lubahn CL, Schaller JA, Bellinger DL, Sweeney S, Lorton D (2004) The importance of timing of adrenergic drug delivery in relation to the induction and onset of adjuvant-induced arthritis.Brain Behav Immun 18: 563-571.
  90. Härle P, Pongratz G, Albrecht J, Tarner IH, Straub RH (2008) An early sympathetic nervous system influence exacerbates collagen-induced arthritis via CD4+CD25+ cells.Arthritis Rheum 58: 2347-2355.
  91. Härle P, Möbius D, Carr DJ, Schölmerich J, Straub RH (2005) An opposing time-dependent immune-modulating effect of the sympathetic nervous system conferred by altering the cytokine profile in the local lymph nodes and spleen of mice with type II collagen-induced arthritis.Arthritis Rheum 52: 1305-1313.
  92. Coderre TJ, Basbaum AI, Helms C, Levine JD (1991) High-dose epinephrine acts at alpha 2-adrenoceptors to suppress experimental arthritis.Brain Res 544: 325-328.
  93. Coderre TJ, Basbaum AI, Dallman MF, Helms C, Levine JD (1990) Epinephrine exacerbates arthritis by an action at presynaptic ß2-adrenoceptors.Neuroscience 34: 521-523.
  94. Levine JD, Coderre TJ, Helms C, Basbaum AI (1988) Beta 2-adrenergic mechanisms in experimental arthritis.Proc Natl Acad Sci U S A 85: 4553-4556.
  95. Malfait AM, Malik AS, Marinova-Mutafchieva L, Butler DM, Maini RN, et al. (1999) The beta2-adrenergic agonist salbutamol is a potent suppressor of established collagen-induced arthritis: mechanisms of action.J Immunol 162: 6278-6283.
  96. Lorton D, Lubahn C, Lindquist CA, Schaller J, Washington C, et al. (2005) Changes in the density and distribution of sympathetic nerves in spleens from Lewis rats with adjuvant-induced arthritis suggest that an injury and sprouting response occurs.J Comp Neurol 489: 260-273.
  97. Lorton D, Lubahn C, Sweeney S, Major A, Lindquist CA, et al. (2009) Differences in the injury/sprouting response of splenic noradrenergic nerves in Lewis rats with adjuvant-induced arthritis compared with rats treated with 6-hydroxydopamine.Brain Behav Immun 23: 276-285.
  98. Baerwald C, Graefe C, Muhl C, Von Wichert P, Krause A (1992) Beta 2-adrenergic receptors on peripheral blood mononuclear cells in patients with rheumatic diseases.Eur J Clin Invest 22 Suppl 1: 42-46.
  99. Baerwald CG, Laufenberg M, Specht T, von Wichert P, Burmester GR, et al. (1997) Impaired sympathetic influence on the immune response in patients with rheumatoid arthritis due to lymphocyte subset-specific modulation of beta 2-adrenergic receptors.Br J Rheumatol 36: 1262-1269.
  100. Wahle M, Neumann RP, Moritz F, Krause A, Buttgereit F, et al. (2005) Beta2-adrenergic receptors mediate the differential effects of catecholamines on cytokine production of PBMC.J Interferon Cytokine Res 25: 384-394.
  101. Kuis W, de Jong-de Vos van Steenwijk C, Sinnema G, Kavelaars A, Prakken B, et al. (1996) The autonomic nervous system and the immune system in juvenile rheumatoid arthritis.Brain Behav Immun 10: 387-398.
  102. Panina-Bordignon P, Mazzeo D, Lucia PD, D'Ambrosio D, Lang R, et al. (1997) Beta2-agonists prevent Th1 development by selective inhibition of interleukin 12.J Clin Invest 100: 1513-1519.
  103. Zhu J, Paul WE (2008) CD4 T cells: fates, functions, and faults.Blood 112: 1557-1569.
  104. Zhang Y, Zhang Y, Gu W, Sun B (2014) TH1/TH2 cell differentiation and molecular signals.Adv Exp Med Biol 841: 15-44.
  105. Li P, Spolski R, Liao W, Leonard WJ (2014) Complex interactions of transcription factors in mediating cytokine biology in T cells.Immunol Rev 261: 141-156.
  106. Zeng WP1 (2013) 'All things considered': transcriptional regulation of T helper type 2 cell differentiation from precursor to effector activation.Immunology 140: 31-38.
  107. Kaplan MH, Grusby MJ (1998) Regulation of T helper cell differentiation by STAT molecules. J Leukoc Biol 64: 2-5.
  108. Yagi R, Zhu J, Paul WE (2011) An updated view on transcription factor GATA3-mediated regulation of Th1 and Th2 cell differentiation. Int Immunol 23: 415-420.
  109. Martinez GJ, Nurieva RI, Yang XO, Dong C (2008) Regulation and function of proinflammatory TH17 cells. Ann N Y Acad Sci 1143: 188-211.
  110. Nakae S, Iwakura Y, Suto H, Galli SJ (2007) Phenotypic differences between Th1 and Th17 cells and negative regulation of Th1 cell differentiation by IL-17. J Leukoc Biol 81: 1258-1268.
  111. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, et al. (2006) Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235-238.
  112. Diaconu CC, Neagu AI, Lungu R, Tardei G, Alexiu I, et al. (2010) Plasticity of regulatory T cells under cytokine pressure. Roum Arch Microbiol Immunol 69: 190-196.
  113. Korn T, Oukka M, Kuchroo V, Bettelli E (2007) Th17 cells: effector T cells with inflammatory properties. Semin Immunol 19: 362-371.
  114. Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, et al. (2008) TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature 453: 236-240.
  115. Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K, et al. (2008) Foxp3 inhibits RORgammat-mediated IL-17A mRNA transcription through direct interaction with RORgammat. J Biol Chem 283: 17003-17008.
  116. Zhang F, Meng G, Strober W (2008) Interactions among the transcription factors Runx, RORgammat and Foxp3 regulate the differentiation of interleukin 17-producing T cells. Nat Immunol 9: 1297-1306.
  117. Schaible HG, Straub RH (2014) Function of the sympathetic supply in acute and chronic experimental joint inflammation. Auton Neurosci 182: 55-64.
  118. Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, et al. (2007) Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 26: 371-381.
  119. Amadi-Obi A, Yu CR, Liu X, Mahdi RM, Clarke GL, et al. (2007) TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med 13: 711-718.
  120. Littman DR, Rudensky AY (2010) Th17 and regulatory T cells in mediating and restraining inflammation. Cell 140: 845-858.
  121. Sakaguchi S, Yamaguchi T, Nomura T, Ono M (2008) Regulatory T cells and immune tolerance. Cell 133: 775-787.
  122. Shevach EM (2009) Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity 30: 636-645.
  123. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, et al. (2005) Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 6: 1123-1132.
  124. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, et al. (2005) IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201: 233-240.
  125. Guereschi MG, Araujo LP, Maricato JT, Takenaka MC, Nascimento VM, et al. (2013) Beta2-adrenergic receptor signaling in CD4+ Foxp3+ regulatory T cells enhances their suppressive function in a PKA-dependent manner. Eur J Immunol 43: 1001-1012.
  126. Vida G, Peña G, Kanashiro A, Thompson-Bonilla Mdel R, Palange D, et al. (2011) ß2-Adrenoreceptors of regulatory lymphocytes are essential for vagal neuromodulation of the innate immune system. FASEB J 25: 4476-4485.
  127. Freier E, Weber CS, Nowottne U, Horn C, Bartels K, et al. (2010) Decrease of CD4(+)FOXP3(+) T regulatory cells in the peripheral blood of human subjects undergoing a mental stressor. Psychoneuroendocrinology 35: 663-673.
  128. Seiffert K, Hosoi J, Torii H, Ozawa H, Ding W, et al. (2002) Catecholamines inhibit the antigen-presenting capability of epidermal Langerhans cells. J Immunol 168: 6128-6135.
  129. Aggarwal S, Ghilardi N, Xie MH, de Sauvage FJ, Gurney AL (2003) Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J Biol Chem 278: 1910-1914.
  130. Langrish CL, McKenzie BS, Wilson NJ, de Waal Malefyt R, Kastelein RA, et al. (2004) IL-12 and IL-23: master regulators of innate and adaptive immunity. Immunol Rev 202: 96-105.
  131. Joffre O, Nolte MA, Spörri R, Reis e Sousa C (2009) Inflammatory signals in dendritic cell activation and the induction of adaptive immunity. Immunol Rev 227: 234-247.
  132. Miossec P (2008) Dynamic interactions between T cells and dendritic cells and their derived cytokines/chemokines in the rheumatoid synovium. Arthritis Res Ther 10: S2.
  133. Kim BJ, Jones HP (2010) Epinephrine-primed murine bone marrow-derived dendritic cells facilitate production of IL-17A and IL-4 but not IFN-γ by CD4+ T cells. Brain Behav Immun 24: 1126-1136.
  134. Maestroni GJ (2002) Short exposure of maturing, bone marrow-derived dendritic cells to norepinephrine: impact on kinetics of cytokine production and Th development. J Neuroimmunol 129: 106-114.
  135. Maestroni GJ, Mazzola P (2003) Langerhans cells beta 2-adrenoceptors: role in migration, cytokine production, Th priming and contact hypersensitivity. J Neuroimmunol 144: 91-99.
  136. Ross PJ, Lavelle EC, Mills KH, Boyd AP (2004) Adenylate cyclase toxin from Bordetella pertussis synergizes with lipopolysaccharide to promote innate interleukin-10 production and enhances the induction of Th2 and regulatory T cells. Infect Immun 72: 1568-1579.
  137. Goyarts E, Matsui M, Mammone T, Bender AM, Wagner JA, et al. (2008) Norepinephrine modulates human dendritic cell activation by altering cytokine release. Exp Dermatol 17: 188-196.
  138. Manni M, Granstein RD, Maestroni G (2011) ß2-Adrenergic agonists bias TLR-2 and NOD2 activated dendritic cells towards inducing an IL-17 immune response. Cytokine 55: 380-386.
  139. Iwakura Y, Ishigame H (2006) The IL-23/IL-17 axis in inflammation. J Clin Invest 116: 1218-1222.
  140. Bhowmick S, Singh A, Flavell RA, Clark RB, O'Rourke J, et al. (2009) The sympathetic nervous system modulates CD4(+)FoxP3(+) regulatory T cells via a TGF-beta-dependent mechanism. J Leukoc Biol 86: 1275-1283.
  141. Cao J, Zhang X, Wang Q, Wang X, Jin J, et al. (2013) Cyclic AMP suppresses TGF-ß-mediated adaptive Tregs differentiation through inhibiting the activation of ERK and JNK. Cell Immunol 285: 42-48.
  142. Lund JM, Hsing L, Pham TT, Rudensky AY (2008) Coordination of early protective immunity to viral infection by regulatory T cells. Science 320: 1220-1224.
  143. Yuan X, Cheng G, Malek TR (2014) The importance of regulatory T-cell heterogeneity in maintaining self-tolerance. Immunol Rev 259: 103-114.
  144. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330-336.
  145. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057-1061.
  146. Bopp T, Becker C, Klein M, Klein-Hessling S, Palmetshofer A, et al. (2007) Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. J Exp Med 204: 1303-1310.
  147. Bazhin AV, Kahnert S, Kimpfler S, Schadendorf D, Umansky V (2010) Distinct metabolism of cyclic adenosine monophosphate in regulatory and helper CD4+ T cells. Mol Immunol 47: 678-684.
  148. Fassbender M, Gerlitzki B, Ullrich N, Lupp C, Klein M, et al. (2010) Cyclic adenosine monophosphate and IL-10 coordinately contribute to nTreg cell-mediated suppression of dendritic cell activation. Cell Immunol 265: 91-96.
  149. Mosenden R, Taskén K (2011) Cyclic AMP-mediated immune regulation--overview of mechanisms of action in T cells. Cell Signal 23: 1009-1016.
  150. Vaeth M, Gogishvili T, Bopp T, Klein M, Berberich-Siebelt F, et al. (2011) Regulatory T cells facilitate the nuclear accumulation of inducible cAMP early repressor (ICER) and suppress nuclear factor of activated T cell c1 (NFATc1). Proc Natl Acad Sci U S A 108: 2480-2485.
  151. Stokka AJ, Mosenden R, Ruppelt A, Lygren B, Taskén K (2009) The adaptor protein EBP50 is important for localization of the protein kinase A-Ezrin complex in T-cells and the immunomodulating effect of cAMP. Biochem J 425: 381-388.
  152. Vang T, Torgersen KM, Sundvold V, Saxena M, Levy FO, et al. (2001) Activation of the COOH-terminal Src kinase (Csk) by cAMP-dependent protein kinase inhibits signaling through the T cell receptor. J Exp Med 193: 497-507.
  153. Vang AG, Housley W, Dong H, Basole C, Ben-Sasson SZ, et al. (2013) Regulatory T-cells and cAMP suppress effector T-cells independently of PKA-CREM/ICER: a potential role for Epac. Biochem J 456: 463-473.
  154. Bodor J, Habener JF (1998) Role of transcriptional repressor ICER in cyclic AMP-mediated attenuation of cytokine gene expression in human thymocytes. J Biol Chem 273: 9544-9551.
  155. Higai K, Tsukada M, Moriya Y, Azuma Y, Matsumoto K (2009) Prolonged high glucose suppresses phorbol 12-myristate 13-acetate and ionomycin-induced interleukin-2 mRNA expression in Jurkat cells. Biochim Biophys Acta 1790: 8-15.
  156. Powell JD, Lerner CG, Ewoldt GR, Schwartz RH (1999) The -180 site of the IL-2 promoter is the target of CREB/CREM binding in T cell anergy. J Immunol 163: 6631-6639.
  157. Bodor J, Bodorova J, Gress RE (2000) Suppression of T cell function: a potential role for transcriptional repressor ICER. J Leukoc Biol 67: 774-779.
  158. Bodor J, Fehervari Z, Diamond B, Sakaguchi S (2007) ICER/CREM-mediated transcriptional attenuation of IL-2 and its role in suppression by regulatory T cells. Eur J Immunol 37: 884-895.
  159. Hogan PG, Chen L, Nardone J, Rao A (2003) Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev 17: 2205-2232.
  160. Chuvpilo S, Jankevics E, Tyrsin D, Akimzhanov A, Moroz D, et al. (2002) Autoregulation of NFATc1/A expression facilitates effector T cells to escape from rapid apoptosis. Immunity 16: 881-895.
  161. Sumpter TL, Payne KK, Wilkes DS (2008) Regulation of the NFAT pathway discriminates CD4+CD25+ regulatory T cells from CD4+CD25- helper T cells. J Leukoc Biol 83: 708-717.
  162. Baillie GS, Scott JD, Houslay MD (2005) Compartmentalisation of phosphodiesterases and protein kinase A: opposites attract. FEBS Lett 579: 3264-3270.
  163. Houslay MD, Baillie GS (2005) Beta-arrestin-recruited phosphodiesterase-4 desensitizes the AKAP79/PKA-mediated switching of beta2-adrenoceptor signalling to activation of ERK. Biochem Soc Trans 33: 1333-1336.
  164. Almahariq M, Mei FC, Wang H, Cao AT, Yao S, et al. (2015) Exchange protein directly activated by cAMP modulates regulatory T-cell-mediated immunosuppression. Biochem J 465: 295-303.
  165. Fuld S, Borland G, Yarwood SJ (2005) Elevation of cyclic AMP in Jurkat T-cells provokes distinct transcriptional responses through the protein kinase A (PKA) and exchange protein activated by cyclic AMP (EPAC) pathways. Exp Cell Res 309: 161-173.
  166. Tiwari S, Felekkis K, Moon EY, Flies A, Sherr DH, et al. (2004) Among circulating hematopoietic cells, B-CLL uniquely expresses functional EPAC, but EPAC1-mediated Rap1 activation does not account for PDE4 inhibitor-induced apoptosis. Blood 103: 2661-2667.
  167. Berruti G (2003) CAMP activates Rap1 in differentiating mouse male germ cells: a new signaling pathway mediated by the cAMP-activated exchange factor Epac? Cell Mol Biol (Noisy-le-grand) 49: 381-388.
  168. de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, Nijman SM, et al. (1998) Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature 396: 474-477.
  169. Hara S, Nakaseko C, Yamasaki S, Hattori M, Bos JL, et al. (2009) Involvement of Rap-1 activation and early termination of immune synapse in CTLA-4-mediated negative signal. Hematology 14: 150-158.
  170. Dillon TJ, Carey KD, Wetzel SA, Parker DC, Stork PJ (2005) Regulation of the small GTPase Rap1 and extracellular signal-regulated kinases by the costimulatory molecule CTLA-4. Mol Cell Biol 25: 4117-4128.
  171. Sansom DM, Walker LS (2006) The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev 212: 131-148.
  172. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, et al. (1995) Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3: 541-547.
  173. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, et al. (1995) Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270: 985-988.
  174. Vendetti S, Riccomi A, Sacchi A, Gatta L, Pioli C, et al. (2002) Cyclic adenosine 5'-monophosphate and calcium induce CD152 (CTLA-4) up-regulation in resting CD4+ T lymphocytes. J Immunol 169: 6231-6235.
  175. Finn PW, He H, Wang Y, Wang Z, Guan G, et al. (1997) Synergistic induction of CTLA-4 expression by costimulation with TCR plus CD28 signals mediated by increased transcription and messenger ribonucleic acid stability. J Immunol 158: 4074-4081.
  176. Krinzman SJ, De Sanctis GT, Cernadas M, Mark D, Wang Y, et al. (1996) Inhibition of T cell costimulation abrogates airway hyperresponsiveness in a murine model. J Clin Invest 98: 2693-2699.
  177. Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle NK, et al. (1991) CTLA-4 is a second receptor for the B cell activation antigen B7. J Exp Med 174: 561-569.
  178. Krummel MF, Allison JP (1996) CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J Exp Med 183: 2533-2540.
  179. Johnson KW, Davis BH, Smith KA (1988) cAMP antagonizes interleukin 2-promoted T-cell cycle progression at a discrete point in early G1. Proc Natl Acad Sci U S A 85: 6072-6076.
  180. Perkins D, Wang Z, Donovan C, He H, Mark D, et al. (1996) Regulation of CTLA-4 expression during T cell activation. J Immunol 156: 4154-4159.
  181. Bodor J, Fehervari Z, Diamond B, Sakaguchi S (2007) Regulatory T cell-mediated suppression: potential role of ICER. J Leukoc Biol 81: 161-167.
  182. Vendetti S, Patrizio M, Riccomi A, De Magistris MT (2006) Human CD4+ T lymphocytes with increased intracellular cAMP levels exert regulatory functions by releasing extracellular cAMP. J Leukoc Biol 80: 880-888.
  183. Lee YK, Turner H, Maynard CL, Oliver JR, Chen D, et al. (2009) Late developmental plasticity in the T helper 17 lineage. Immunity 30: 92-107.
  184. Lexberg MH, Taubner A, Förster A, Albrecht I, Richter A, et al. (2008) Th memory for interleukin-17 expression is stable in vivo. Eur J Immunol 38: 2654-2664.
  185. Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, et al. (2007) Phenotypic and functional features of human Th17 cells. J Exp Med 204: 1849-1861.
  186. Shi G, Cox CA, Vistica BP, Tan C, Wawrousek EF, et al. (2008) Phenotype switching by inflammation-inducing polarized Th17 cells, but not by Th1 cells. J Immunol 181: 7205-7213.
  187. Bending D, De la Peña H, Veldhoen M, Phillips JM, Uyttenhove C, et al. (2009) Highly purified Th17 cells from BDC2.5NOD mice convert into Th1-like cells in NOD/SCID recipient mice. J Clin Invest 119: 565-572.
  188. Martin-Orozco N, Chung Y, Chang SH, Wang YH, Dong C (2009) Th17 cells promote pancreatic inflammation but only induce diabetes efficiently in lymphopenic hosts after conversion into Th1 cells. Eur J Immunol 39: 216-224.
  189. Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, et al. (2008) Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity 29: 44-56.
  190. Xu L, Kitani A, Fuss I, Strober W (2007) Cutting edge: regulatory T cells induce CD4+CD25-Foxp3- T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-beta. J Immunol 178: 6725-6729.
  191. Osorio F, LeibundGut-Landmann S, Lochner M, Lahl K, Sparwasser T, et al. (2008) DC activated via dectin-1 convert Treg into IL-17 producers. Eur J Immunol 38: 3274-3281.
  192. Wei G, Wei L, Zhu J, Zang C, Hu-Li J, et al. (2009) Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 30: 155-167.
  193. Ayyoub M, Deknuydt F, Raimbaud I, Dousset C, Leveque L, et al. (2009) Human memory FOXP3+ Tregs secrete IL-17 ex vivo and constitutively express the T(H)17 lineage-specific transcription factor RORgamma t. Proc Natl Acad Sci U S A 106: 8635-8640.
  194. Goodnow CC, Sprent J, Fazekas de St Groth B, Vinuesa CG (2005) Cellular and genetic mechanisms of self tolerance and autoimmunity. Nature 435: 590-597.
  195. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155: 1151-1164.
  196. Piccirillo CA, Shevach EM (2004) Naturally-occurring CD4+CD25+ immunoregulatory T cells: central players in the arena of peripheral tolerance. Semin Immunol 16: 81-88.
  197. Faria AM, Weiner HL (2006) Oral tolerance and TGF-beta-producing cells. Inflamm Allergy Drug Targets 5: 179-190.
  198. Wan YY, Flavell RA (2006) The roles for cytokines in the generation and maintenance of regulatory T cells. Immunol Rev 212: 114-130.
  199. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, et al. (2001) The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 27: 20-21.
  200. Godfrey VL, Wilkinson JE, Russell LB (1991) X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am J Pathol 138: 1379-1387.
  201. Piccirillo, CA, Thornton AM (2004) Cornerstone of peripheral tolerance: naturally occurring CD4+CD25+ regulatory T cells. Trends Immunol 25: 374-380.
  202. Wang W, Shao S, Jiao Z, Guo M, Xu H, et al. (2012) The Th17/Treg imbalance and cytokine environment in peripheral blood of patients with rheumatoid arthritis. Rheumatol Int 32: 887-893.
  203. Niu Q, Cai B, Huang ZC, Shi YY, Wang LL (2012) Disturbed Th17/Treg balance in patients with rheumatoid arthritis. Rheumatol Int 32: 2731-2736.
  204. Kim JM, Rasmussen JP, Rudensky AY (2007) Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol 8: 191-197.
  205. Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T, et al. (2001) Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev 182: 18-32.
  206. Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6: 345-352.
  207. Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, et al. (2006) Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. J Immunol 177: 4376-4383.
  208. Field EH, Kulhankova K, Nasr ME (2007) Natural Tregs, CD4+CD25+ inhibitory hybridomas, and their cell contact dependent suppression. Immunol Res 39: 62-78.
  209. Nakamura K, Kitani A, Strober W (2001) Cell contact-dependent immunosuppression by CD4(+)CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med 194: 629-644.
  210. Patel M, Xu D, Kewin P, Choo-Kang B, McSharry C, et al. (2005) Glucocorticoid-induced TNFR family-related protein (GITR) activation exacerbates murine asthma and collagen-induced arthritis. Eur J Immunol 35: 3581-3590.
  211. Santucci L, Agostini M, Bruscoli S, Mencarelli A, Ronchetti S, et al. (2007) GITR modulates innate and adaptive mucosal immunity during the development of experimental colitis in mice. Gut 56: 52-60.
  212. Nocentini G, Riccardi C (2009) GITR: a modulator of immune response and inflammation. Adv Exp Med Biol 647: 156-173.
  213. Schaer DA, Cohen AD, Wolchok JD (2010) Anti-GITR antibodies--potential clinical applications for tumor immunotherapy. Curr Opin Investig Drugs 11: 1378-1386.
  214. Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, et al. (2010) Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS One 5: e10436.
  215. Cooles FA, Isaacs JD, Anderson AE (2013) Treg cells in rheumatoid arthritis: an update.Curr Rheumatol Rep 15: 352.
  216. Skapenko A, Leipe J, Lipsky PE, Schulze-Koops H (2005) The role of the T cell in autoimmune inflammation. Arthritis Res Ther 7: S4-14.
  217. Schulze-Koops H, Kalden JR (2001) The balance of Th1/Th2 cytokines in rheumatoid arthritis. Best Pract Res Clin Rheumatol 15: 677-691.
  218. Kusaba M, Honda J, Fukuda T, Oizumi K (1998) Analysis of type 1 and type 2 T cells in synovial fluid and peripheral blood of patients with rheumatoid arthritis. J Rheumatol 25: 1466-1471.
  219. Morita Y, Yamamura M, Kawashima M, Harada S, Tsuji K, et al. (1998) Flow cytometric single-cell analysis of cytokine production by CD4+ T cells in synovial tissue and peripheral blood from patients with rheumatoid arthritis. Arthritis Rheum4: 1669-1676.
  220. Nanki T, Lipsky PE (2000) Cytokine, activation marker, and chemokine receptor expression by individual CD4(+) memory T cells in rheumatoid arthritis synovium. Arthritis Res 2: 415-423.
  221. Yamada H, Nakashima Y, Okazaki K, Mawatari T, Fukushi JI, et al. (2008) Th1 but not Th17 cells predominate in the joints of patients with rheumatoid arthritis. Ann Rheum Dis 67: 1299-1304.
  222. Boissier MC, Chiocchia G, Bessis N, Hajnal J, Garotta G, et al. (1995) Biphasic effect of interferon-gamma in murine collagen-induced arthritis. Eur J Immunol 25: 1184-1190.
  223. Rosloniec EF, Latham K, Guedez YB (2002) Paradoxical roles of IFN-gamma in models of Th1-mediated autoimmunity. Arthritis Res 4: 333-336.
  224. Mauritz NJ, Holmdahl R, Jonsson R, Van der Meide PH, Scheynius A, et al. (1988) Treatment with gamma-interferon triggers the onset of collagen arthritis in mice. Arthritis Rheum 31: 1297-1304.
  225. Vermeire K, Heremans H, Vandeputte M, Huang S, Billiau A, et al. (1997) Accelerated collagen-induced arthritis in IFN-gamma receptor-deficient mice. J Immunol 158: 5507-5513.
  226. Frey O, Mitera T, Kelchtermans H, Schurgers E, Kamradt T, et al. (2011) Ameliorated course of glucose-6-phosphate isomerase (G6PI)-induced arthritis in IFN-γ receptor knockout mice exposes an arthritis-promoting role of IFN-γ. J Autoimmun 36: 161-169.
  227. Matthys P, Lories RJ, De Klerck B, Heremans H, Luyten FP, et al. (2003) Dependence on interferon-gamma for the spontaneous occurrence of arthritis in DBA/1 mice. Arthritis Rheum 48: 2983-2988.
  228. Kageyama Y, Koide Y, Yoshida A, Uchijima M, Arai T, et al. (1998) Reduced susceptibility to collagen-induced arthritis in mice deficient in IFN-gamma receptor. J Immunol 161: 1542-1548.
  229. Doodes PD, Cao Y, Hamel KM, Wang Y, Farkas B, et al. (2008) Development of proteoglycan-induced arthritis is independent of IL-17. J Immunol 181: 329-337.
  230. Doncarli A, Stasiuk LM, Fournier C, Abehsira-Amar O (1997) Conversion in vivo from an early dominant Th0/Th1 response to a Th2 phenotype during the development of collagen-induced arthritis. Eur J Immunol 27: 1451-1458.
  231. Okamoto Y, Gotoh Y, Tokui H, Mizuno A, Kobayashi Y, et al. (2000) Characterization of the cytokine network at a single cell level in mice with collagen-induced arthritis using a dual color ELISPOT assay. J Interferon Cytokine Res 20: 55-61.
  232. Wang D, Hill JA, Jevnikar AM, Cairns E, Bell DA (2002) Induction of transient arthritis by the adoptive transfer of a collagen II specific Th1 clone to HLA-DR4 (B1*0401) transgenic mice. J Autoimmun 19: 37-43.
  233. Doodes PD, Cao Y, Hamel KM, Wang Y, Rodeghero RL, et al. (2010) IFN-gamma regulates the requirement for IL-17 in proteoglycan-induced arthritis. J Immunol 184: 1552-1559.
  234. Straub RH (2014) Rheumatoid arthritis: Stress in RA: a trigger of proinflammatory pathways? Nat Rev Rheumatol 10: 516-518.
  235. McCray CJ, Agarwal SK (2011) Stress and autoimmunity. Immunol Allergy Clin North Am 31: 1-18.
  236. Satoh M, Chan EK, Ho LA, Rose KM, Parks CG, et al. (2012) Prevalence and sociodemographic correlates of antinuclear antibodies in the United States. Arthritis Rheum 64: 2319-2327.
  237. Nielen MM, van Schaardenburg D, Reesink HW, van de Stadt RJ, van der Horst-Bruinsma IE, et al. (2004) Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis Rheum 50:380–386.
  238. Rantapää-Dahlqvist S, de Jong BA, Berglin E, Hallmans G, Wadell G, et al. (2003) Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheum 48: 2741-2749.
  239. Majka DS, Deane KD, Parrish LA, Lazar AA, Baron AE, et al (2008) Duration of preclinical rheumatoid arthritis-related autoantibody positivity increases in subjects with older age at time of disease diagnosis. Ann Rheum Dis.67: 801-807.
  240. Jagannathan M, Hasturk H, Liang Y, Shin H, Hetzel JT, et al. (2009) TLR cross-talk specifically regulates cytokine production by B cells from chronic inflammatory disease patients. J Immunol 183: 7461-7470.
  241. Deane KD, O'Donnell CI, Hueber W, Majka DS, Lazar AA, et al. (2010) The number of elevated cytokines and chemokines in preclinical seropositive rheumatoid arthritis predicts time to diagnosis in an age-dependent manner. Arthritis Rheum 62: 3161-3172.
  242. Kokkonen H, Söderström I, Rocklöv J, Hallmans G, Lejon K, et al. (2010) Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis Rheum 62: 383-391.
  243. Marrack P, Kappler J, Kotzin BL (2001) Autoimmune disease: why and where it occurs. Nat Med 7: 899-905.
  244. Atanackovic D, Schnee B, Schuch G, Faltz C, Schulze J, et al. (2006) Acute psychological stress alerts the adaptive immune response: stress-induced mobilization of effector T cells. J Neuroimmunol 176:141-152.
  245. Xiang L, Del Ben KS, Rehm KE, Marshall GD Jr. (2012)Effects of acute stress-induced immunomodulation on TH1/TH2 cytokine and catecholamine receptor expression in human peripheral blood cells. Neuropsychobiology 65:12-19.
  246. Viswanathan K, Dhabhar FS (2005) Stress-induced enhancement of leukocyte trafficking into sites of surgery or immune activation. Proc Natl Acad Sci U S A 102: 5808-5813.
  247. Sandberg S, Paton JY, Ahola S, McCann DC, McGuinness D, et al. (2000) The role of acute and chronic stress in asthma attacks in children. Lancet 356: 982-987.
  248. Mohr DC, Hart SL, Julian L, Cox D, Pelletier D (2004) Association between stressful life events and exacerbation in multiple sclerosis: a meta-analysis. BMJ 328: 731.
  249. Wahle M, Krause A, Ulrichs T, Jonas D, von Wichert P, et al. (1999) Disease activity related catecholamine response of lymphocytes from patients with rheumatoid arthritis. Ann N Y Acad Sci 876: 287-296.
  250. Szelenyi J, Selmeczy Z, Brozik A, Medgyesi D, Magocsi M (2006) Beta-adrenergic modulation in the immune system. Stimulus-dependent effect of isoproterenol on MAPK activation and inflammatory mediator production in macrophages. Neurochem Int 49: 94-103.
  251. Wang WC, Mihlbachler KA, Brunnett AC, Liggett SB (2009) Targeted transgenesis reveals discrete attenuator functions of GRK and PKA in airway beta2-adrenergic receptor physiologic signaling. Proc Natl Acad Sci U S A106:15007-15012.
  252. Gerlo S, Kooijman R, Beck IM, Kolmus K, Spooren A, et al. (2011) Cyclic AMP: a selective modulator of NF-κB action. Cell Mol Life Sci 68: 3823-3841.
  253. Lefkowitz R, Goldstein J, Brown M (2012) A conversation with Robert Lefkowitz, Joseph Goldstein, and Michael Brown. Interview by Ushma S. Neil and Howard A. Rockman. J Clin Invest 122: 1586-1587.
  254. Shenoy SK, Drake MT, Nelson CD, Houtz DA, Xiao K, et al. (2006) Beta-arrestin-dependent, G protein-independent ERK1/2 activation by the beta2 adrenergic receptor. J Biol Chem 281: 1261-1273.
  255. Lefkowitz RJ, Shenoy SK (2005)Transduction of receptor signals by ß-arrestins. Science 308:512-517.
  256. Clark RB, Knoll BJ, Barber R (1999) Partial agonists and G protein-coupled receptor desensitization. Trends Pharmacol Sci 20: 279-286.
  257. Rockman HA, Koch WJ, Lefkowitz RJ (2002) Seven-transmembrane-spanning receptors and heart function. Nature 415: 206-212.
  258. Tran TM, Friedman J, Qunaibi E, Baameur F, Moore RH, et al. (2004) Characterization of agonist stimulation of cAMP-dependent protein kinase and G protein-coupled receptor kinase phosphorylation of the beta2-adrenergic receptor using phosphoserine-specific antibodies. Mol Pharmacol. 65:196-206.
  259. Hausdorff WP, Bouvier M, O'Dowd BF, Irons GP, Caron MG, et al. (1989) Phosphorylation sites on two domains of the beta 2-adrenergic receptor are involved in distinct pathways of receptor desensitization. J Biol Chem 264: 12657-12665.
  260. Lohse MJ, Benovic JL, Codina J, Caron MG, Lefkowitz RJ (1990) beta-Arrestin: a protein that regulates beta-adrenergic receptor function. Science 248: 1547-1550.
  261. Lefkowitz RJ (1998) G protein-coupled receptors. III. New roles for receptor kinases and beta-arrestins in receptor signaling and desensitization. J Biol Chem 273: 18677-18680.
  262. Freedman NJ, Lefkowitz RJ (1996) Desensitization of G protein-coupled receptors. Recent Prog Horm Res 51: 319-351.
  263. Krupnick JG, Benovic JL (1998) The role of receptor kinases and arrestins in G protein-coupled receptor regulation. Annu Rev Pharmacol Toxicol 38: 289-319.
  264. Goodman OB Jr, Krupnick JG, Santini F, Gurevich VV, Penn RB, et al. (1996) Beta-arrestin acts as a clathrin adaptor in endocytosis of the beta2-adrenergic receptor. Nature 383: 447-450.
  265. Laporte SA, Oakley RH, Zhang J, Holt JA, Ferguson SS, et al. (1999) The beta2-adrenergic receptor/betaarrestin complex recruits the clathrin adaptor AP-2 during endocytosis. Proc Natl Acad Sci U S A 96: 3712-3717.
  266. Luttrell LM, Roudabush FL, Choy EW, Miller WE, Field ME, et al. (2001) Activation and targeting of extracellular signal-regulated kinases by beta-arrestin scaffolds. Proc Natl Acad Sci U S A 98: 2449-2454.
  267. McDonald PH, Chow CW, Miller WE, Laporte SA, Field ME, et al. (2000) Beta-arrestin 2: a receptor-regulated MAPK scaffold for the activation of JNK3. Science 290: 1574-1577.
  268. Luttrell LM, Ferguson SS, Daaka Y, Miller WE, Maudsley S, et al. (1999) Beta-arrestin-dependent formation of beta2 adrenergic receptor-Src protein kinase complexes. Science 283: 655-661.
  269. Sun Y, Cheng Z, Ma L, Pei G (2002) Beta-arrestin2 is critically involved in CXCR4-mediated chemotaxis, and this is mediated by its enhancement of p38 MAPK activation. J Biol Chem 277: 49212-49219.
  270. Kim KS, Park DH, Wessel TC, Song B, Wagner JA, et al. (1993) A dual role for the cAMP-dependent protein kinase in tyrosine hydroxylase gene expression. Proc Natl Acad Sci U S A 90: 3471-3475.
  271. Trester-Zedlitz M, Burlingame A, Kobilka B, von Zastrow M (2005) Mass spectrometric analysis of agonist effects on posttranslational modifications of the beta-2 adrenoceptor in mammalian cells. Biochemistry 44: 6133-6143.
  272. Ferguson SS, Downey WE 3rd, Colapietro AM, Barak LS, Ménard L, et al. (1996) Role of beta-arrestin in mediating agonist-promoted G protein-coupled receptor internalization.Science 271: 363-366.
  273. Krause A, Steitz A, von Wichert P, Baerwald C (1995) Influence of cytokines on the density of beta 2-adrenergic receptors on peripheral blood mononuclear cells in vitro. Cytokine 7: 273-276.
  274. Korichneva IL, Tkachuk VA (1990) Alterations in beta-adrenoceptor density on T-lymphocytes upon activation with interleukin-2 and phytohaemagglutinin. Biomed Sci 1: 84-88.
  275. Nakane T, Szentendrei T, Stern L, Virmani M, Seely J, et al. (1990) Effects of IL-1 and cortisol on beta-adrenergic receptors, cell proliferation, and differentiation in cultured human A549 lung tumor cells. J Immunol 145: 260-266.
  276. Aderem A, Ulevitch RJ (2000) Toll-like receptors in the induction of the innate immune response. Nature 406: 782-787.
  277. Akira S, Takeda K (2004) Toll-like receptor signalling. Nat Rev Immunol 4: 499-511.
  278. Beutler B, Jiang Z, Georgel P, Crozat K, Croker B, et al. (2006) Genetic analysis of host resistance: Toll-like receptor signaling and immunity at large. Annu Rev Immunol 24: 353-389.
  279. Janeway CA Jr, Medzhitov R (2002) Innate immune recognition. Annu Rev Immunol 20: 197-216.
  280. Fan J, Malik AB (2003) Toll-like receptor-4 (TLR4) signaling augments chemokine-induced neutrophil migration by modulating cell surface expression of chemokine receptors. Nat Med 9: 315-321.
  281. Loniewski K, Shi Y, Pestka J, Parameswaran N (2008) Toll-like receptors differentially regulate GPCR kinases and arrestins in primary macrophages. Mol Immunol 45: 2312-2322.
  282. Heldwein KA, Fenton MJ (2002) The role of Toll-like receptors in immunity against mycobacterial infection. Microbes Infect 4: 937-944.
  283. Quesniaux V, Fremond C, Jacobs M, Parida S, Nicolle D, et al. (2004) Toll-like receptor pathways in the immune responses to mycobacteria. Microbes Infect 6: 946-959.
  284. Heldwein KA, Liang MD, Andresen TK, Thomas KE, Marty AM, et al. (2003) TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. J Leukoc Biol 74: 277-286.
  285. Reiling N, Hölscher C, Fehrenbach A, Kröger S, Kirschning CJ, et al. (2002) Cutting edge: Toll-like receptor (TLR)2- and TLR4-mediated pathogen recognition in resistance to airborne infection with Mycobacterium tuberculosis. J Immunol 169: 3480-3484.
  286. Shim TS, Turner OC, Orme IM (2003) Toll-like receptor 4 plays no role in susceptibility of mice to Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 83: 367-371.
  287. Abel B, Thieblemont N, Quesniaux VJ, Brown N, Mpagi J, et al. (2002) Toll-like receptor 4 expression is required to control chronic Mycobacterium tuberculosis infection in mice. J Immunol 169: 3155-3162.
  288. Brightbill HD, Libraty DH, Krutzik SR, Yang RB, Belisle JT, et al. (1999) Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science 285: 732-736.
  289. Means TK, Wang S, Lien E, Yoshimura A, Golenbock DT, et al. (1999) Human toll-like receptors mediate cellular activation by Mycobacterium tuberculosis. J Immunol 163: 3920-3927.
  290. Underhill DM, Ozinsky A, Smith KD, Aderem A (1999) Toll-like receptor-2 mediates mycobacteria-induced proinflammatory signaling in macrophages. Proc Natl Acad Sci U S A 96: 14459-14463.
  291. Means TK, Lien E, Yoshimura A, Wang S, Golenbock DT, et al. (1999) The CD14 ligands lipoarabinomannan and lipopolysaccharide differ in their requirement for Toll-like receptors.J Immunol 163: 6748-6755.
  292. Thoma-Uszynski S, Stenger S, Takeuchi O, Ochoa MT, Engele M, et al. (2001) Induction of direct antimicrobial activity through mammalian toll-like receptors. Science 291: 1544-1547.
  293. Buwitt-Beckmann U, Heine H, Wiesmüller KH, Jung G, Brock R, et al. (2005) Toll-like receptor 6-independent signaling by diacylated lipopeptides. Eur J Immunol 35: 282-289.
  294. Morr M, Takeuchi O, Akira S, Simon MM, Mühlradt PF (2002) Differential recognition of structural details of bacterial lipopeptides by toll-like receptors. Eur J Immunol 32: 3337-3347.
  295. Müller SD, Müller MR, Huber M, Esche Uv Uv, Kirschning CJ, et al. (2004) Triacyl-lipopentapeptide adjuvants: TLR2-dependent activation of macrophages and modulation of receptor-mediated cell activation by altering acyl-moieties. Int Immunopharmacol 4: 1287-1300.
  296. Okusawa T, Fujita M, Nakamura J, Into T, Yasuda M, et al. (2004) Relationship between structures and biological activities of mycoplasmal diacylated lipopeptides and their recognition by toll-like receptors 2 and 6. Infect Immun 72:1657-1665.
  297. Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY (2004) TLR2 is expressed on activated T cells as a costimulatory receptor. Proc Natl Acad Sci U S A 101: 3029-3034.
  298. Islam KN, Koch WJ (2012) Involvement of nuclear factor κB (NF-κB) signaling pathway in regulation of cardiac G protein-coupled receptor kinase 5 (GRK5) expression. J Biol Chem 287: 12771-12778.
  299. Kitas GD, Salmon M, Farr M, Gaston JS, Bacon PA (1988) Deficient interleukin 2 production in rheumatoid arthritis: association with active disease and systemic complications. Clin Exp Immunol 73: 242-249.
  300. Avramescu C, Vere CC, Margaritescu C, Turculeanu A, Balasoiu M, et al. (2005) Cytokinin panel in rheumatoid arthritis and correlation with histological patterns of synovitis -- active type of disease. Rom J Morphol Embryol 46: 87-92.
  301. Badou A, Savignac M, Moreau M, Leclerc C, Foucras G, et al. (2001) Weak TCR stimulation induces a calcium signal that triggers IL-4 synthesis, stronger TCR stimulation induces MAP kinases that control IFN-gamma production. Eur J Immunol 31: 2487-2496.
  302. Zhang S, Kaplan MH (2000) The p38 mitogen-activated protein kinase is required for IL-12-induced IFN-gamma expression. J Immunol 165: 1374-1380.
  303. Cantrell DA1 (2002) T-cell antigen receptor signal transduction. Immunology 105: 369-374.
  304. Dong C, Davis RJ, Flavell RA (2002) MAP kinases in the immune response. Annu Rev Immunol 20: 55-72.
  305. Huang Y, Wange RL (2004) T cell receptor signaling: beyond complex complexes. J Biol Chem 279: 28827-28830.
  306. Alberola-Ila J, Forbush KA, Seger R, Krebs EG, Perlmutter RM (1995) Selective requirement for MAP kinase activation in thymocyte differentiation. Nature 373: 620-623.
  307. Li X, Chaudry IH, Choudhry MA (2009) ERK and not p38 pathway is required for IL-12 restoration of T cell IL-2 and IFN-gamma in a rodent model of alcohol intoxication and burn injury. J Immunol 183: 3955-3962.
  308. Straub RH, Rauch L, Fassold A, Lowin T, Pongratz G (2012) Neuronally released sympathetic neurotransmitters stimulate splenic interferon-gamma secretion from T cells in early type II collagen-induced arthritis. Arthritis Rheum. 58:3450-3460. Erratum in: Arthritis Rheum 64:1695-1696.
  309. Koopman FA, Stoof SP, Straub RH, Van Maanen MA, Vervoordeldonk MJ, et al. (2011) Restoring the balance of the autonomic nervous system as an innovative approach to the treatment of rheumatoid arthritis. Mol Med 17: 937-948.
  310. Leden I, Eriksson A, Lilja B, Sturfelt G, Sundkvist G (1983) Autonomic nerve function in rheumatoid arthritis of varying severity. Scand J Rheumatol 12: 166-170.
  311. Perry F, Heller PH, Kamiya J, Levine JD (1989) Altered autonomic function in patients with arthritis or with chronic myofascial pain. Pain 39: 77-84.
Citation: Lorton D, Bellinger DL (2015) Sympathetic Nervous System Regulation of Th Cells in Autoimmunity: Beyond Th1 and Th2 Cell Balance. J Clin Cell Immunol 6:356.

Copyright: © 2015 Lorton D, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top