Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson’s disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity.
Significance Statement Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
I. Introduction
A. Overview
Dopamine, or 3-hydroxytyramine, is an endogenous catecholamine that is important to both neuronal and nonneuronal processes. Dopamine was first synthesized in 1910, and initial studies examined its biologic effect as a weak sympathomimetic,although the mechanism of action was not clear (Barger and Dale, 1910). Neurotransmitters, initially acetylcholine, were defined as the chemical agents mediating communication in nerve pulses by Drs. Otto Loewi and Henry Dale in the first decades of the 20th century (Valenstein, 2002). The catecholamines norepinephrine and dopamine were determined to be neurotransmitters in the middle of the 20th century, norepinephrine by Drs. Ulf von Euler, Bernard Katz, and Julius Axelrod and dopamine by Drs. Arvid Carlsson and Paul Greengard (Benes, 2001; Snyder, 2006; Iversen and Iversen, 2007).
The biosynthetic pathways associated with the synthesis of dopamine were first hypothesized by Hermann Blaschko (Blaschko, 1957), and with the discovery of dopamine in the peripheral tissues of mammals (Goodall, 1951; Von Euler and Hellner, 1951), it was shown to be a precursor of the catecholamines norepinephrine and epinephrine. Carlsson and colleagues found that dopamine has a unique distribution pattern throughout the brain, plasma, and other tissues within the human body (Carlsson et al., 1957, 1958; Weil-Malherbe and Bone, 1957; Bertler and Rosengren, 1959; Imai et al., 1970). Early reports identified the largest amounts of dopamine in the striatum, particularly the caudate nucleus. Studies using both dopamine and dopaminergic drugs found that dopamine could inhibit neuronal discharge and was critical for extrapyramidal function, particularly motor function (Hornykiewicz, 1966). In 1960, Ehringer and Hornykiewicz demonstrated dopamine deficits in patients with parkinsonism, indicating that extrapyramidal activity made dopamine a central factor in Parkinson’s disease (PD) (Ehringer and Hornykiewicz, 1960, 1998). Today, central nervous system (CNS) dopamine has a well-established role in motor control, cognition, learning, and reward. In the periphery, dopamine regulates gastrointestinal (GI) motility, sodium levels, blood pressure maintenance, hormone release, and many other functions (Baines and Drangova, 1997; Fitzgerald and Dinan, 2008; Harris and Zhang, 2012; Bove et al., 2019). Reductions in dopamine underlie diseases such as PD and attention-deficit/hyperactivity disorder (ADHD), while elevated dopamine states have been implicated in schizophrenia (Birtwistle and Baldwin, 1998).
In addition, research over the past few decades has shown that dopamine can have a substantial impact on immune cell function in both the CNS and periphery. Dopaminergic immunomodulation affects both innate and adaptive immunity and has become increasingly important as a possible target for drug discovery and disease management. While there have been numerous reviews that detail individual parts of these topics (Levite et al., 2017; Pinoli et al., 2017; Matt and Gaskill, 2020; Thomas Broome et al., 2020; Vidal and Pacheco, 2020), this field is progressing rapidly, and there is still considerable controversy regarding the understanding of dopamine as an immunoregulatory factor. This review aims to explore, organize, and consolidate what is known about the immunoregulatory actions of dopamine, from the regulation of specific immune functions to its role in disease pathogenesis, providing a firm foundation on which to move these types of studies forward. Of note, while all substance use disorders (SUDs) dysregulate the dopaminergic system, the direct effects of addictive substances on the immune system, is an overlapping but distinct topic from the role of dopamine itself. There is a growing interest in the bidirectional interaction(s) between the immune system and SUDs, and this topic has been extensively covered in other reviews (Cui et al., 2014; Lacagnina et al., 2017; Namba et al., 2021). Therefore, it is only discussed briefly in reference to other topics.
B. Dopamine Signaling
Dopamine primarily mediates its effects on different cell types by signaling through dopamine receptors, which are G-protein coupled receptors (GPCRs). An overview of these signaling cascades, delineating the pathways described in this section, is shown in Fig. 1. Paul Greengard and colleagues defined two distinct signaling pathways that activate or inhibit the ubiquitous second messenger 3′-5′ cAMP and are mediated by different types of dopamine receptors (Hemmings et al., 1984). Based on these pathways, the five dopamine receptor subtypes are often grouped into stimulatory D1-like dopamine receptors (D1 and D5) and inhibitory D2-like dopamine receptors (D2, D3, and D4) (Missale et al., 1998; Beaulieu and Gainetdinov, 2011). Dopamine receptors can also be grouped into low affinity (D1 and D2) and high affinity (D3, D4, and D5) receptors depending on their affinity for dopamine. The regulation of cAMP production is mediated by the release of heterotrimeric G proteins that are coupled to each dopamine receptor; D1-like receptors stimulate the production of cAMP, and D2-like receptors inhibit it.
Dopamine signaling through cognate receptors. Dopamine signaling is mediated through its GPCRs. D1-like receptors (D1 and D5, red) classically couple to Gas to mediate activation of adenylate cyclase, leading to cAMP production, PKA activation, and downstream activation of PKA targets. The D2-like receptors (D2, D3, and D4, blue) couple to the Gai pathway to inhibit adenylate cyclase production and oppose D1-like signaling. The D1-like receptors can also lead to activation of PLCβ, thus enhancing calcium flux and PKC activation. The D2-like receptors can also activate this pathway via Gbg. D2-like stimulation can additionally inhibit AKT phosphorylation through the formation of a b-arrestin/PP2A signaling complex. Both D1-like and D2-like stimulation leads to AKT phosphorylation through its activity on the phosphatidylinositol 3-kinase (PI3K)/Akt signaling axis, but the mechanisms behind this are not clear. Downstream, both receptors can activate members of the MAPK family. This occurs through various mechanisms including, but not limited to, cAMP activation, PKC and calcium signaling, and activation of the PI3K/Akt signaling cascade. Created with BioRender.com.
More specifically, dopamine binding to a dopamine receptor triggers a conformational shift that allows a guanine exchange factor to exchange a guanosine diphosphate for the guanosine triphosphate bound to the α subunit of the heterotrimeric G-protein. This releases the coupled G-protein and facilitates its dissociation into α and βγ subunits. The α subunit then acts on adenylate cyclase, an enzyme that catalyzes the conversion of ATP into cAMP. The specific effect on adenylate cyclase depends on the released G-protein. D1-like receptor activation releases Gs alpha subunits of the Gs heterotrimeric G protein (Gαs), which activate adenylate cyclase, increasing intracellular concentrations of cAMP (Kebabian and Greengard, 1971; Kebabian, 1978). Activation of both D1-like receptors increases production of cAMP in transfected cells, but some knockout studies suggest that D1 is more strongly linked to this response relative to D5 (Undie and Friedman, 1994; Undieh, 2010). In contrast to D1-like receptors, D2-like receptors are coupled to the inhibitory Gi alpha subunit of the Gi heterotrimeric G protein (Gαi). D2-like receptor activation inhibits the activity of adenylate cyclase, reducing cAMP production and decreasing the downstream activity induced by D1-like receptors (Boyd and Mailman, 2012).
cAMP primarily activates protein kinase A (PKA), but studies have shown that cAMP can also activate exchange factor directly activated by cAMP (EPAC) and protein kinase C (PKC). PKA activation leads to the transcription of cAMP response element binding protein (CREB), which triggers the transcription of a variety of genes (Beaulieu et al., 2015; Wang, Xu et al., 201). In neurons, dopamine and cAMP-regulated phosphoprotein is also a major target of PKA and is a critical mediator of dopamine signaling (Svenningsson et al., 2004), although the activity of dopamine and cAMP-regulated phosphoprotein in nonneuronal cells is poorly understood. PKA also modulates mitogen-activated protein kinase (MAPK) family activation, although this appears to be cell type–specific and may involve other downstream signaling effectors (Zhen et al., 1998; Skalhegg and Tasken, 2000; Han et al., 2007).
Although cAMP signaling is most associated with dopamine receptor activation, a growing number of studies have shown important effects of alternative GPCR-dependent and -independent pathways. Activation of both D1-like and D2-like receptors can activate protein lipase C-β (PLCβ) to induce inositol triphosphate (IP3)-mediated intracellular calcium flux. Unlike the effect on cAMP signaling, both dopamine receptor subtypes mediate similar effects on IP3-mediated calcium flux despite different signaling cascades (Beaulieu and Gainetdinov, 2011; Beaulieu et al., 2015). In this pathway, D1-like receptors couple to Gαq and stimulate PLCβ, leading to diacylglycerol and IP3 production, and the subsequent activation of Ca2+ and PKC (Jin et al., 1998; Wang et al., 1995; Jin et al., 2001; Zhang et al., 2009; Undieh, 2010; Medvedev et al., 2013). Knockout studies showed that dopamine activated PLCβ in Drdr1 knockout animals but not Drd5 knockout animals (Sahu et al., 2009), and earlier studies showed that only one D1-like isoform could modulate PLCβ activity (Friedman et al., 1997). This finding suggests that D5 may be the specific D1-like receptor that mediates Gαq activity and that the two D1-like receptors may mediate different effects, although this may be tissue-specific (Undieh, 2010). D2-like receptor activation can also stimulate PLCβ (Di Marzo et al., 1993; Hernandez-Lopez et al., 2000), acting through Gβγ subunits rather than Gαq subunits (Choi et al., 1999; Hernandez-Lopez et al., 2000; Beaulieu et al., 2015). Dopamine-mediated PLCβ activation has been observed in a number of cell types, including striatal neurons (Friedman et al., 1997; Sahu et al., 2009), renal proximal tubule cells (Felder et al., 1989; Vyas et al., 1992), and immune cells (Nickoloff-Bybel et al., 2019), suggesting that this effect may be central to many of the actions of dopamine.
Dopamine signaling can also be mediated through G-protein-independent pathways, such as the activation of β-arrestins. Classically, these proteins regulate receptor internalization but have also been shown to trigger specific signaling activities (Lefkowitz and Shenoy, 2005; DeWire et al., 2007; Defea, 2008; Jean-Charles et al., 2017). In addition, at higher concentrations, dopamine can activate α- and β-adrenergic receptors (Lei, 2014), triggering a distinct but often overlapping set of downstream pathways. Dopamine-induced β-arrestin signaling is primarily mediated by D2-like receptors (Beaulieu et al., 2005, 2015; Peterson et al., 2015) which recruit protein phosphatase 2A (PP2A) and cause the subsequent inhibition of protein kinase B (Akt) (Beaulieu et al., 2004; Radl et al., 2013; Zhang, Jiang et al., 2016; Han et al., 2017; Wu et al., 2020). These and other data indicate that the view of D1-like receptors as stimulatory (Gαs) and D2-like receptors as inhibitory (Gαi) is only partially accurate, as D1- and D2-like receptors can stimulate both opposing and overlapping signaling mechanisms through non-cAMP pathways.
It is challenging to define all the dopamine-driven signaling cascades that are downstream of cAMP, Ca2+, and PLCβ, as dopamine receptors can activate a wide range of additional proteins and effectors, including the MAPKs extracellular signal-regulated kinase 1/2 (ERK1/2), p38 MAPK, and c-Jun N-terminal kinase (JNK)/stress-activated protein kinase, L-type calcium channels, Akt, and AMP-activated protein kinase (Welsh et al., 1998; Zhen et al., 1998; Cussac et al., 1999; Yan et al., 1999; Zhen et al., 2001; Brami-Cherrier et al., 2002; Nair et al., 2003; Nair and Sealfon, 2003; Wang et al., 2005; Ming et al., 2006; Han et al., 2007; Liu et al., 2009; Mannoury la Cour et al., 2011; Chen, Ruan et al., 2012; Perreault et al., 2013; Yoon and Baik, 2013; Franz et al., 2015; Bone et al., 2017; Fan et al., 2018). The precise signaling pathways involved in the activation of these effectors are complicated, often overlapping, and vary by cell type. For example, both D1- and D2-like receptors have been shown to activate all three members of the MAPK family: p38 MAPK, JNK, and ERK1/2; however, the specific MAPKs involved and the distinct roles of dopamine receptors vary from system to system. In mouse embryonic stem cells, dopamine, the D1-like agonist SKF-38393, and the D2-like agonist quinpirole activated all three members of the MAPK family (Lee et al., 2006). However, in neuroblastoma cells, D1-like agonists only activated p38 MAPK and JNK, and no MAPKs were activated by quinpirole (Zhen et al., 1998). Moreover, in CD4+ T-cells, D5 stimulation activates ERK1/2, while D3 stimulation was linked to inhibition of this protein (Franz et al., 2015).
Similarly, dopamine may have varying impacts on Akt activation in different systems. Dopamine both positively and negatively regulates Akt (Zhen et al., 2001; Brami-Cherrier et al., 2002; Nair et al., 2003; Nair and Sealfon, 2003; Beaulieu and Gainetdinov, 2011; Mannoury la Cour et al., 2011; Chen, Ruan et al., 2012; Perreault et al., 2013; Radl et al., 2013; Mirones et al., 2014; Beaulieu et al., 2015; Tolstanova et al., 2015; Zhang, Jiang et al., 2016; Gao et al., 2017; Han et al., 2017; Wu et al., 2020; Yan et al., 2020), although D2-like receptors are most commonly associated with Akt inhibition (Beaulieu et al., 2007, 2015; Beaulieu and Gainetdinov, 2011; Radl et al., 2013; Tolstanova et al., 2015; Zhang, Jiang et al., 2016; Han et al., 2017). Akt inhibition is likely mediated by β-arrestin-induced recruitment of PP2A, which inhibits the phosphorylation and activation of Akt (Beaulieu et al., 2005, 2007, 2015; Beaulieu and Gainetdinov, 2011).
A substantial amount of dopaminergic immunomodulation can be mediated by MAPK and Akt activity, as these proteins modulate many immune activities. MAPKs can regulate functions such as cytokine/chemokine production and phagocytosis (Karin, 1995; Cuenda and Rousseau, 2007; Kaminska et al., 2009; Cargnello and Roux, 2011; Kyriakis and Avruch, 2012), while Akt inhibition may mediate some of the anti-inflammatory effects of dopamine receptors (Zhang, Jiang et al., 2016; Han et al., 2017; Wu et al., 2020). Taken together, these data highlight the complexity of dopamine signaling and indicate that cell type–specific variations in dopamine receptor responses to the same ligands likely account for the diverse effects of dopamine on the functions of different immune cells.
C. Dopamine Production and Metabolism
The synthesis of dopamine has been most widely studied in dopaminergic neurons in the CNS, in which dopamine is synthesized from tyrosine. An overview of the dopamine synthesis and degradation pathways described in this section is found in Fig. 2. Tyrosine is produced from phenylalanine in the liver through the action of phenylalanine hydroxylase and is transported into dopaminergic neurons after crossing the blood–brain barrier through the large neutral amino acid transporter. In dopaminergic neurons, tyrosine hydroxylase (TH) catalyzes the addition of a hydroxyl group to the meta position of tyrosine to produce levodopa (L-DOPA) (Meiser et al., 2013). This process is considered the rate limiting step in this pathway and is susceptible to end-product inhibition by high levels of catecholamines. However, it is much less affected by changes in tyrosine levels, as TH is typically saturated with substrate. Once produced, L-DOPA is rapidly converted to dopamine by aromatic L-amino acid decarboxylase (AADC) (Meiser et al., 2013). In dopaminergic neurons, newly synthesized dopamine is transported from the cytoplasm into synaptic vesicles through vesicular monoamine transporter 2 (VMAT2). In adrenergic neurons containing dopamine-β-hydroxylase, dopamine can be further hydrolyzed to generate norepinephrine (Hoffman et al., 1998). Once produced and packaged into vesicles, dopamine can be released when the vesicles fuse with the cell membrane to release the packaged dopamine.
Metabolic pathway of dopamine biosynthesis and degradation. Dopamine synthesis is initiated with the hydroxylation of tyrosine by the enzyme TH to generate L-DOPA. L-DOPA is converted to dopamine by AADC. Dopamine beta hydroxylase (DBH) hydroxylates dopamine to form norepinephrine, which is converted to epinephrine by phenylethanolamine-N-methyltransferase (PNMT). Dopamine is primarily metabolized by two enzymatic pathways, COMT and MAO. COMT converts dopamine to 3-methoxytyramine, which is subsequently converted to 3-methoxy-4-hydroxyacetaldehyde by MAO. In contrast, MAO converts dopamine to 3,4-dihydroxyphenylacetaldehyde, which is then converted by aldehyde dehydrogenase (ALDH) to DOPAC. In the final steps of dopamine degradation, ALDH and COMT convert 3-methoxy-4-hydroxyacetaldehyde and DOPAC to HVA, respectively. Created with BioRender.com
Extracellular dopamine mediates its effects by binding to dopamine receptors and can be recycled via active transport back into dopaminergic neurons by monoamine transporters. The primary transporter that mediates dopamine uptake is the dopamine transporter (DAT), but in brain regions with low DAT expression, dopamine is taken up by the norepinephrine transporter (Moron et al., 2002). Once dopamine is taken back up, it can be repackaged into VMAT2-expressing vesicles, which are then re-released, recycling the neurotransmitter. Alternatively, in both neurons and other cell types, dopamine can be metabolized to form homovanillic acid (HVA). The two major degradation pathways that generate HVA begin with oxidative deamination by monoamine oxidases (MAOs), or O-methylation by catechol-O-methyltransferase (COMT). These pathways generate a variety of intermediate metabolites, including dihydroxyphenylacetaldehyde, 3,4 dihydroxyphenylacetic acid (DOPAC), and 3-methoxytyramine, which are ultimately metabolized to HVA (Kopin, 1985; Thomas Broome et al., 2020). These regulatory pathways are often the targets of therapeutic drugs prescribed for diseases mediated by dopaminergic dysregulation, such as neuropsychiatric disorders (Laatikainen et al., 2012; Finberg, 2019). Importantly, the processes described here apply mainly to the production and metabolism of dopamine in the CNS as mediated by dopaminergic neurons. As discussed in the following text, many immune cells also express these proteins, and data indicate that these processes occur in several immune cell types. However, the processes regulating dopamine synthesis and metabolism in immune cells, how they play into the function of these cells, and the role of dopamine in disease processes remain poorly understood.
D. Dopamine Release and Uptake
Classically, dopaminergic neurons release dopamine when an action potential reaches the axon terminal, inducing dopamine-containing vesicles to fuse with the cell membrane and release dopamine via exocytosis (Liu and Kaeser, 2019). Within dopaminergic neurons, two temporally distinct modes of signaling are traditionally discussed: phasic and tonic firing. Phasic firing involves synchronized burst firing, which results in fast and transient dopamine release (seconds), while tonic firing involves asynchronous spontaneous firing that produces slow and widespread dopamine release (minutes to hours) (Hauber, 2010). The amount of dopamine that is released in both the CNS and in the periphery depends on the region or tissue being examined and the stimulus involved, but the precise amounts in humans remain largely unclear. However, studies in rodents suggest that tonic firing releases dopamine in the nanomolar range while phasic firing increases the released dopamine concentrations up to micromolar levels (Matt and Gaskill, 2020).
Extracellular dopamine mediates communication by binding to dopamine receptors on neighboring cells. Classic neurotransmission refers to dopamine receptors on the postsynaptic neuron and point-to-point transfer of dopamine in the synaptic cleft. This type of direct interaction within well-defined physical boundaries, such as axons and their synapses, represents a discrete type of transmission known as wired transmission. This type of communication was long considered to be the primary form of dopaminergic neurotransmission (Agnati et al., 2010). However, more precise mapping of neurotransmitter location has shown that a slower, less directed form of communication known as volume transmission also plays a substantial role in dopaminergic neurotransmission. Volume transmission is characterized by extrasynaptic dopamine receptor activation via dopamine spillover or by nonsynaptic release of dopamine into the extracellular space (Venton et al., 2003; Fuxe et al., 2015; Borroto-Escuela et al., 2018). Recent data suggest that dopamine spillover from the synapse plays a minor role in this process, suggesting that volume transmission is largely due to focused release and the diffusion of dopamine at extrasynaptic sites (Wiencke et al., 2020).
Because dopaminergic neurons have broad arbors, dopamine released via spillover and extrasynaptic release increase dopamine concentrations in a large area of surrounding tissue. This can activate extrasynaptic dopamine receptors on neighboring neurons, as well as other nearby cells, such as immune cells, that may be a considerable distance away from the synapse (Rice and Cragg, 2008; Rice et al., 2011). Because increased concentrations of dopamine lead to the diffusion of dopamine throughout a greater area of CNS tissue, the area and number of cells exposed to dopamine is directly proportional to the amount of dopamine released. However, the rates and distances of diffusion can vary widely across the CNS, depending on the concentration and volume of dopamine release, DAT uptake dynamics, the regional specificity of the dopamine receptors, and the volume of extracellular fluid (Sulzer et al., 2016; Matt and Gaskill, 2020).
The function of DAT is particularly important for the regulation of dopaminergic signaling, as this transporter is a critical regulator of extracellular dopamine concentrations. DAT dysregulation or neuronal death/dysfunction that reduces the amount of functional DAT in a specific area could expose larger brain areas and the immune cells in those regions to increased concentrations of dopamine. Indeed, DAT dysregulation is implicated in several distinct pathologies, including ADHD, PD, and bipolar disorder (Vaughan and Foster, 2013; Bastos et al., 2018; Mackie et al., 2018). Most studies that model DAT regulation of dopamine concentrations focus on neuronal DAT, but DAT is also expressed and active on numerous cell types, including glia (Schomig et al., 1998; Meng et al., 1999; Takeda et al., 2002) and many types of immune cells (McKenna et al., 2002; Meredith et al., 2005; Mackie et al., 2018, 2022). The role of immune cell DAT, although not well understood, will be further discussed in subsequent sections.
E. Dopamine and Oxidative Stress
The induction of oxidative stress is another important mechanism by which dopamine drives several disparate effects that are primarily associated with the development of pathologic conditions. Oxidative damage associated with dopamine includes dopamine auto-oxidation, α-synuclein aggregation, glial cell activation, alterations in calcium signaling, mitochondrial dysfunction, and excess free iron (Juarez Olguin et al., 2016). Dopamine and its metabolites contain 2-hydroxyl residues, which generate highly reactive dopamine and DOPA quinones (Miyazaki and Asanuma, 2008; Meiser et al., 2013). The production of these highly reactive quinones not only can be the result of enzymatic oxidation by metal catalysis (Fe3+) or by cyclooxygenases, tyrosinases, or other enzymes but can also occur spontaneously. These reactions can then generate superoxide radical anions by donating electrons to oxygen (Graham et al., 1978; Stokes et al., 1999; Sulzer and Zecca, 2000; Meiser et al., 2013). In the presence of iron, dopamine quinones can also further react to form 6-hydroxydopamine, which is a neurotoxin (Simola et al., 2007). These products [dopamine quinones, 6-hydroxydopamine and reactive oxygen species (ROS) such as O2-] can be particularly harmful to cells by reacting nonspecifically, causing irreversible cell damage and apoptosis. The interplay of oxidative stress and neuroinflammation is a major factor in the impact of dopamine on immune function and has been shown to synergistically promote the progression of neurodegenerative diseases such as PD and Alzheimer’s disease (Asanuma et al., 2003; Jiang, Sun et al., 2016).
F. Dopaminergic Pathways in the CNS
The dopaminergic neurons that make up the nigrostriatal, mesolimbic, and mesocortical pathways are largely present in midbrain regions, specifically the substantia nigra (SbN), ventral tegmental area (VTA), and arcuate nucleus of the hypothalamus (Scarr et al., 2013; Nolan and Gaskill, 2019). Their efferent projections reach the striatum, nucleus accumbens (NAc), and several regions in the cortex, respectively. Classic slow-firing properties are seen in dopaminergic neurons that project to the dorsal striatum and NAc shell, while fast-firing properties are seen in dopaminergic neurons in the medial VTA that project to the amygdala or NAc core (Lammel et al., 2008; Hauber, 2010). Dopamine concentrations in the nigrostriatal, mesolimbic, and mesocortical pathways range from 10−8M to 10−5M. The microglia and macrophages in these pathways are in relatively close proximity to neurons and could be exposed to these dopamine concentrations during homeostatic function. A more in-depth discussion of the concentrations of dopamine to which CNS immune cells may be exposed can be found in the recent review by Matt and Gaskill (2020).
Within the tuberoinfundibular pathway, which is the fourth major dopaminergic pathway in the CNS, neurons project to the median eminence and are primarily responsible for the regulation of prolactin (Gudelsky, 1981; Scarr et al., 2013). Unlike the majority of dopaminergic neurons, most tuberoinfundibular dopaminergic neurons are categorized as secretory neurons due to their lack of synaptic contacts (Ben-Jonathan and Hnasko, 2001). Dopamine released by tuberoinfundibular dopaminergic neurons can diffuse through the perivascular space and is transported to the pituitary gland by portal blood. Moreover, in this pathway, continuously high exposure to dopamine signals via D2 receptors to suppress the activity of pituitary lactotrophs by inhibiting prolactin through the control of calcium flux (Ben-Jonathan and Hnasko, 2001). In the pituitary, studies have identified macrophages (Fujiwara et al., 2017) and dendritic cells (DCs) (Glennon et al., 2015) that could play a role in communicating immune activation to the hypothalamic–pituitary–adrenal (HPA) axis. Dopamine concentrations in the tuberoinfundibular pathway range from 10−8M to 10−4M in both the hypothalamus and pituitary, and immune cells could be exposed to significant dopamine fluctuations due to diet (Volkow et al., 2011) or the regulation of prolactin production (Lyons et al., 2012).
G. Peripheral Dopamine
The role of dopamine in the periphery was first described in 1972 in the renal and cardiovascular systems (Goldberg, 1972). Since then, peripheral dopamine has been shown to influence many critical functions in the periphery, such as blood pressure, GI motility, respiration, and immune activation (Goldstein et al., 1995; Rubí and Maechler, 2010; Grassi and Ram, 2016; Matt and Gaskill, 2020). Peripheral and central nervous systems share some mechanisms and molecular machinery, but studies suggest that peripheral dopaminergic systems act via pathways distinct from those in the CNS. There are substantial genetic and expression level differences in dopaminergic proteins in peripheral cells relative to CNS cells, as well as different dopamine release and uptake mechanisms (Wong et al., 1995; Myohanen et al., 2010; Zeng and Jose, 2011). The concentrations of dopamine in peripheral regions vary and were previously believed to originate from the nervous system via crosstalk or from mesenteric regions. It is now clear that while dopamine is released from these regions, it can also be generated in other cell types and peripheral organs. For example, in the kidney or adrenal medulla, dopamine can act as an autocrine/paracrine regulator of local organ function (Eisenhofer et al., 1997; Matt and Gaskill, 2020).
Changes in plasma dopamine are partially determined by sympathetic nerve activity, as increased peripheral dopamine is associated with increases in sympathetic activity, and patients with a loss of sympathetic nerve function have low plasma dopamine concentrations (Goldstein and Holmes, 2008). Sympathetic nerves release dopamine through vesicles that release both dopamine and norepinephrine via exocytosis (Goldstein and Holmes, 2008). Dopamine is also directly released into the circulation from chromaffin cells in the adrenal medulla, as well as amine precursor uptake and decarboxylation cells, which are found predominantly in the kidney (Wolfovitz et al., 1993). The high levels of plasma dopamine are not well understood, but one possible explanation is an underestimation of the amount of dopamine synthesis and metabolism that occurs in nonneuronal cells in the periphery. This notion is supported by high urinary excretion of DOPAC and HVA, as well as very high concentrations of dopamine conjugates in the periphery (Eisenhofer et al., 1997). Free dopamine levels in the circulation are in the picomolar to femtomolar range and only make up 5% of dopamine in plasma (Kuchel and Kuchel, 1991). Most peripheral dopamine is conjugated as sulfates or glucuronides, which are biologically inactive (Yoneda et al., 1983). There are also substantial amounts of both types of dopamine conjugates in the CNS (Suominen et al., 2013), again exceeding the concentrations of free dopamine in these regions. Data suggest that the source of these dopamine conjugates may be the adrenal gland (Wang et al., 1983; Uutela et al., 2009).
Dopamine sulfate has a half-life of a few hours compared with a few minutes for unmodified dopamine (Eldrup, 2004). The sulfo-conjugation mechanism seems relatively independent of sympathetic nerves, as the loss of sympathetic nerve function does not decrease plasma levels of dopamine sulfate. However, ingestion of a standard meal increases plasma dopamine sulfate concentrations by more than 50-fold, suggesting that sulfo-conjugation may depend on diet and dopamine conjugation in the GI tract (Goldstein et al., 1999). Dopamine sulfate conjugation is hypothesized to take place before dopamine enters the bloodstream, and very little dopamine sulfate is formed from circulating dopamine. This mechanism may localize the effects of bioactive dopamine due to diet or local dopamine produced by peripheral tissues, and/or it may be used to inactivate dopamine when it enters circulation to prevent toxicity and catecholamine buildup (Goldstein et al., 1999). Inactivating circulating dopamine may be important as variations in the ability to sulfo-conjugate dopamine increases the risk of certain diseases (Suominen et al., 2015).
Unlike dopamine inactivation by deamination or O-methylation, sulfo-conjugation is reversible. Dopamine sulfate can be converted back to bioactive dopamine by the enzyme arylsulfatase A (Strobel et al., 1990), which is found in the liver, lung, brain, and adipose tissue (Richard et al., 2001; Borcherding et al., 2011). Small amounts of dopamine sulfate can cross the blood–brain barrier (Suominen et al., 2015), and the levels of the UDP-glucuronosyltransferases and phenol sulfotransferases (needed for dopamine conjugation have been reported in rat and human brain (King et al., 1999; Kauffman, 2004). It has also been shown that dopamine can induce its own sulfation metabolism and that inhibiting the sulfotransferase SULT1A3 significantly increased the susceptibility of cells to dopamine toxicity (Sidharthan et al., 2013). This suggests that sulfation is a mechanism to protect cells from damage and could be involved in neurodegenerative pathology. However, so far this has only been studied in vitro with SK-N-MC and Neuro2A cells (Sidharthan et al., 2013).
Levels of glucuronidated dopamine vary with sympathetic input to the periphery (Claustre et al., 1983; Alexander et al., 1984), although the mechanisms underlying this variability is unclear. Glucuronidation is common in the gut, and while human cells do not express glucuronidase enzymes, many gut bacteria express β-glucuronidase that can reverse this process (Pellock and Redinbo, 2017). In the gut lumen of mice, levels of free and glucuronidated dopamine are regulated by the activity of gut microbiota, such as Clostridium, with high levels of β-glucuronidase activity (Asano et al., 2012). More broadly, numerous bacterial species in the human gut can release catecholamines (Kim and Shin, 2018; Xue, Zhang et al., 2018; González-Arancibia et al., 2019), and dietary changes that alter the gut microbiome directly influence neurotransmitter levels in the brain and gut (Guo et al., 2021). Further, changes in gut microbial composition that reduced TH expression in the gut exacerbated invariant natural killer (NK) T-cell–mediated hepatitis (Xue, Zhang et al., 2018). Taken together, these data indicate that the microbiome plays an important role in the regulation of peripheral dopamine levels, potentially by modulating levels of conjugated dopamine. More research is needed to define how both sulfation and glucuronidation can impact peripheral dopamine levels, but these studies can be challenging as there are large differences in the extent of conjugation between different species (Claustre et al., 1983; Eisenhofer et al., 1997). Still, the active production, degradation, conjugation, and excretion of dopamine indicate an active peripheral dopaminergic system, but the importance of peripheral regulatory mechanisms, and indeed many of the activities of peripheral dopamine, remains understudied.
II. Considerations and Caveats Regarding the Study of Dopaminergic Immunology
A. Interpreting In Vivo Measurements of Dopamine
Despite an increasing focus on the immunomodulatory effects of dopamine, there is still a substantial gap in our knowledge regarding the specific impact of this neurotransmitter on different types of immune cells and diseases. Determining specific dopamine concentrations to use in vitro can be difficult, as dopamine concentrations in specific organ systems or to which immune cells could be exposed are not well defined. In animal models, studies of CNS dopamine levels are more common, particularly in rodents, and a number of studies have examined peripheral dopamine levels in animal systems, although to a much lesser extent than the brain. Direct access to a living human brain is limited, and although we can examine DAT and dopamine receptor densities, we are still developing the tools needed to define dopamine levels by neuroimaging in humans. While it is possible to directly measure peripheral dopamine levels, there has been relatively little interest in studying peripheral dopamine, so studies that make these determinations are scarce. Our recent review discusses these data in detail, aggregating studies to present the general ranges of dopamine levels throughout much of the CNS and periphery (Matt and Gaskill, 2020).
However, these ranges are relatively wide in most organs, with considerable differences within each region. A substantial amount of this variation is likely due to differences in populations, species, or environment between the different studies. However, some differences may also be due to specific technical issues that should be carefully considered when assessing dopamine concentrations. Measuring dopamine in postmortem tissue requires careful consideration of the isolation procedures used, as exposure to oxygen, metal ions, or specific enzymes can induce oxidation of the catechol ring and result in the production of dopamine quinones (Graham et al., 1978; Stokes et al., 1999; Sulzer and Zecca, 2000). This could reduce the amount of detectable dopamine present in the sample and potentially underestimate dopamine levels. Oxidation can also affect dopamine metabolites and may be more pronounced in certain regions, generating artificial distortions in dopamine:dopamine metabolite ratios and regional differences in dopamine levels and metabolism (Sloviter and Connor, 1977; Graham et al., 1978; Spokes and Koch, 1978; Kontur et al., 1994; Sun et al., 2018). Rapid tissue processing, processing under anoxic conditions (Morelli et al., 2011; Emanuel et al., 2022), and isolation and storage at colder temperatures can prevent oxidation and may provide a more accurate measurement of tissue dopamine levels.
Other technical considerations include interference by molecules with similar oxidation potential, and the sensitivity and specificity of the neurochemical techniques used for analysis, as this can have significant effects on the assessed dopamine concentration. Many of the methods from which our current understanding of anatomic dopamine levels has come (high-performance liquid chromatography, microdialysis or fast scan cyclic voltammetry) have considerable associated errors (McLaurin et al., 2021). More precise and potentially higher levels of dopamine (Patriarchi et al., 2018) will likely be measured as more studies use newer electrochemical detection techniques that are enhanced with aptamers or molecularly imprinted polymers and enzyme-, aptamer-, and antibody-based biosensors, as well as more sensitive in vivo tools including fiber photometry and genetic dopamine sensors such as dLight (Patriarchi et al., 2018; Leopold et al., 2019; Sabatini and Tian, 2020; Labouesse and Patriarchi, 2021; McLaurin et al., 2021).
Consideration must also be given to the relationship between the amount of dopamine measured in a particular tissue and the amount of dopamine to which immune cells in that compartment are exposed. In the brain and likely other compartments, biogenic amines and their metabolites are generally found in three regions: the extracellular space, cytoplasmic vesicles, and the cytoplasm itself (Best et al., 2009, 2010). Postmortem tissue collection and analysis limits the measurement of dopamine to that found within cytosolic and vesicular pools but not the extracellular pool as this is lost in processing, and most in vivo studies examine dopamine levels in the brain in close proximity to neurons. Further, the overall dopaminergic tone in a particular tissue does not account for microenvironments. These are created because dopamine levels throughout tissues are dynamic and based on the density and activity of cells that produce, take up, and degrade dopamine in that compartment (Kawagoe et al., 1992; Wightman et al., 2007). In many studies focusing on changes in dopamine in the context of behavior or neurotransmission, these concerns should be noted, but they are not likely to change the results, as it is the amount of change rather than baseline dopamine concentrations that are critical. However, in vitro studies or studies that expose immune cells to a specific amount of dopamine should consider these issues, as the amount of dopamine to which immune cells respond in vivo may only be approximated by the range determined for that tissue.
B. In Vitro Dopamine Concentrations
To account for uncertainty in the physiologically relevant concentrations of dopamine, in vitro examinations of dopaminergic immunomodulation should use dopamine concentrations that approximate dopamine levels to which the immune cell being studied could be exposed. In most tissues, this range is likely to be approximately 10−5M to 10−11M, although the utility of higher and lower levels of dopamine should be determined based on the experimental question. While the higher end of dopamine concentrations are >10−5M in some brain regions and peripheral compartments such as the adrenal gland, gut, and carotid body, most immune cells will generally encounter lower concentrations of dopamine during homeostatic conditions (Matt and Gaskill, 2020). In addition, regulation of dopamine levels is likely to be disrupted by a number of diseases, as well as by many therapeutics, particularly neuropsychiatric drugs (Matt and Gaskill, 2019).
Of particular note, the dopamine levels encountered by immune cells in the CNS are very likely to be increased in the context of SUDs, as all addictive substances acutely increase CNS dopamine in the mesocorticolimbic system, as well as other brain regions (Di Chiara and Imperato, 1988; Pierce and Kumaresan, 2006; Volkow, Fowler et al., 2009). Data suggest that methamphetamine generally induces the greatest increase, resulting in concentrations of approximately 1 to 5 × 10−5M (Matt and Gaskill, 2020). The effects of neuropsychiatric and addictive drugs on peripheral dopamine are not clear and need further study, although whole-body positron emission tomography (PET) scanning in mice showed that cocaine, ketamine, and methamphetamine altered dopamine concentrations in several peripheral organs (Yeh et al., 2014). The major effect of exogenous increases in dopamine is likely to be an increase in the number of immune cells exposed to lower dopamine levels, as increased dopamine release expands the area of tissue exposed to dopamine, and lower concentrations cover a larger area and contact more cells (Peters and Michael, 2000; Venton et al., 2003; Spuhler and Hauri, 2013). Thus, even studying the immunoregulatory effects of stimulant-induced dopamine levels does not necessarily require the use of dopamine levels much higher than 10−5M.
The concern regarding the use of extraphysiologic dopamine concentrations (>10−5M) is that it may create confounding results by initiating immune functions that do not occur in vivo. One mechanism by which this could occur is through the activation of lower affinity dopamine receptors that would not be activated when an immune cell encounters lower dopamine levels. Different dopamine receptors, as well as nondopamine receptors, also have discrete affinities for dopamine (Richtand, 2006; Beaulieu and Gainetdinov, 2011). Thus, lower dopamine levels may activate high affinity receptors (D3, D4, and D5) to drive one function, while high dopamine levels could activate both the low- (D1 and D2) and high-affinity dopamine receptors, potentially activating other distinct functions through multiple receptors. For example, dopamine concentrations from 10−10M to 10−6M dose-dependently decrease glucagon secretion by human pancreatic islet cells, but 10−4 and 10−5M dopamine increases glucagon release (Aslanoglou et al., 2021). In splenic macrophages from wall lizards, lower dopamine levels (10−11–10−15M) increased phagocytosis, while higher dopamine levels (10−7–10−5M) decreased phagocytosis (Roy and Rai, 2004). In schizophrenia, it has been suggested that lower dopamine levels selectively stimulate high-affinity dopamine receptors (D3 and D5) and trigger inflammation, while high dopamine levels stimulate low-affinity dopamine receptors (D1 and D2), inducing an anti-inflammatory effect (Pacheco, 2017; Vidal and Pacheco, 2020).
Exposure to increased dopamine levels may also mediate effects through nondopamine receptors, such as adrenergic receptors, as higher concentrations of dopamine can bind to adrenergic receptors in different tissues and species (Cornil et al., 2002; Cornil et al., 2008; Lei, 2014; Ozkan et al., 2017; Aslanoglou et al., 2021), which can also drive inflammatory changes. For example, the β-adrenergic receptor antagonist propranolol inhibited dopamine-induced increases in nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB), interleukin (IL)-6, and IL-8 and blocked dopamine-mediated IL-12p40 suppression in human keratinocytes and rodent macrophages (Hasko et al., 2002; Parrado et al., 2012; Parrado et al., 2017). In the RAW264.7 rodent macrophage cell line, dopamine only affected nitric oxide (NO) production at a concentration of 5 × 10−6M, while much lower concentrations of the adrenergic receptor agonists norepinephrine and epinephrine increased lipopolysaccharide (LPS)-induced NO production, suggesting that dopamine may act through adrenergic receptors in this system (Chi et al., 2003).
Exposure to high levels of dopamine could also induce nonspecific changes in immune cell function through cytotoxicity and oxidative stress induced by the formation of dopamine quinones and other ROS (Graham et al., 1978; Stokes et al., 1999; Sulzer and Zecca, 2000). Studies in BV-2 microglia showed that quinone formation resulting from dopamine pre-treatment (10−6M–10−4M) for 24 hours attenuated LPS-induced expression of IL-6, tumor necrosis factor alpha (TNF-α), and IL-1β by inhibiting NF-κB (Yoshioka et al., 2016, 2020). Treatment with 0.5 to 2 × 10−5M dopamine induced cell cycle arrest and apoptosis in rapidly dividing B-cells via oxidative stress (Meredith et al., 2006), and in peripheral blood lymphocytes (PBLs), 1 to 5 × 10−4M dopamine induced intracellular ROS levels and apoptotic cell death through oxidative stress. Similar effects have been seen in other studies of primary human lymphocytes, which showed increased apoptotic marker levels and dose-dependent decreases in proliferation, differentiation, and the synthesis of IL-4 and interferon gamma (IFN-γ) in response to high dopamine concentrations (10−5–5 × 10−4M) (Bergquist et al., 1994, 1997). In addition, the antioxidant glutathione prevented high dopamine levels (6–10 × 10−5M) from reactivating latent human immunodeficiency virus (HIV) in a chronically infected T-cell line (Scheller et al., 2000), indicating that reactivation was induced by oxidative stress. Thus, the use of extraphysiologic dopamine levels could produce confounding results by increasing the activation of receptors that may not be physiologically relevant and inducing aberrant effects due to cell death and the subsequent response to factors released from apoptotic cells.
C. Evaluating and Accounting for Dopamine Receptor Expression
Cell type- and species-specific differences in dopamine receptor expression and signaling have important implications in the effects of dopamine on immune cell function and regulation. As has been reviewed recently (Levite, 2016; Pacheco, 2017; Pinoli et al., 2017; Matt and Gaskill, 2020) and will be discussed in subsequent sections, the majority of immune cells have been shown to express all types of dopamine receptors. Further, dopamine receptors are also expressed in several other nonneuronal cell types, such as renal proximal tubule cells (Dawirs and Teuchert-Noodt, 1992; Fardoun et al., 2007; Han et al., 2007), pancreatic beta cells (Rubi et al., 2005), brown adipocytes (Kohlie et al., 2017), and cells throughout the gut such as stomach parietal cells (Mezey et al., 1999; Li, Schmauss et al., 2006). However, the relative expression levels of the different dopamine receptor subtypes are not consistent between studies.
Dopamine receptor expression levels are typically analyzed at the mRNA level using in situ hybridization and reverse-transcription polymerase chain reaction or at the protein level by Western blotting, immunohistochemistry, or immunofluorescent staining. While mRNA expression does not equate to protein expression and cannot reveal the receptor expression levels in the plasma membrane, the absolute specificity of the probe/primer sequences can indicate expression, enable precise subtype differentiation, and suggest the relative ratios between the different subtypes. In contrast, protein analysis can theoretically demonstrate surface expression and be used to quantify receptor density. However, dopamine receptor antibodies often lack specificity (Bodei et al., 2009; Michel et al., 2009), likely due to the high homology among dopamine receptor subtypes within a given subfamily (Platania et al., 2012). Thus, antibodies may not sufficiently discriminate different receptor proteins on the cell surface, nor do they bind to denatured proteins in blotted membranes. Moreover, because the molecular sizes of dopamine receptor subtypes are relatively similar [D1, 49 kD; D5, 53 kD; D2 short isoform, 47 kD; D2 long isoform, 59 kD; D3, 44 kD; however, several shorter isoforms have been described (e.g., D4, 41 kD), although weight vary slightly based on the number of 48-base pair variable tandem repeats in exon 3] (Van Tol et al., 1992; Fishburn et al., 1993; Khan et al., 1998; Richtand, 2006; Beaulieu and Gainetdinov, 2011), their immunoblot signals could be, at least in part, superimposed (Bucolo et al., 2019).
These technical issues, as well as differences in the information provided by these methodologies, creates difficulties when trying to make comparisons across the literature. For instance, mRNA and protein expression levels of dopamine receptors vary greatly among human and rodent immune cell types, making comparisons across species complicated (Davis, 2008; Shay et al., 2013). Even within species, there can be large differences, as primary human macrophages have high expression of D1-like receptors and D2, but much lower expression of D3 and D4 (Nickoloff-Bybel et al., 2019; Nolan et al., 2019), while human THP-1 monocytic cells have substantially higher levels of D4, particularly relative to D1-like receptor expression (Basova et al., 2018). The murine immature osteoblast line, MC3T3-E1, only expresses D1 and D4, while all subtypes of dopamine receptors except D3 are expressed in primary osteoblasts (Motyl et al., 2017).
These differences are especially important when considering the impact of dopamine on cells isolated during different disease states or in activated and resting cells. Diverse stimuli, as well as many pathologies, can alter both dopamine concentrations and dopamine receptor expression levels, which could potentially activate different dopamine receptors than would normally respond to dopamine and promote important homeostatic activity. Changes in the expression of some dopamine receptors and not others could also affect function by altering the ratios of different dopamine receptors. As changes in dopamine receptors may be critical for determining the effects of dopamine on distinct cell types, it is important to define baseline expression levels, how those levels compare with other systems, and how those levels change throughout experiments. Moving forward, examining dopamine receptor ratios may be more relevant than measuring individual receptor expression. Further, examining receptor ratios in relation to other receptors that modulate dopamine signaling (e.g., adrenergic receptors) may be a better strategy for understanding dopamine-mediated patterns in immune function and disease.
D. Species-Specific Dopamine Signaling and Immune Function
Classically, dopamine signaling has been defined in neurons, but several studies have shown dopamine signaling differs between neurons and immune cells, as well as other nonneuronal cell types (Wang et al., 2005; Beaulieu and Gainetdinov, 2011; Beaulieu et al., 2015), and between species. This is particularly important regarding rodent–human differences, as rodents and rodent immune cells are commonly used to model human immune function. There are substantial differences between rodent and human immune responses due to genetic differences and the experimental environment (Beura et al., 2016; Tao and Reese, 2017). For example, LPS amplifies toll-like receptor (TLR)2/6 responses and downregulates CXCR4 in murine macrophages but does not affect TLR2/6 responses and increases CXCR4 expression in human macrophages (Ariffin and Sweet, 2013). There are also differences in the transcriptional profiles of rodent and human immune systems (Shay et al., 2013), and studies of inflammatory diseases using rodents often poorly translate to humans (Mestas and Hughes, 2004; Seok et al., 2013).
This disconnection is also seen regarding dopamine. In human primary monocyte-derived macrophages (hMDMs), D1-like receptor activation does not stimulate cAMP production (Nickoloff-Bybel et al., 2019) and primes the nucleotide-binding oligomerization–like receptor family pyrin domain containing 3 (NLRP3) inflammasome (Nolan et al., 2020). However, studies in LPS-primed murine bone marrow–derived macrophages (BMDMs), D1 activation inhibits NLRP3 activity through a cAMP-dependent pathway, although D1-mediated increases in cAMP were not directly observed (Yan et al., 2015). Although both studies examined D1-like receptors, there are clear differences in the activated pathways. These variations may result from species-specific differences in dopamine receptor or inflammasome activity, the presence of LPS, or the different concentrations of dopamine used, which may have activated different dopamine receptors. Similarly, both human and mouse pancreatic α- and β-cells express dopamine and adrenergic receptors, but there are large, species-specific differences in the ratios of dopamine receptors to adrenergic receptors. Further, dopamine induces dose-dependent changes in glucagon in human islets, but low levels decrease glucagon, while higher levels increase glucagon. In contrast, dopamine only dose-dependently increases glucagon in mouse islets. This may be due to variations in the ratios of receptors with different dopamine affinities (Aslanoglou et al., 2021). These types of dissimilarities could, at least partially, explain species-specific differences in dopaminergic immunomodulatory effects. Differences in environmental stimuli could also contribute to these effects, such as the comparison of LPS-stimulated and nonstimulated cells (Gaskill et al., 2012) or between cell activity in vitro and in vivo.
Additionally, dopamine receptors can form heteromers with other dopamine receptors, other types of GPCR, and even ion channels. This includes D1–D2, D1–D3, D2–D3, D2–D4, and D2–D5 complexes (Marcellino et al., 2008; So et al., 2009; González et al., 2012; Perreault et al., 2014), although there has been controversy about the existence of D1–D2 receptors under physiologic conditions (Rashid et al., 2007; Chun et al., 2013; Frederick et al., 2015; Hasbi, Sivasubramanian et al., 2020). Dopamine receptors also oligomerize with many other types of GPCR, mostly receptors associated with neurotransmission. This includes, but is not limited to, adenosine, N-methyl-D-aspartate, corticotrophin-releasing hormone, neurotensin, serotonin, histamine, and metabotropic and ionotropic glutamate receptors (Borroto-Escuela et al., 2013; Cahill et al., 2014; Ferre et al., 2014; Fuenzalida et al., 2014; Moreno et al., 2014; Perreault et al., 2014; Andrianarivelo et al., 2021). Some studies show sex differences (Hasbi, Nguyen et al., 2020) in dopamine receptor heteromers, and there may also be cell type or species-specific differences in the type or frequency of dopamine receptor oligomers, although this has not been well studied. Heteromeric receptors often display distinct signaling capacity and functional selectivity in ligand binding (Ferre et al., 2014), and the formation of distinct types of heteromeric complexes may be associated with the frequency of different receptors on distinct cell types. Thus, immune cells may be more likely to generate heteromeric complexes between dopamine receptors and cytokine or chemokine receptors that enable dopamine to influence immune activity. For example, a recent study showed that D5R and C-C motif chemokine receptor 9 (CCR9) form a heteromer on both mouse and human CD4+ T-cells and that these D5R:CCR9 complexes are increased during gut inflammation and drive colonic homing of these T-cells (Osorio-Barrios et al., 2021). Thus, whether immune dopamine receptors exist as monomers or as part of an oligomeric complex could have substantial impact on the dopamine-mediated signaling processes and immune functions in those cells. These and other data clearly show that much more research is needed to effectively interpret and synchronize our understanding of the broad impacts of dopamine, particularly across species.
E. Considerations Regarding the Effects of Dopamine Induced by Addictive Substances or Pharmacological Agents
Another concern when interpreting the immunologic effects of dopamine is the potential imprecision associated with the use of pharmacologic agents to activate dopamine receptors. Specificity of a pharmacologic agent is determined and/or relies on a distinct experimental system, so specificity may differ between systems with varying dopamine receptor levels, and more specific drugs or additional mechanistic studies may be needed to precisely target a particular receptor. This is problematic because when agonists and antagonists are not specific or selective enough, they do not provide the basis for the univocal and unambiguous identification of particular receptors (Salomone and Waeber, 2011).
For example, studies that treat immune cells in vitro with dopamine receptor antagonists often show modulatory effects. But if there is no dopamine in the system, it is not clear that the antagonist is preventing the effect that results from receptor activation. In these cases, antagonists may have effects on systems that are dopamine receptor–independent. Antagonists could also be acting on dopamine receptors in unexpected ways because the affinity for the dopamine receptors expressed in these systems are distinct from the system in which the antagonist was defined. It is also possible that the antagonist is blocking the effects of endogenous dopamine release and autocrine activation (we note in the next section that many immune cells produce and secrete dopamine as part of their communication strategy). Because it is often not clear how the pharmacologic agent is acting on the dopamine receptor being studied or what is the appropriate concentration of agonist/antagonist to use, extrapolations about the effects of dopamine based on pharmacologic drugs could be misleading.
Another similar issue occurs in studies evaluating the immunomodulatory effects of addictive drugs. As previously noted, all addictive substances, including stimulants such as cocaine and methamphetamine (Di Chiara and Imperato, 1988; Kimmel et al., 2005), alcohol (Wozniak et al., 1991; Kegeles et al., 2018), cannabis (Chen et al., 1993), or opioids such as heroin (Hemby et al., 1995), acutely increase CNS dopamine levels through distinct mechanisms of action (Pierce and Kumaresan, 2006; Volkow, Fowler et al., 2009). Further, many of these substances also act on other systems, such as the opioid or endocannabinoid system, or specific receptors such as sigma-1 (Tsai et al., 2015; Lever et al., 2016; Cai et al., 2017) or trace-amine associated receptor 1 (Cotter et al., 2015; Sriram et al., 2016). Thus, the in vivo immunomodulatory effects of these drugs could result from the dopamine released by the use of these substances or the interactions of the substances themselves with other receptors. While in vitro monocultures of immune cells can produce some dopamine, they lack the capacity to release dopamine as it is produced in vivo, and the presence and activity of additional receptors in these systems is often undefined. Thus, it is not necessarily accurate to attribute the immunomodulatory effects of addictive substances to dopamine when these effects are defined in vitro. As a result, many of these studies are actually evaluating the immunologic effects of the substances themselves rather than the dopaminergic impact of their use, and follow-up studies examining the specific receptors or signaling pathways induced by these substances could be fruitful. However, to fully address the immunomodulatory effects of dopamine induced when using addictive substances, in vitro mechanistic studies using the dopamine concentrations induced during SUDs should be combined with in vivo analyses to give a more complete picture of the impact of each addictive substance.
III. Dopamine and Immune Cells
A. Introduction
A growing body of evidence has shown that dopamine can modulate a variety of immune functions, including proliferation, chemotaxis, antigen presentation, phagocytosis, cytokine secretion, and cell adhesion (Cosentino et al., 1999; Sarkar et al., 2010; Levite, 2016; Nolan et al., 2018). Research defining the immunomodulatory effects of dopamine often generates conflicting data, suggesting that dopamine elicits activity or quiescence, or both pro- and anti-inflammatory states depending on the cell type, model system, and experimental context (Tarazona et al., 1995; Hasko et al., 1996, 2002; Sarkar et al., 2006; Capellino et al., 2010; Nakano et al., 2011; Gaskill et al., 2012; Franz et al., 2015; Yan et al., 2015; Zhang et al., 2015; Zhang, Jiang et al., 2016; Nolan and Gaskill, 2019; Yoshioka et al., 2020). Much of this research has focused on myeloid cells, particularly macrophages and microglia, and T-lymphocytes, although dopamine has been shown to affect immune function in most immune cell types.
Almost all types of immune cells express various levels of both the D1- and D2-like receptors, as well as other proteins involved in the synthesis, reuptake, transport, and metabolism of dopamine, such as DAT, TH, VMAT2, and MAOs (McKenna et al., 2002; Farber et al., 2005; Cosentino et al., 2007; Gaskill et al., 2009, 2012; Mastroeni et al., 2009; Kustrimovic et al., 2014; Coley et al., 2015; Huck et al., 2015; Levite, 2016; Nolan and Gaskill, 2019; Prado et al., 2021; Wieber et al., 2022). This suggests that most immune cells interact with dopamine through surface receptors and can take up dopamine through active transport. Uptake and release of dopamine can modulate both the releasing cell and neighboring cells, mediating a variety of functions, such as transforming growth factor (TGF)-β and IL-10 production or B-cell activation (Faraj et al., 1991; Cosentino et al., 2007; Pacheco et al., 2009; Arreola et al., 2016; Papa et al., 2017). It is not entirely clear whether the dopamine that mediates these interactions is produced de novo or taken up from the surrounding environment and released. However, studies inhibiting TH activity and measuring catecholamine levels in human neutrophils (Cosentino et al., 1999), lymphocytes (Musso et al., 1996; Qiu et al., 2005), peripheral blood mononuclear cells (PBMCs) (Marino et al., 1999; Cosentino et al., 2002), and B-cells (Honke et al., 2022) show changes in dopamine levels in immune cells, and follicular T-helper cells specifically produce and store dopamine in dense-core granules marked by chromogranin B (Papa et al., 2017). Stimulation of rodent macrophages and neutrophils with LPS also resulted in the production of catecholamines (Flierl et al., 2007). These studies indicate that immune cells produce small quantities of dopamine de novo, and many of these cells use the produced dopamine for autocrine or paracrine regulation by activating dopamine receptors on neighboring cells. The following sections will further discuss the dopaminergic systems in the immune cells of the CNS and periphery, as well as specific immune cell responses to dopamine signaling. Then, the bidirectional interaction of dopamine and inflammation will be reviewed, discussing the role of dopamine in driving inflammatory functions such as cytokine production and the impact of inflammation on dopaminergic machinery.
B. Dopaminergic Machinery and Activity in CNS Immune Cells
Dopamine levels in dopaminergic regions of the CNS, including the striatum, VTA, NAc, and prefrontal cortex (PFC), are among the highest in the body. Therefore, CNS immune cells, particularly in these regions, are likely to frequently encounter immunomodulatory levels of dopamine, and dopamine-mediated effects may be associated with homeostatic function in these regions (Matt and Gaskill, 2020). The most common immune cells in the CNS are myeloid cells, particularly microglia, as well as other types of CNS macrophages (Herz et al., 2017). Microglia are dynamic, yolk sac–derived, tissue-resident macrophages that make up a unique myeloid population in the CNS parenchyma (Prinz et al., 2019). These cells interact with neighboring neurons and other glia physically and through the production of cytokines and neurotrophic factors. Microglia play a critical role in the maintenance of neuronal health, synaptic pruning, and the organization of neuronal circuits (Schafer et al., 2012; Schafer et al., 2013; Li and Barres, 2018). These cells also protect the CNS by surveilling the parenchyma for pathologic insults or infection and differentiate into various activation states depending on the pathologic stimuli detected (Ousman and Kubes, 2012). Notably, microglia are overrepresented in dopaminergic midbrain pathways relative to other brain regions, so these areas may be particularly sensitive to the inflammatory effects of dopamine (Kim et al., 2017; Treadway et al., 2019). Activated cells can induce the production of inflammatory modulators such as IL-1, NO, IL-10, TNF-α, superoxide, and prostaglandin E2 to promote and/or quell inflammation (Wolf et al., 2017).
All five dopamine receptor subtypes have been identified on human microglia (McKenna et al., 2002; Mastroeni et al., 2009), human microglial cell lines (Matt et al., 2021) and rodent microglial cells (Farber et al., 2005; Kettenmann et al., 2011; Huck et al., 2015; Kopec et al., 2018) although not every study detected all subtypes. There may be an age-associated effect, as cultured human microglia from elderly individuals did not express Drd5 mRNA despite the presence of mRNA for the remaining dopamine receptors (Mastroeni et al., 2009). In rodents, microglial dopamine receptors and transcriptomes (De Biase et al., 2017) vary among brain region and local environment (Kuric and Ruscher, 2014; Huck et al., 2015), although this has not been studied in primates. Rodent microglia have also been shown to express DAT and the metabolic enzyme COMT (Myohanen et al., 2010; Fan et al., 2018), but studies are still needed to define the full dopaminergic system present in human microglia.
Dopamine has several effects on microglia, many pertaining to neuroinflammation and the activation state of these cells. In wild-type mice, no D2 expression was detected in Iba-1+ microglia, but cerebral ischemia and the associated inflammation induced high levels of D2 expression in these cells (Huck et al., 2015). In a separate study, both D1 and D2 were present in resting murine microglia, but only D2 mediated anti-inflammatory changes through the expression of renin-angiotensin receptors. In contrast, in LPS-induced microglia, both D1 and D2 receptors mediated this effect (Dominguez-Meijide et al., 2017). Furthermore, microglial activation was reduced by global ablation of D2 in a murine 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD, although this was likely through T-cell-mediated inflammation (Liu, Zhai et al., 2021).
Studies in microglial cell lines and primary adult microglia show that with stimulation with phorbol myristate acetate or LPS, but not in unstimulated cells, dopamine (2.5 × 10−6M) induced the formation of extracellular traps. Although extracellular traps are normally generated in granulocytes, dopamine has not been shown to affect trap formation in that cell type. The novelty of this process was increased by the finding that these extracellular traps were formed independent of ROS production, actin polymerization, or cell death (Agrawal et al., 2021; Wu et al., 2021). This phenomenon may be particularly relevant to glioblastoma multiforme, a malignant brain tumor in which sterile neuroinflammation occurs along with release of dopamine (Agrawal et al., 2021; Alghamri et al., 2021; Roesler et al., 2021), suggesting that dopamine may be involved in the inflammation associated with this disease. There may also be sex-dependent differences in dopamine receptor expression associated with microglial functions. Microglial and complement-mediated phagocytosis both eliminate D1-like receptors in males but not females, and this elimination shapes NAc development (Kopec et al., 2018). These studies indicate that microglia express a fully functional dopaminergic system that may affect many microglial functions (summarized in Fig. 3). However, the effects of this system could vary widely depending on the brain region and local environment, and the specific role of dopamine in the function of human microglia in both health and disease requires further study.
Dopamine receptor signaling in myeloid cells. Current knowledge of immunomodulatory effects of dopamine signaling in monocytes, macrophages, and microglia are summarized. In monocytes, dopamine signaling through the dopamine receptors leads to an increase in chemokine production, chemokinesis, and transmigration. Studies in macrophages show that dopamine can have bidirectional effects on phagocytosis and NO production, while stress response genes, NF-kB activation, and release of proinflammatory mediators are all increased in response to dopamine. In microglia, dopamine signaling through its receptors increases chemotaxis, phagocytosis, formation of extracellular traps and pro-inflammatory mediator production while decreasing NO production. Additionally, in microglia, in general D1-like receptor stimulation inhibits NF-kB while D2-like receptor stimulation activates NF-kB. Created with BioRender.com.
In addition to microglia, there are other immune cell populations in the CNS, including peripheral immune cells such as T-cells and monocytes, a small number of which transiently survey the CNS during homeostasis (Prinz and Priller, 2017). The dopaminergic system in peripheral immune cells is discussed in subsequent sections. There are also many specialized CNS-resident macrophage populations, including perivascular macrophages, choroid plexus macrophages, and meningeal macrophages, although these cells are much less well studied than microglia. Classically, these macrophage populations were thought to be more closely related to peripheral macrophages than microglia, but fate mapping studies suggest that CNS macrophage populations are also yolk sac-derived and make up a stable, low-turnover population that is relatively transcriptionally related to microglia (Goldmann et al., 2016; Prinz and Priller, 2017). Further, recent studies suggest that some CNS macrophage populations are derived from the skull and vertebral bone marrow, and these populations occupy unique niches within the CNS (Cugurra et al., 2021). The functions of these specialized macrophage populations are not well defined, although major functions include sampling and removing various types of debris, providing trophic support to neurons and glia, and regulating the immune responses at different CNS boundaries such as the perivascular space, the choroid plexus, and the lymphatic and glymphatic systems (Herz et al., 2017). Although these cells are similar to microglia, it is not clear how much overlap is present between the dopaminergic systems in these specialized CNS macrophages and microglia.
Finally, while they are not considered immune cells, astrocytes are the largest population of cells in the CNS and play a central role in neuronal health and function, often having an immunomodulatory role in response to CNS insult or disease (Sofroniew, 2014; Giovannoni and Quintana, 2020). Like microglia, astrocytes can take on a reactive phenotype in response to CNS damage, inflammatory stimuli, and microglial activation (Hamby et al., 2012; Sofroniew, 2014; Liddelow et al., 2017; Giovannoni and Quintana, 2020). Activated astrocytes have both neuroprotective and neurotoxic effects, secreting both inflammatory and anti-inflammatory cytokines and chemokines that contribute to tissue repair and neuroinflammation (Markiewicz and Lukomska, 2006; Sofroniew, 2014; Giovannoni and Quintana, 2020; Linnerbauer and Rothhammer, 2020). An in-depth discussion of astrocytes is outside the scope of this review, but studies have shown that rodent astrocytes express all dopamine receptor subtypes (Bal et al., 1994; Zanassi et al., 1999; Reuss et al., 2000; Miyazaki et al., 2004; Montoya et al., 2019) as well as other dopaminergic proteins such as DAT (Takeda et al., 2002), monoamine oxidase B (MAO-B), and COMT (Fitzgerald et al., 1990; Myohanen et al., 2010; Winner et al., 2017). Astrocytes can also take up and metabolize dopamine (Pelton et al., 1981; Inazu et al., 1999), with at least one study indicating dopamine transport is regulated by the norepinephrine transporter (Takeda et al., 2002). Astrocytic dopamine receptors may play a role in inflammation (Shao et al., 2013; Zhang et al., 2015), as well as neuronal health and survival (Ohta et al., 2003, 2010; Li, Guo et al., 2006), indicating that dopamine responsiveness is important for glial cells within the CNS. There are species-dependent differences in astrocyte dopamine receptor expression and activity, and some studies have shown regional variations in dopamine receptor expression and responses. Astrocytes in dopaminergic regions such as the striatum, VTA or PFC express dopamine receptors and respond to dopamine, while those in other regions, such as the cerebellum, do not (Vermeulen et al., 1994; Khan et al., 2001; Reuss and Unsicker, 2001; Miyazaki et al., 2004; Xin et al., 2019). This finding indicates that the effects of dopamine on astrocytes, like many other cell types, is context- and environment-dependent.
C. Dopaminergic Machinery and Activity in Peripheral Immune Cells
1. Innate Immune Cells
The innate immune response is considered the first line of defense against invading pathogens and mediates rapid, nonspecific inflammatory responses. In innate cells, particularly myeloid cells, granulocytes, NK cells, and DCs, these responses are generally initiated by the exposure of extracellular and intracellular pattern recognition receptors (PRRs) to various stimuli associated with pathogens or other insults. The four main families of PRRs are toll-like receptors (TLRs), nucleotide-binding oligomerization domain-like receptors (NLRs), C-type lectin receptors, and RIG-1 like receptors (Janeway and Medzhitov, 2002; Takeuchi and Akira, 2010; Pinoli et al., 2017). TLRs are membrane proteins that are localized on endosomes and mediate extracellular recognition of pathogens, whereas NLRs are cytosolic proteins that recognize intracellular pathogens (Janeway and Medzhitov, 2002; Franchi et al., 2009; Kumar et al., 2011). There are currently 10 known functional TLRs in humans and 12 in mice, while there are 22 human NLRs (Kawai and Akira, 2008; Takeuchi and Akira, 2010). Formyl peptide receptors and scavenger receptors are also PRRs, as they bind N-formyl peptides produced by bacterial degradation and acetylated or oxidized low-density lipoproteins, respectively (Janeway and Medzhitov, 2002; Takeuchi and Akira, 2010; Pinoli et al., 2017).
PRRs act primarily by recognizing a variety of molecules known as pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), which signal the presence of danger. Examples of PAMPs and DAMPs include LPS, which is a component of Gram-negative bacterial cell walls; single- or double stranded RNA, which is associated with viral infection; β-glucans, which are components of fungal cell walls; and immunostimulants, such as polyinosinic:polycytidylic acid, which mimics activation caused by viral RNA. The expression and activation of PRRs on innate immune cells mediates coordinated effector responses upon contact with an invading pathogen (Janeway and Medzhitov, 2002; Li and Wu, 2021). Dopamine receptors and other dopamine-related proteins have been detected on most of these cells, suggesting the potential for broad effects of dopamine on many branches of the innate immune system (Pinoli et al., 2017).
a. Monocytes and Macrophages
Monocytes are myeloid cells that circulate through the blood and lymphatic system and can be rapidly recruited to sites of tissue damage and infection. Within tissues, these cells differentiate into macrophages, which secrete inflammatory cytokines and trophic factors, engulf and eliminate pathogens, regulate development and homeostasis, and mediate tissue repair/wound healing, thereby playing major roles in protective immunity. The influence of dopamine on myeloid cell activity is show in Fig. 3. Monocytes and macrophages in humans and other mammals express all dopamine receptor subtypes, as well as other dopamine-related proteins including DAT, VMAT2, TH, AADC, COMT, and both MAO-A and -B (Tarazona et al., 1995; McKenna et al., 2002; Brown et al., 2003; Liang et al., 2008; Gaskill et al., 2009, 2012; Coley et al., 2015; Bone et al., 2017; Nolan et al., 2019). Studies show that primary human (Cosentino et al., 1999; Mackie et al., 2022) and rodent myeloid cells (Flierl et al., 2009), as well as rice stem borer (Chilo suppressalis) hemocytes, blood cells that are analogous to macrophages in vertebrates (Wu et al., 2015), can take up, store, and produce dopamine.
The capacity of macrophages to synthesize dopamine is supported by studies that show that intracellular dopamine levels in myeloid cells are decreased by the catecholamine synthesis inhibitor alpha-methyl-p-tyrosine (Freeman et al., 2001) and that M2-polarization in rodent macrophages may reduce dopamine levels in these cells (Fischer et al., 2017). Both the monocytic murine cell line RAW264.7 and the human myeloid cell line U937 store dopamine and express L-DOPA decarboxylase, and LPS stimulation of RAW264.7 cells increased TH mRNA expression and intracellular dopamine within 48 hours (Brown et al., 2003; Kokkinou et al., 2009). The expression of TH and VMAT2 in U937 cells is only seen in response to LPS, suggesting that dopamine production in this cell line could be tied to activation (Capellino et al., 2010). This is also seen in human monocytes, where stimulation with TNF-α increases the number of TH+ cells and the amount of TH present in each cell (Gopinath et al., 2021). Notably, hMDMs do not express dopamine-β-hydroxylase (Nolan and Gaskill, 2019), indicating that dopamine cannot be converted to norepinephrine and that it is produced for use as dopamine.
In addition to myeloid cells in the blood, there are many tissue-specific macrophages, heterogeneous populations of myeloid cells that fulfill niche-specific functions. These populations include but are not limited to a variety of CNS myeloid cells like microglia and meningeal macrophages (brain), alveolar macrophages (lung), Kupffer cells (liver), adipose-associated macrophages (adipose tissue), Langerhans cells (skin), osteoclasts and bone marrow macrophages (bone), and intestinal macrophages (gut). There is very little specific data on the effect of dopamine on tissue-specific macrophages other than microglia, although recent studies have identified DAT on a subset of gut macrophages in the lamina propria (Mackie et al., 2022). This lack of information is likely because identifying, isolating, and working with specific tissue macrophage populations is technically challenging, and many studies on macrophages, including the vast majority of those examining the dopaminergic system, are done with hMDMs or BMDMs matured in vitro. While the use of hMDMs and BMDMs has and continues to provide valuable data regarding macrophage function, it is important to recognize that these cells are distinct from tissue-specific macrophages in many ways (Davies et al., 2013; Gordon and Pluddemann, 2017). Further, dopamine increases the expression of stress response genes such as hypoxia inducible factor-1a and nuclear factor erythroid 2-related factor in murine BMDMs via the uptake of unbound iron (Dichtl et al., 2018). This finding suggests that dopamine influences many more macrophage functions than are currently understood. Future studies examining the impact of dopamine on tissue-specific macrophages, as well as on general macrophage function, should account for these differences and improve understanding of the specific impact of tissue macrophages on homeostasis and disease.
b. Granulocytes
There has been relatively little research on the impact of dopamine on the four types of granulocytes: neutrophils, basophils, eosinophils, and mast cells. Dopamine has been shown to mediate functional changes in neutrophils, eosinophils, and mast cells (Fig. 4) with little current research on basophils. The majority of the research has been studied in neutrophils (Pinoli et al., 2017). These cells are relatively short-lived granulocytes derived from the bone marrow that quickly move into the blood. Neutrophils are the most abundant granulocyte in circulation (40%–70% of blood leukocytes) and are often the first line of host defense, inhibiting or eliminating invading pathogens through the generation of cytokines, ROS, neutrophil extracellular traps, and other factors. Recent studies have shown that neutrophils play a critical role in communication between the innate and adaptive immune response through direct interactions or cytokine signaling (Rosales, 2020). The expression of both D1-like and D2-like receptors on human neutrophils has been confirmed at both the mRNA and protein levels (Cosentino et al., 1999; Sookhai et al., 1999; McKenna et al., 2002; Pereira et al., 2003; Chen et al., 2014).
Dopamine receptor signaling in granulocytes. Current knowledge of the immunomodulatory effects of dopamine signaling in neutrophils, eosinophils, and mast cells is summarized. In neutrophils, dopamine acts through its receptors to increase formation of extracellular traps and to decrease NO production and transendothelial migration, while having reported bidirectional effects on neutrophil phagocytosis. Dopamine signaling in eosinophils has been shown to affect eosinophil counts with high doses of dopamine leading to eosinopenia and low doses of dopamine leading to eosinophilia. In mast cells, dopamine acting at D1-like receptors has been shown to increase degranulation while D2-like receptor stimulation decreases cytokine production. Created with BioRender.com.
Dopamine regulates neutrophil activity, as dopamine treatment (2.61 × 10−7 M) attenuated the expression of CD62l (L-selectin) and CD11b in neutrophils stimulated with N-formyl-methionyl-leucyl-phenylalanine, a potent neutrophil activator that mimics the actions of factors released by bacteria. This effect was not seen in resting neutrophils or in response to lower dopamine concentrations (2.61 × 10−10M) (Trabold et al., 2007). The downregulation of L-selectin and CD11b could substantially interfere with adherence to and rolling on activated endothelium, which is a primary function of these cells. Higher levels of dopamine (10−4M–10−5M) increased apoptosis in human neutrophils (Sookhai et al., 1999; Aslan et al., 2011), and the dopamine receptor antagonists chlorpromazine and pimozide blocked the increases in neutrophil counts associated with exposure to ovalbumin peptide in rats. In contrast, the dopamine receptor agonist apomorphine increased neutrophil numbers (Altenburg et al., 1995), suggesting that dopamine receptor activity could regulate neutrophil numbers and function and that higher levels of dopamine reduced neutrophil numbers, while lower levels may increase them.
Eosinophils are another granulocyte subset that are much less common than neutrophils (2%–3% of blood leukocytes) that have a key role in defense against helminth infection, injured tissue repair, and allergic diseases. These cells are released into the circulation like neutrophils but generally reside in tissues, mediating their effects through the release of cytoplasmic granules, cytokines, chemokines, and growth factors (Aoki et al., 2021). Very few studies have examined the dopaminergic system in these cells, but they have been shown to express all five subsets of dopamine receptors on their surface (McKenna et al., 2002). Eosinophils also express TH and can synthesize and release dopamine. Dopamine synthesis is slightly increased following activation with IL-5 and eotaxin, and inhibiting eosinophil TH activity with alpha-methyl-p-tyrosine decreased murine vascular relaxation in vitro. This suggests that the release of dopamine or other catecholamines is important for eosinophil function (Withers et al., 2017). In rats, administration of low-dose L-DOPA or apomorphine increased blood eosinophil count, while higher doses of these agents decreased blood eosinophil count, suggesting that dopamine synthesis and dopamine receptor activity affects eosinophil viability (Podolec et al., 1979). The usefulness of these findings is limited by our understanding of the dopaminergic system in eosinophils and is an area in need of further study.
Mast cells are myeloid lineage cells that originate in the bone marrow, but unlike other granulocytes, these cells reside in regions that are exposed to the external environment, such as mucosal and epithelial tissues. Mast cells are also common in connective tissues, and the many types of mast cells depend on the tissue environment in which they reside. These cells are important effectors of host defense against bacteria, venomous toxins, and triggers of allergic responses and anaphylaxis. During the immune host response, these cells release factors that rapidly recruit other innate and adaptive immune cells, which creates a balanced response to infection. Mast cells contain granules that store a variety of inflammatory mediators including cytokines and chemokines such as IL-4, C-C motif chemokine ligand 2 (CCL2), C-C motif chemokine ligand 5, and TNF-α; growth factors such as TGF-β and vascular endothelial growth factor; proteoglycans, proteases, and other enzymes; and biogenic amines such as histamine, serotonin, and dopamine (DeBruin et al., 2015). Mast cells express some dopamine receptors, and several studies indicate that dopamine plays an important role in mast cell function. Human synovial mast cells from patients with rheumatoid arthritis (RA) express D3, although these receptors were only seen on a subset of mast cells (Xue, Li et al., 2018). mRNA for D1 and D5 receptors was found in bone marrow and fetal skin derived mast cells (Mori et al., 2013). Bone marrow-derived murine mast cells also express TH and store dopamine in granules via a process that seems to be dependent on the presence of serglycin, an intracellular proteoglycan. Degranulation and the release of intracellular dopamine may be triggered by intracellular Ca2+ flux, which is mediated by IgE crosslinking (Ronnberg et al., 2012). Other studies have confirmed dopamine storage in murine (Freeman et al., 2001) and bovine mast cells (Edvinsson et al., 1977).
The release of dopamine is important for mast cell function, as dopamine-mediated (10−9M–10−7M) activation of D1-like receptors dose-dependently increased murine mast cell degranulation. Antagonizing D1-like receptors with SCH23390 also reduced ear swelling caused by passive cutaneous anaphylaxis, which is a mast cell–mediated process (Mori et al., 2013). Although not all D2-like receptors are present on mast cells, treating the rat cell line RBL-2H3 with bromocriptine, 7-OH-DPAT, haloperidol, and clozapine resulted in potent dose-dependent inhibition of degranulation (Seol et al., 2004). These ligands all bind to D2-like dopamine receptors, including D3, suggesting that D2-like receptors may also affect mast cell activity, although this cell type is actually derived from basophils, and it is not clear if it is entirely representative of mast cell biology (Passante and Frankish, 2009). Other studies suggest that D3 activity may be anti-inflammatory in mast cells, as there is a negative correlation between the numbers of D3-expressing mast cells in synovial fluid and disease severity in RA (Xue, Li et al., 2018). Furthermore, studies using bone marrow–derived mast cells from Drd3 knockout mice showed that D3 regulated the inhibitory effects of methamphetamine on LPS-induced cytokine production in mast cells. As methamphetamine does not bind to dopamine receptors, these data suggest that methamphetamine facilitates dopamine release in these cells by activating an autocrine signaling pathway by which the mast cells activate D3 to block inflammation (Xue et al., 2016). Taken together, these findings suggest that D1-like receptors may drive inflammatory activity in mast cells, while D2-like receptors may act in an anti-inflammatory manner.
Basophils are the largest and least common type of granulocyte, making up less than 1% of blood leukocytes. Basophils are very similar to mast cells but are found in the circulation rather than in tissues. After entering the peripheral blood, basophils transmigrate to various organs, such as the spleen, lymph nodes, and other inflammatory sites, where they can be activated. Like all granulocytes, these cells regulate innate inflammation and specialize in fighting parasites and regulating allergic responses. Basophils store inflammatory mediators such as IL-4 and TNF-α, as well as histamine, releasing them upon IgE crosslinking with surface receptors (Schwartz et al., 2016). The expression of dopamine receptors and other dopamine-related proteins on basophils is largely unclear, although human basophils do express VMAT2 (Anlauf et al., 2006), suggesting these cells can store monoamines in cytoplasmic vesicles. While there are no specific data on the functional effects of dopamine in this population, treatment with very low doses of epinephrine reduced histamine release (Mannaioni et al., 2010), and serotonin exposure significantly decreased IL-4 release (Schneider et al., 2011). These findings suggest that monoamines can regulate basophil function and that dopaminergic modulation may occur in regions with sufficient dopamine. For example, murine basophil activation leads to the release of serine proteases such as mMCP-8 and mMCP-11, which enhance microvascular permeability, allowing T-cells and other immune cells to transmigrate to sites of inflammation (Miyake and Karasuyama, 2017; Yamanishi et al., 2017). Dopamine has a known role in vascular regulation (Bhattacharya et al., 2008) and has been detected in the spleen and lymph nodes, which basophils frequent (Matt and Gaskill, 2020), suggesting that these cells are involved in this process. These interactions are currently unclear because of the lack of research on dopamine signaling in basophils, preventing a more comprehensive understanding of the role of dopamine in diseases characterized by basophilia, such as allergy or myeloproliferative disorders.
c. Natural Killer Cells
NK cells are granular lymphocytic cells that are often described as being in the gap between innate and adaptive immunity. These cells are named for their capacity to kill virus-infected and tumor cells without priming via antigen presentation or the recognition of major histocompatibility complexes (MHC) on target cells. Indeed, one of the primary functions of these cells is to recognize and eliminate other cells that lack MHC class I molecules. NK cells act by producing and secreting cytokines and chemokines that can influence the immune response and/or induce death pathways in infected cells (Ljunggren and Karre, 1990; Biron et al., 1999; Capellino et al., 2020). A variety of neurotransmitters, including dopamine, have been shown to influence NK cell function (Capellino et al., 2020), and human NK cells highly and consistently express all five dopamine receptors (McKenna et al., 2002; Zhao et al., 2013; Mikulak et al., 2014).
Dopamine may have both inhibitory and activating effects on NK cells. In NK cells isolated from human PBMCs, activation with IL-2 increases the expression of D5, allowing low levels of dopamine (10−9–10−12M) to inhibit cellular proliferation and reduce the synthesis of IFN-γ (Mikulak et al., 2014). In contrast, in NK cells isolated from mouse spleens, SKF38393 (D1-like agonist) treatment increased D1-like receptor expression and signaling through the cAMP–PKA–CREB pathway, enhancing cytotoxic activity against YAC-1 lymphoma cells. This study also showed that quinpirole (D2-like receptor agonist) reduced the expression of D3 and D4, as well as cytotoxic activity, and that antagonizing D2-like receptors with haloperidol blocked this effect (Zhao et al., 2013). Haloperidol also decreased NK cell activity in a different murine model (Nozaki et al., 1996), and in APO-SUS rats, which have a hyperreactive dopaminergic system characterized by increased expression of TH mRNA and D2 receptors, there is decreased splenic NK cell activity (Teunis et al., 2004). While this finding suggests that D2-like receptors generally suppress NK activity, one study assessing the effects of seven different dopamine receptor inhibitors on NK activity showed that only the pan-dopamine receptor antagonists thiothixene, fluphenazine, and trifluoperazine suppressed murine splenic NK cell cytotoxicity and effector-target cell conjugation (Won et al., 1995). This finding suggests that activation of multiple types of dopamine receptors or crosstalk with other receptors is needed to suppress NK activity or that these agents may affect NK cell immune function through alternative, nondopaminergic mechanisms.
d. Dendritic Cells
Dendritic cells (DCs) are professional antigen-presenting cells derived from both myeloid and lymphoid progenitors. The primary function of these cells is to migrate to secondary lymphoid organs and interact with T-cells to promote the differentiation of various effector T-cell subsets (Patente et al., 2019). Both human and rodent DCs express all dopamine receptor subtypes and other dopamine-related proteins such as MAO-A and -B, TH, and VMAT2. As in macrophages, dopamine-β-hydroxylase is not expressed in DCs, and interestingly, DAT was also not found, suggesting that these cells may not be able to take up dopamine from the extracellular space (Chen, Tsai et al., 2012; Prado et al., 2012; Figueroa et al., 2017; Arce-Sillas et al., 2019). Human DCs can produce dopamine de novo, and intracellular dopamine levels increase in response to synthetic L-DOPA. Dopamine production seems to be regulated by D2-mediated cAMP signaling that activates TH, as D2 blockade and treatment with forskolin, an activator for adenylate cyclase, increased stored dopamine. By inhibiting D2, dopamine signals predominately through D1-like receptors, which canonically stimulate the formation of cAMP (Beaulieu and Gainetdinov, 2011), increase phosphorylation of TH, and increase dopamine production (Nakano et al., 2009). Once produced, dopamine is stored in vesicles near the plasma membrane, which release dopamine. It is possible that the release acts in an autocrine fashion, binding to DC dopamine receptors to regulate proper dopamine levels, as dopamine is important in their role in antigen presentation and T-cell activation. These and other effects of dopamine on DC activity are summarized in Fig. 5.
Dopamine receptor signaling in T-cells dendritic cells. Current knowledge of dopaminergic immunomodulation in dendritic cells and T-cells. Dopamine receptor balance on dendritic cells appear to impact T-cell differentiation. Increased expression of D1, D2, and D3 receptors on dendritic cells induces a Th1 phenotype in T-cells. Increased D1 expression with low D2 expression drives Th2 phenotype while increased D1, D3, and D5 along with low D4 expression has been linked to Th17 phenotype. Double arrows before receptor expression indicate a stronger correlation of the corresponding receptor on dendritic cells that drive T-cell differentiation. On activated CD4+ T-cells, dopamine stimulates D1-like receptors to increase IL-5 and IL-17 production and stimulates D2-like receptors leading to reduced IL-2, IL-4, and IFN-y production. On CD8+ T-cells, dopamine stimulation of D1-like receptors leads to decrease cytotoxicity and decreased immune suppressive activity. By acting on D2-like receptors on resting CD8+ T-cells, dopamine increases IL-10 and TNF-α production. Created with BioRender.com.
When DCs interact with CD4+ T-cells, dopamine is released and activates D1-like receptors to increase cAMP signaling, which regulates T-helper (Th)1–Th2 polarization (Nakano et al., 2009). However, D2 may also be involved in this process, as blocking D2-like receptors in human monocyte-derived DCs with risperidone but not haloperidol inhibited DC production of Th1 cytokines (IL-6, IL-8, TNF-α). Blocking D2-like receptors also increased Th2 cytokine (IL-10) production and reduced Th1 polarization in cocultured T-cells (Chen, Tsai et al., 2012). Interestingly, haloperidol did affect the activity of murine bone marrow–derived DC, reducing the expression of MHC class II, CD80, and CD86, and decreasing the production of IL-12p40, an important factor for DC maturation (Matsumoto et al., 2015). This finding suggests species-specific differences in receptor crosstalk on murine and human DC, as both haloperidol and risperidone have strong affinities for D2-like receptors, but risperidone has much higher affinity for serotonin receptors.
Several studies from the Pacheco group indicate that the specific D1-like receptor that mediates DC–T-cell interactions is D5, as D5-mediated increases in cAMP increase the activation of signal transducer and activator of transcription and drive inflammation (Prado et al., 2018). The LPS-induced maturation of murine DCs decreased D5 expression, which impaired the activation and proliferation of antigen-specific CD4+ T-cells in vitro. Transplanting these D5-deficient DCs into a mouse showed a significant reduction in the percentage of Th17 cells infiltrating the CNS compared with that in wild-type animals (Prado et al., 2012). In the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis (MS), adoptive transfer of D5-deficient DCs reduced the severity of EAE and the frequency of inflammatory CD4+ T-cell subsets in the CNS. Reserpine-mediated dopamine depletion in wild-type DCs prior to transfer into EAE animals also reduced clinical severity, indicating that autocrine dopamine activation of D5 on DCs mediated the inflammatory effects of dopamine in this system (Prado et al., 2018). Because SCH23390 also blocks D5 activity, these data are supported by a separate study in which the inhibition of D1-like receptors with SCH-23390 blocked DC-mediated Th17 differentiation in EAE mice, although treatment with the D4-selective antagonist L750667 induced Th17 differentiation (Nakano et al., 2008). Adoptive transfer of D3-deficient DCs into mice had no effect on CD4+ T-cell responses but increased antigen cross presentation and CD8+ T-cell activity against tumors (Figueroa et al., 2017). This finding indicates that different dopamine receptors have discrete roles in antigen presentation and T-cell activation-associated signaling pathways in DCs. Because changes in antigen presentation impact the ability of the adaptive immune system to create memory, defining the overlapping and discrete dopamine pathways by which DCs regulate T-cell responses could be of substantial value in the treatment of autoimmunity.
2. Adaptive Immune Cells
The primary cells that mediate adaptive immunity are T- and B-lymphocytes, which interact with the innate immune system to generate specific responses to invading pathogens, killing infected cells and generating specific antibodies against pathogens. A critical feature of adaptive immunity is immune memory, which enables the body to rapidly respond to repeat insults using factors that specifically target those pathogens. Lymphocytes, particularly T-cells, can differentiate into numerous subpopulations that have a wide variety of functions depending on the maturation process and stimuli to which they are exposed. The major T-cell subsets are classified by surface expression of CD4 or CD8. The CD4+ “helper” T-cells coordinate the immune response, activating other adaptive immune cells, such as memory B-cells and effector T-cells, and innate immune cells, such as macrophages. These cells are activated by exposure to peptide antigens on the surface of antigen-presenting cells, such as DCs and macrophages and differentiate into Th subtypes that rapidly proliferate and secrete various cytokines. The subsets into which CD4+ T-cells differentiate include Th1, Th2, Th17, and others depending on the stimulus encountered and the type of immune function they need to execute. The CD4+ regulatory T-cell (Treg) subset primarily prevents a response to self-antigens and suppresses detrimental immune responses. CD8+ cells, also called cytotoxic T-cells or cytotoxic T lymphocytes, are activated by CD4+ T-cells or antigen-presenting cells, producing cytotoxins such as perforin and granzyme, Fas ligand, and various cytokines that destroy target cells such as virus-infected cells or tumor cells.
The other major lymphocyte that mediates adaptive immunity is the B-cell, which primarily produces antibodies, although B-cells can also act as antigen-presenting cells and produce cytokines. Naïve or memory B-cells express B-cell receptors on the plasma membrane, and when this receptor is activated by a specific antigen, the B-cell proliferates and differentiates into a plasma cell, producing large quantities of specific antibodies to fight invading pathogens. Both T-cells and B-cells can develop into memory cells that can persist in a quiescent state for long periods of time until activated by the specific antigen to which they respond. Once the memory cell is activated, it generates a rapid adaptive immune response. Lymphocyte differentiation, the specific roles of lymphocytes subsets, and the formation of immune memory are central to the adaptive immune response, and this topic is an extraordinarily expansive and well-researched field, with numerous excellent reviews (Murphy et al., 2008; Chaplin, 2010; Kurosaki et al., 2015; Netea et al., 2020). The role of dopamine in adaptive immunity has also been well described, and many studies and reviews have discussed the different functions dopamine can modulate in T-lymphocytes (Pacheco et al., 2009; Gaskill et al., 2013; Levite, 2016; Levite et al., 2017), although there has been much less study of B-cells (Tsao et al., 1997; Meredith et al., 2006).
a. T-Lymphocytes
T-cells are among the most well-characterized immune cell types associated with dopaminergic regulation and have been reviewed in detail, so we will discuss them more briefly. All dopamine receptor subtypes are expressed on the T-cell surface (Levite et al., 2001; McKenna et al., 2002; Besser et al., 2005; Kustrimovic et al., 2014). The binding profiles of dopamine receptor-specific ligands in these cells is similar to those observed in neuronal membranes, suggesting that the receptors may act similarly to those found in neurons (Takahashi et al., 1992). T-cells also express TH, DAT, VMAT2, and COMT, and different types of T-cells can synthesize, store, release, and take up dopamine (Bergquist et al., 1994; Josefsson et al., 1996; Cosentino et al., 2007; Papa et al., 2017), although the dopamine concentrations involved in these processes are not the same among T-cell subsets. For example, human CD4+CD25+ Tregs contained significantly higher dopamine levels than CD4+CD25− T-cells (Cosentino et al., 2007), and human T follicular helper cells (Tfh) stored much higher concentrations of dopamine than naïve T-cells (Papa et al., 2017). Some studies suggest that T-cells tightly regulate intracellular dopamine concentrations through a balance of synthesis, uptake and release (Bergquist et al., 1994). Treatment of lymphocytes with IFN-β, extracellular dopamine (10−6M–10−8M), or the PKC activator phorbol myristate acetate increased the production and release of dopamine and other catecholamines (Musso et al., 1996; Ferrari et al., 2004; Cosentino et al., 2005). The effects of dopamine on T-cell activity and maturation are summarized in Fig. 5.
As many of these effects are also distinct between T-cell subsets, the effects of dopamine are likely influenced by distinct dopamine receptor profiles. For example, D1-like receptors are highly expressed in both naïve and memory T-cells, while D2-like receptors are expressed mainly in memory T-cells and only marginally in naïve cells (Nakano et al., 2008; Mignini et al., 2013). The density of D1 expression is also significantly lower in human CD4+CD25+ Tregs than in CD4+CD25− T-cells (Cosentino et al., 2007), and there is significantly higher D3 expression in CD8+ T-cells than in CD4+ T-cells (Watanabe et al., 2006). In rat thymocytes, the expression of dopamine-associated proteins such as DAT and VMAT2 was higher in CD8+ cells than CD4+ cells, and DAT and D1-like receptors are expressed at higher levels than D2-like receptors on rat thymocytes and rat peripheral lymphocytes (Mignini et al., 2013). In cells derived from the blood of individuals with PD, the levels of D1-like receptors are increased in total CD4+ T-cells and naïve CD4+ T-cells relative to those of healthy patients, and a decrease in D1 expression was associated with increased motor dysfunction (Kustrimovic et al., 2016). The variations in dopamine receptor expression and dopamine levels across T-cell subsets, as well as the release of dopamine in response to multiple types of stimuli, suggest that dopamine influences multiple T-cell functions and that this depends on the type of T-cell, the activation state, and local environment.
For example, in mice, dopamine (10−5 and 10−7M) downregulates the suppressive activity of Tregs but does not affect the D1-mediated response of T-effector cells to regulatory cell suppression, which may be mediated by ERK signaling (Kipnis et al., 2004). D1-like receptor expression was also associated with reduced Treg function in human CD4+ Tregs (Cosentino et al., 2018). However, adoptive transfer of T-cells from Drd5 knockout mice showed that D5-specific signaling enhanced the suppressive activity of murine Tregs in vivo and increased the expression of glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) (Osorio-Barrios et al., 2018). Thus, broad stimulation of D1-like receptors reduced the suppressive activity of Tregs, but specific signaling through D5 enhanced this function.
Further, while adoptive transfer experiments showed that D5 signaling drives Treg suppressive functions, in naïve CD4+ T-cells, D5 signaling increased proliferation and increased the differentiation of the Th17 inflammatory phenotype in the early stages of EAE (Osorio-Barrios et al., 2018). In contrast, when T-cells derived from individuals with lung carcinoma and those undergoing stress were treated with dopamine, it significantly inhibited the proliferation of human CD4+ and CD8+ T-cells and significantly reduced CD8+ T-cell cytotoxicity through D1-mediated cAMP signaling (Saha, Mondal, Basu et al., 2001, Saha, Mondal, Majumder et al., 2001). D1-like receptor activation with SKF-38393 also decreased the suppressive activity of human CD8+ Tregs (Nasi et al., 2019).
These differences in dopamine signaling may also regulate T-cell differentiation, and D3 and D5 seem to play particularly important roles. In naïve CD4+ T-cells, D3 signaling shifts the inflammatory balance toward the Th1 and Th17 phenotypes, and antigen recognition by naïve CD4+ T-cells induces D3 expression (Contreras et al., 2016). D3 stimulation in human T-cells also promotes integrin activation and the expression of IFN-γ and TNF-α (Levite et al., 2001; Ilani et al., 2004; Besser et al., 2005), indicating that D3 mediates increases in the number of Th1 cells. This finding was confirmed in two studies using murine CD4+ T-cells, which suggested that D3 mediates the increase in Th1-differentiation by reducing ERK phosphorylation, while a D3-mediated reduction in cAMP levels also increased CD4+ T-cell activity (González et al., 2013; Franz et al., 2015). One of these studies also showed that D5 signaling mediated increases in ERK phosphorylation and T-cell activity (Franz et al., 2015). Although this effect was not specific to D5, stimulation of D1-like receptors on human lymphocytes increased cAMP production and enhanced T-cell proliferation more than D2 receptor stimulation (Cosentino et al., 2018). However, D3 and D5 were not the only receptors affecting T-cell differentiation, as ablation of D2 receptors in a murine MPTP model of PD also increased Th1 and Th17 polarization of CD4+ T-cells in vivo. Treatment of isolated splenic CD4+ T-cells in vitro with sumarinole (D2 agonist), also blocked Th1 and Th17 polarization, and this effect was blocked by D2 antagonism with L-741,626 (Liu, Zhai et al., 2021). This differs from what is seen with D3 signaling, suggesting distinct roles among dopamine receptor subtypes. Finally, in naïve murine CD4+ T-cells, stimulation of D4 induced IL-2-STAT5 signaling, which promoted a Th2 phenotype. This effect was blocked by specific D4-inhibition with L-745,870 and confirmed in Drd4−/− mice, which showed profound age-dependent reduction in Th2 inflammation with altered allergen exposure response in the lung of neonatal animals (Wang, Cohen et al., 2019).
These data suggest that all types of dopamine receptors can activate T-cells, particularly through D3 and D5, but dopamine receptor activation more often inhibits activated immune cells. Dopamine alone activates naïve human T-cells, and CD8+ T-cells seem to be more responsive to dopamine-mediated inhibition than CD4+ T-cells. Dopaminergic regulation of T-cells is important in many disease processes such as skin inflammation or psoriasis (Keren et al., 2019) and PD (González et al., 2013; Ambrosi et al., 2017). We will expand on how the mechanisms discussed in this section can be applied to disease pathophysiology later in this review.
b. B-Lymphocytes
There is historically far less research on the role of dopamine in B-cells than on almost any other immune cell type, but recent publications in this area demonstrate a role for dopamine in B-cell immune function. These cells express TH and can store and produce dopamine. Additionally, in activated B-cells, increased production of endogenous catecholamines, including dopamine, norepinephrine and epinephrine, is associated with autocrine activation of β-adrenergic receptors and increased IL-10 production (Ferrari et al., 2004; Honke et al., 2022). This is further supported by the finding that Tfh, upon interaction with B-cells, release stored dopamine to facilitate Tfh–B-cell interactions and accelerate germinal center output (Papa et al., 2017). B-cells also express dopamine receptors on their surface (Santambrogio et al., 1993; McKenna et al., 2002; Meredith et al., 2006; Wei et al., 2016), indicating that these cells can also respond to dopamine. Treatment with dopamine (10−9M–10−6M), SKF-38393 (D1-agonist), and LY171555 (D2-agonist) all increased LPS-induced murine splenocyte proliferation (Tsao et al., 1997), suggesting that dopamine receptor signaling could drive B-cell proliferation. However, in B-cell neoplasia, dopamine toxicity may target cycling B-cells independent of their receptors to induce cell cycle arrest and apoptosis (Meredith et al., 2006). This suggests that dopamine regulates B-cell activity via both receptor dependent and independent mechanisms.
The D2-like receptors, particularly D3, may have an important role in B-cell immunity. In a rodent model of EAE requiring effective antigen presentation, the number of D3+/CD20+ B-cells (antigen-presenting B-cells) was increased in the CNS due to increases CNS tropism mediated by upregulation of CXC chemokine receptor 3 (CXCR3). Further, this subset of cells seems important to the onset of EAE, because knocking out Drd3 in B-cells completely abrogated disease symptoms. Expression of CXCR3 is increased in the CNS of patients with MS, and natalizumab, which blocks lymphocyte entry into the CNS, reduces CXCR3+ B-cells in MS brains. However, in a separate EAE model that does not require antigen-presentation, Drd3 knockout in B-cells exacerbated disease progression and increased the number of inflammatory CD4+ T-cells in the CNS. This suggested that D3 activation had discrete effects on antigen presenting versus nonantigen-presenting, anti-inflammatory B-cell subsets, demonstrating the importance of dopamine in regulating the function of several types of B-cells (Prado et al., 2021). In another study, expression of D2 on peripheral B-cells was negatively correlated with plasma TNF-α levels in patients with RA, suggesting dopamine receptors could influence disease progression by modulating inflammation (Wei et al., 2016). These data indicate an important role for dopamine receptors in the functional regulation of distinct B-cell subsets. Further studies are necessary to better understand the role of other dopamine receptors on B-cells and to determine how these functions influence the behavior of B-cell subsets.
D. Immunomodulatory Effects of Dopamine
1. Inflammatory Regulation of Dopaminergic Machinery
As dopamine receptors, transporters, and metabolic machinery are expressed in nearly all immune cells, it is not surprising that there is significant crosstalk between the dopaminergic system and the immune function of these cells. While dopamine can modulate the responses of immune cells to inflammatory insults, inflammatory factors released from different immune cells can also impact dopaminergic machinery, altering the function and expression of dopamine receptors and impacting dopamine metabolism. In the CNS, cytokines and chemokines influence the development, maintenance, and functional properties of midbrain dopaminergic neurons (Zalcman et al., 1994; Ling et al., 1998; Dunn, 2006; Felger and Miller, 2012). Inflammatory cytokines such as IL-1β promote the differentiation of mesencephalic progenitor cells into dopaminergic neurons (Ling et al., 1998), and IL-6 modulates dopaminergic activity in various mesolimbic structures in a dose-dependent manner (Zalcman et al., 1994; Dunn, 2006).
Inflammation can also regulate dopamine synthesis and release. A number of studies show NO induced changes in dopamine regulation in different brain regions (Lorrain and Hull, 1993; Seilicovich et al., 1995; Hartung et al., 2011; Motahari et al., 2016). Acute exposure to IL-1β and IL-6 can stimulate TH expression and dopaminergic activity in vivo and in vitro (Abreu et al., 1994; Zalcman et al., 1994; Ling et al., 1998). In rice stem borer (Chilo suppressalis) hemocytes, exposure to LPS stimulates dopamine synthesis (Wu et al., 2015). Inflammatory cytokines also impair the conversion of phenylalanine to tyrosine (Felger et al., 2013), potentially limiting tyrosine availability for dopamine synthesis. In addition to altering the biosynthesis of dopamine, some cytokines can regulate dopamine storage. IL-1β and TNF-α can decrease the expression of VMAT2, thereby limiting the availability of presynaptic dopamine. Conversely, TGF-α increases VMAT2 expression, favoring the storage of presynaptic dopamine (Kazumori et al., 2004). In hMDMs, DAT is highly responsive to inflammatory stimuli, as LPS treatment reduces DAT-mediated uptake and increases DAT-mediated dopamine efflux by promoting an efflux-favoring conformation in these cells. These activities were shown to be part of a TLR4-dependent autocrine loop by which DAT dynamically regulates the amount of dopamine in the local microenvironment to modulate immune functions such as cytokine release and phagocytosis (Mackie et al., 2022).
These results indicate that inflammatory cytokines can exert complex regulatory effects on the amount of dopamine available in dopaminergic cells by modifying the biosynthesis rate and the storage of this neurotransmitter. Finally, it is possible that oxidative stress dysregulates dopamine receptors. In rat renal proximal tubes, oxidative stress caused hyperphosphorylation of DR1, leading to uncoupling of the receptor and Gαs via NF-κB (Banday et al., 2007; Fardoun et al., 2007). This effect was also induced by the TLR4 agonist LPS and may be one mechanism by which dysregulated dopamine receptor signaling can lead to hypertension (Wang, Luo et al., 2014; Yang, Villar et al., 2021). Together, these data support a bidirectional interaction between the immune system and dopamine metabolism, uptake, storage, and receptor signaling.
2. Dopamine-Mediated Impacts on the Inflammatory Response
Multiple studies have demonstrated that dopamine can impact the production of inflammatory mediators in various cell systems, although the precise receptors and signaling pathways involved in this process are complex and not well understood (Tarazona et al., 1995; Hasko et al., 1996, 2002; Capellino et al., 2010; Nakano et al., 2011; Gaskill et al., 2012; Zhang et al., 2015, Zhang, Jiang 2016; Arreola et al., 2016; Nolan and Gaskill, 2019; Yoshioka et al., 2020). These effects are broadly summarized in Table 1. As previously discussed, innate immune cells recognize PAMPs and DAMPs via PRRs. Dopamine often acts on pathways downstream of several of these PRRs, so a baseline understanding of their signaling processes is important for understanding dopamine signaling in the immune response. TLRs coordinate innate immune functions and initiate adaptive immune responses through activation of the NF-κB/activator protein 1 and/or IRF3/7 pathways. Similarly, NOD-like receptor and C-type lectin receptor (dectin-1) signaling activates NF-κB and inflammasomes during bacterial and antifungal infections, respectively. Antiviral immunity is most often mediated by the upregulation of type I interferons via cytoplasmic RIG-I/MDA5, which is a cytoplasmic caspase-recruiting domain helicase (Lee and Kim, 2007). Several of these signaling pathways converge on NF-κB and its downstream effectors. Production of many cytokines is regulated by NF-κB transcriptional activity (Liu et al., 2017), and NF-κB activation is the first step in inflammasome activation and the secretion of IL-1 cytokines in a variety of cell types (He et al., 2016; Herman and Pasinetti, 2018). Signaling pathways involve numerous secondary messengers, kinases, and other signaling effectors (Kawai and Akira, 2009; Takeuchi and Akira, 2010), many of which can be activated by distinct dopamine signaling pathways (Beaulieu and Gainetdinov, 2011; Beaulieu et al., 2015), demonstrating substantial crosstalk between PRRs and dopamine signaling.
Dopamine-mediated immune function
The transcription factor NF-κB is central to the initiation and regulation of PRR-mediated inflammatory responses (Liu et al., 2017), and the impact of dopamine on NF-κB has been examined in a variety of immune cell subsets (Takeuchi and Fukunaga, 2003; Zhang et al., 2015; Nolan et al., 2019; Wang, Chen et al., 2019; Wu et al., 2020; Yoshioka et al., 2020; Yue et al., 2021). A number of these studies suggest that dopamine inhibits NF-κB, indicating anti-inflammatory activity. In the macrophage RAW264.7 cell line, very high levels of dopamine (10−3–10−5 M) reduced both LPS-induced expression of NO synthase and the expression of the NLRP3 inflammasome components NLRP3 and caspase-1 (Liu and Ding, 2019). Although this study did not directly demonstrate that dopamine impacted NF-κB, activation of this transcription factor is necessary for NLRP3 expression, and so the downregulation of NLRP3 indicates reductions in NF-κB activity (He et al., 2016; Herman and Pasinetti, 2018). Rotigotine (D2-like agonist) reduced TNF-α production and NF-κB activation in a mouse model of acute liver injury (Yue et al., 2021), and D2 signaling inhibited NF-κB activation by inhibiting Akt in a murine model of acute pancreatitis (Han et al., 2017). These effects are not unique to myeloid cells, as D2 activation in resting peripheral human T-cells was associated with IL-10 secretion, which regulates cytokine production via NF-κB (Besser et al., 2005). In CD3-stimulated human T-cells, D2 and D3 activation inhibited the dopamine-mediated release of IL-2, IFN-γ, and IL-4 (Ghosh et al., 2003). In a separate study, D4 blocked IL-2 production in CD3/CD28-stimulated human T-cells (Sarkar et al., 2006), indicating that under certain conditions, the activation of all three D2-like receptors can be anti-inflammatory. Dopamine treatment of astrocytes may also inhibit NF-κB and the NLRP3 inflammasome through D2-like receptor signaling (Shao et al., 2013; Zhu, Hu et al., 2018).
Dopamine signaling through D1-like receptors may also reduce NF-κB activity, potentially via PP2A activation (Wu et al., 2020), which is also linked to Akt inhibition (Manning and Toker, 2017). Dopamine (∼6–18 × 10−5M) also decreased inflammatory cytokine production and NLRP3 activity in murine lung tissue in ventilator-induced lung injury models (Yang et al., 2017). Treatment with A68,930 (D1-like agonist) inhibited NLRP3 activation in doxorubicin-treated murine cardiac myoblasts, as well as in mice with cardiac inflammation induced by doxorubicin, CNS inflammation induced by intracerebral hemorrhage, and acute kidney injury caused by renal ischemia/reperfusion (Wang, Nowrangi et al., 2018; Cao et al., 2020; Liu, Jin et al., 2021).
In murine BMDMs treated with the TLR2 ligand Pam3CSK4, D5 activation mediated the downregulation of NLRP3 (Wu et al., 2020), and signaling through D1 decreased NLRP3 activity by increasing NLRP3 ubiquitination in LPS-induced dopamine treated (1.5–2.5 × 10−4M) cells (Yan et al., 2015). These studies show that high levels of dopamine or specific D1-like receptor activation in rodents and rodent macrophages can downregulate NLRP3 activity. Several studies also suggest that this effect is mediated by cAMP signaling, although D1 signaling was not shown to act directly on cAMP production (Yan et al., 2015; Wang, Villar et al., 2018; Liu, Jin et al., 2021). The connection may therefore be indirect or circumstantial, as data in human macrophages suggest that D1-like receptors do not activate cAMP in this cell type (Nickoloff-Bybel et al., 2019).
Although these aforementioned studies showed an anti-inflammatory effect of D1-like receptor signaling, they examined the effects of high levels of dopamine or D1-specific agonists on existing inflammation. A number of other studies indicate an inflammatory role for dopamine, which can also be mediated by NF-κB and NLRP3 activity (Nakano et al., 2008, 2011; Gaskill et al., 2012; Parrado et al., 2012; Trudler et al., 2014; Wang, Villar et al., 2018; Nolan et al., 2019, 2020). In hMDMs, dopamine (10−8–10−6M) increased the production of the inflammatory mediators CCL2, IL-6, C-X-C motif chemokine ligand (CXCL) 8, IL-1β, CXCL9, and CXCL10 (Gaskill et al., 2012; Nolan et al., 2019). Similarly, dopamine (2 × 10−6M–10−5M) increased IL-6 and IL-1β production in activated primary murine microglia, NK cells, and BV-2 murine microglial cells (Trudler et al., 2014; Fan et al., 2018; Nolan et al., 2019), and one study showed increased ERK1/2 and p38MAPK activation in LPS-induced rodent primary microglia and BV2 cells relative to resting cells (Fan et al., 2018). IL-1β production was also increased in LPS-induced peritoneal macrophages (Kawano et al., 2018). In hMDMs, specifically blocking DAT activity enhanced LPS-mediated production of IL-6, TNF-α, and mitochondrial superoxide levels, showing that the regulation of dopamine uptake and release, as well as dopamine receptor activity, plays a role in dopaminergic immunomodulation in macrophages (Mackie et al., 2022).
These effects are not limited to myeloid cells. In naïve human CD4+ T-cells stimulated with anti-CD3/anti-CD28, dopamine (10−8–10−7M) increased the production of IL-5, IL-17, IL-1β, and IL-6 and the dopamine-mediated changes in IL-5 and IL-17 were blocked by the presence of SCH23390 (D1-like receptor antagonist) (Nakano et al., 2011). The secretion of TNF-α by resting human peripheral T-cells also increased in response to D1/D5 and D3 activation (Besser et al., 2005). Activation of D3 also drives inflammatory effects in several other human and rodent systems, biasing the generation of inflammatory Th1 and Th17 cells and suppressing the development of Th2 cells (González et al., 2013; Ilani et al., 2004; Contreras et al., 2016). In a mouse model of intestinal inflammation, the transplantation of naïve Drd3−/− T-cells resulted in milder weight loss and mucosal inflammation than the transplantation of wild-type cells (Contreras et al., 2016). In an MPTP model of PD, Drd3 knockout animals showed decreased microglial activation and reductions in TNF-α and IFN-γ production due to a lack of D3 signaling in T-cells (González et al., 2013). In addition to traditional immune cells, in human adipocytes, pharmacological stimulation of D2 induced the expression and release of IL-6 (Wang, Villar et al., 2018), and in human keratinocytes, D2 activation increased IL-6 and IL-8 levels (Parrado et al., 2012).
Inflammatory responses were also suppressed by SCH23390 in a SCID-mouse model of murine RA, while D2-like receptor suppression with haloperidol increased the accumulation of IL-6+ and IL-17+ T-cells (Nakano et al., 2011). In a murine model of EAE, SCH23390 (D1-like receptor antagonist) decreased IL-17 production (Nakano et al., 2008), and in a different murine model of EAE, dopamine was found to exacerbate the disease, as removal of dopamine reduced EAE severity. Much of this effect was found to be driven by D5 signaling in DCs, which increased the frequency of inflammatory CD4+ T-cells in the CNS and drove inflammation by blocking signal transducer and activator of transcription and inducing the activity of IL-12 and IL-23 in T-cells (Prado et al., 2018). Further, dopamine (10−6M–10−5M) increased the anti-inflammatory mediator IL-10 in LPS-induced hMDMs (Gaskill et al., 2012; Nolan et al., 2019), LPS-induced primary murine splenocytes, B-cells, and bone marrow–derived DCs (Kawano et al., 2018), although the effect of A77636 (D1-like receptor agonist) on splenocytes was inhibitory (Kawano et al., 2018).
Many studies of innate immune cells such as macrophages indicate that the regulation of NF-κB and the NLRP3 inflammasome is central to the inflammatory impact of dopamine. In hMDMs, dopamine increases the activation of NF-κB, leading to the expression of NLRP3 and IL-1β. Dopamine could also potentiate ATP-induced secretion of IL-1β but did not mediate the secretion of IL-1β on its own, which indicates that dopamine can prime but not activate the NLRP3 inflammasome by activating NF-κB (Nolan et al., 2020). Many previously discussed studies also show that dopamine or specific dopamine receptors increase and decrease NF-κB and NLRP3 inflammasome activity (Nakano et al., 2011; Shao et al., 2013; Trudler et al., 2014; Yan et al., 2015; Yamamoto et al., 2016; Wang, Nowrangi et al., 2018; Wu et al., 2020; Liu, Jin et al., 2021).
These data, along with other, similar research, indicate that NF-κB and the NLRP3 inflammasome may be central to dopaminergic modulation of inflammation, at least in macrophages and other types of innate immune cells. Notably, most studies only examined the NLRP3 inflammasome, and the effects of dopamine on other inflammasomes remain unclear. These data show the broad range of cell types on which dopamine can act and highlight the discrete roles of specific dopamine receptor subtypes, as well as how they could differ between cell types. While many of these data are conflicting, they demonstrate that dopamine acts differently on activated or stimulated cells than on unstimulated cells, mediating anti-inflammatory effects in the presence of inflammation and driving inflammation in non-inflammatory conditions.
It is likely that D1- and D2-like receptors mediate distinct effects, particularly across different cell types and activation states. The hypothesis that D1-like receptors are inflammatory and the D2-like receptors are anti-inflammatory is very appealing (Nolan and Gaskill, 2019) and is supported in many disease models, as well in a number of in vitro studies (Nakano et al., 2008; Nakano et al., 2011; Shao et al., 2013; Zhang et al., 2015; Osorio-Barrios et al., 2018; Prado et al., 2018; Zhu, Hu et al., 2018; Alam et al., 2021; Yue et al., 2021), many of which are discussed in the previous text. In hMDMs, there is a negative correlation between D1-like receptor expression and the anti-inflammatory cytokine IL-10, whereas dopamine increased IL-1β production in hMDMs lacking D2 (Nolan et al., 2019), and D1-like receptor signaling can increase TNF-α production in resting human CD4+ T-cells (Besser et al., 2005). Pharmacological inhibition of D1-like receptors using SCH23390, or knockout of Drd5 specifically on DCs reduced inflammation in an EAE model (Prado et al., 2018), antagonizing D1-like receptors also suppresses Th17 and neutrophilic lung inflammation signaling (Nakagome et al., 2011), both suggesting that D1-like receptor signaling drives inflammation.
In further support of this hypothesis, D2 activation was anti-inflammatory in rodent models of pancreatitis (Han et al., 2017) and acute liver injury (Yue et al., 2021). D2 activation increased the secretion of the anti-inflammatory cytokine IL-10 in human T-cells (Besser et al., 2005), and silencing D2 receptors reduced renal inflammation, which was associated with increased Akt phosphorylation and decreased PP2A activity, suggesting that D2R signaling may act through the activation of PP2A and subsequent inhibition of Akt (Zhang, Jiang et al., 2016). In rodent models of brain injury, the D2 agonist quinpirole protected against glial cell–induced neuroinflammation by inhibiting NF-κB and the activation of αB-crystalline (Zhang et al., 2015; Alam et al., 2021). The precise cell type(s) affected by dopamine were not investigated in this study, but subsequent studies showed that the activation of astrocytic D2-like receptors induces αB-crystalline to alleviate neuroinflammation in response to MPTP-mediated injury and in stroke models (Shao et al., 2013; Qiu et al., 2016). Moreover, activation of astrocytic D2-like receptors inhibited NLRP3 inflammasome activation via a β-arrestin mediated mechanism (Zhu, Hu et al., 2018). While the PP2A/AKT axis has not been examined in astrocytes, β-arrestin-mediated recruitment of PP2A is a major mechanism by which D2-like receptors inhibit Akt (Beaulieu et al., 2005, 2015; Beaulieu and Gainetdinov, 2011). This finding suggests an important role of this signaling axis in mediating the anti-inflammatory effects of D2-like receptors.
While these studies and others support the inflammatory D1–anti-inflammatory D2 dichotomy, this concept is not consistent throughout the literature. For example, A68,930 (D1-like receptor agonist) inhibited NLRP3 activation in mouse lung (Jiang, Li et al., 2016), brain (Wang, Nowrangi et al., 2018), and kidney (Cao et al., 2020). Treatment with A77636 also inhibited inflammation in mouse splenocytes (Kawano et al., 2018), and while the effect was not specifically inflammatory, D5 signaling mediated an increase in T-cell activity (Franz et al., 2015). Extensive mechanistic studies show that both types of D1-like receptors individually inhibit NLRP3 activation in several types of activated murine macrophages (Yan et al., 2015; Liu and Ding, 2019; Wu et al., 2020; Liu, Wu et al., 2021). In the NG108-15 rodent glial cell line, D1-like signaling was also associated with the inhibition of NF-κB through PKA-mediated mechanisms, whereas D2-like activation was associated with the activation of NF-κB (Takeuchi and Fukunaga, 2003). D2 activation also increased the release of inflammatory cytokines by human adipocytes (Wang, Villar et al., 2018) and human keratinocytes (Parrado et al., 2012).
While it is possible that some of these differences may be associated with subtype-specific signaling (D5 is anti-inflammatory and D1 is proinflammatory), this seems unlikely, as both D1 (Yan et al., 2015) and D5 (Wu et al., 2020; Liu, Wu et al., 2021) have been shown to decrease inflammation in mouse model systems. Additionally, dopamine receptors of the same subtype can have distinct effects on the same cells, as D2 activation increases IL-10 in resting human T-cells, while D3 activation increases TNF-α in the same population (Besser et al., 2005). This bifurcation of dopamine-mediated effects on a dopamine receptor subtype seems to be particularly common in T-cells, in which D3 is more inflammatory and D2 is more anti-inflammatory, although this effect is clearly influenced by activation state. There may also be species-specific differences, such as dopamine being more inflammatory in human myeloid cells than rodent myeloid cells, but many additional studies of human immune cells are needed to determine this effect with any certainty. These data suggest that the effects of dopamine receptor signaling are heavily influenced by existing levels of inflammation and likely have distinct effects on different cell types.
3. Dopaminergic Regulation of Phagocytosis
Phagocytosis is a critical immune function in which cells engulf pathogens or debris and, in the case of pathogens, kill them through intracellular or extracellular mechanisms. Phagocytosis is primarily mediated by professional phagocytes, including monocytes, macrophages, DCs, and eosinophils, although most cell types have the capacity for some kind of phagocytosis. A wide range of studies across species indicate that dopamine can modulate phagocytic activity, but the precise mechanisms by which dopamine mediates these effects seem to differ across species. In Litopenaeus vannamei, a species of white prawn, dopamine (10−7M–10−6 M) significantly decreased gene expression of Drd4, Gi heterotrimeric G protein, and exocytosis-related proteins, while phagocytosis-related proteins and Gs heterotrimeric G protein were increased (Tong et al., 2020). In the rice stem borer (Chilo suppressalis), dopamine (10−5M) increased hemocyte phagocytosis via a D1-like receptor. This effect appeared to involve ERK1/2 activation (Wu et al., 2015), which can be induced by dopamine via cAMP/PKA signaling (Zhen et al., 1998; Gerits et al., 2008; Shao et al., 2020). While the implications of these findings to human subjects is unclear, these data suggest that the capacity of dopamine to stimulate the D1–cAMP axis may promote the phagocytic function of immune cells via the Gs heterotrimeric G protein pathway, which signals through the activation of adenylate cyclase and the production of cAMP (Marinissen and Gutkind, 2001).
Studies also suggest that dopamine modulates phagocytic activity in a number of vertebrate systems, although the mechanisms underlying this activity are not clear. In wall lizard splenic macrophages, dopamine induced bimodal changes in phagocytosis; higher concentrations of dopamine (10−7–10−5M) could decrease phagocytosis, while very low dopamine concentrations (10−11–10−15M) could increase phagocytosis (Roy and Rai, 2004). Dopamine (10−8–10−7M) increased the phagocytic activity of murine BMDMs in the presence of IFN-γ, as well as in neutrophils (Sookhai et al., 1999), although another study showed that similar concentrations of dopamine (6.5 × 10−7M) reduced neutrophil phagocytosis (Wenisch et al., 1996). In mice, macrophage phagocytosis was decreased by spiperone (D2-like receptor antagonist) (Sternberg et al., 1987). Chicken macrophages treated with dopamine (0.65–1.6 × 10−6M) and guinea pig macrophages treated with bromocriptine (D2 receptor agonist) and pergolide (D2 receptor agonist) both showed an enhanced binding and phagocytosis of IgG-sensitive red blood cells (Ali et al., 1994; Gomez et al., 1999).
In each of these studies, changes in phagocytic activity were associated with dopamine receptor-induced changes in the expression of surface Fc-gamma receptor (Fcγ) receptors, which are critical regulators of the immune response that mediate a number of factors including antigen uptake (Sternberg et al., 1987). Dopamine-mediated changes in Fcγ receptors could not only disrupt phagocytosis and antigen presentation but also suggest a mechanism by which dopamine affects T-cell activation and promotes the development of autoimmunity (Junker et al., 2020). A D2-mediated effect on phagocytosis was seen in rodent peritoneal macrophages, but antagonizing D2-like receptors with domperidone increased phagocytosis in lactating rats (Carvalho-Freitas et al., 2008). Unlike studies showing effects through regulation of Fcγ expression, this effect was thought to be mediated by the regulation of serum prolactin levels, and phagocytosis was shown to be decreased at other time points in another study by this group (Carvalho-Freitas et al., 2008, 2011). Dopaminergic regulation of phagocytosis was also seen in hMDMs when DAT activity was blocked with nomifensine, downregulating LPS-induced phagocytic activity via autocrine signaling and suggesting that lower dopamine levels inhibit phagocytic activity in activated macrophages (Mackie et al., 2022). A decrease in phagocytosis, as well as ERK1/2 phosphorylation, was also seen in in response to dopamine (2 × 10−6M) treatment of LPS-activated BV2 microglia, but no effect on the phagocytic activity of resting cells was observed, suggesting autocrine control of phagocytosis by dopamine signaling in activated myeloid cells (Fan et al., 2018).
These differences could be due to the different mechanisms through which phagocytosis is regulated, such as Fcγ receptors, prolactin levels, or DAT/MAPK. Lower dopamine concentrations (≤10−6M) and D2 activation increased phagocytosis (Sternberg et al., 1987; Ali et al., 1994; Gomez et al., 1999) and were associated with changes in Fcγ receptors, while reducing D2 activity increased phagocytic activity and was associated with prolactin (Carvalho-Freitas et al., 2008, 2011). However, phagocytic activity was also reduced by the pan-dopamine agonist apomorphine, the D2/D3 agonist bromo-a-ergocryptine, and the β-adrenergic receptor antagonist propranolol in wall lizard macrophages. These data studies suggest that D1-like receptors can also drive phagocytosis, and many of these data suggest the involvement of the cAMP signaling pathway, as D2 activation inhibits cAMP production.
In support of this, in the wall lizard cells, treatment with phosphodiesterase IBMX, which increases cAMP levels, resulted in a greater reduction in phagocytosis when combined with dopamine. Signaling through MAPK is also downstream of cAMP in a number of pathways. However, the use of the cAMP analog db cAMP had a similar biphasic effect (Roy and Rai, 2004), making the directional effects of higher and lower cAMP levels unclear. Moreover, dopamine-mediated cAMP activity was notably absent from human macrophages (Nickoloff-Bybel et al., 2019), suggesting that this pathway might not function in human cells. Taken together, these data form a confusing picture of dopaminergic modulation of phagocytosis, suggesting that the differences in the effects of dopamine are due to the concentration of dopamine or ligand being used, the exposure time, the receptor being activated, and the species. Thus, while these data indicate that dopamine does regulate phagocytic function in multiple cell types, additional work with carefully chosen and controlled experimental conditions is needed to precisely define the mechanism by which this regulation occurs.
4. Dopaminergic Regulation of Chemotaxis
Chemotaxis is the mechanism by which cells move in response to extracellular chemical gradients, allowing immune cells to migrate to sites of inflammation or tissue damage when they detect chemokines and other factors secreted by cells at that site. Chemotaxis involves many chemokines, some of which are species- or cell-specific, and is initiated when soluble chemokines bind to their cognate receptors on immune cells (Rossi and Zlotnik, 2000; Hughes and Nibbs, 2018). A number of studies have shown that dopamine can impact the production of chemotactic factors, particularly CCL2, which is important in the pathogenesis of many chronic inflammatory conditions, as well as neurologic HIV (neuroHIV) (Eugenin et al., 2006; Covino et al., 2016; Fantuzzi et al., 2019; Das et al., 2021). Dopamine increased the mRNA expression of CXCL8 and CCL2 in activated PBMCs (Torres et al., 2005) and the injection of a D1-like agonist increased the mRNA expression levels of CCL2 and C-C motif chemokine ligand 7 in the rodent PFC (Saika et al., 2018). In hMDMs, dopamine (10−6M–10−8M) increased the production of CCL2, CXCL8, CXCL9, and CXCL10 (Gaskill et al., 2012; Nolan et al., 2019), while blocking DAT activity potentiated LPS-induced CCL2 production (Mackie et al., 2022). Both CCL2 and CXCL2 were reduced by D2 signaling in acute pancreatitis (Han et al., 2017; Saika et al., 2018), and in mouse renal proximal tubule cells, D2 knockdown was associated with an increase in CCL2 production. This effect was connected to the loss of PP2A activity, suggesting that the D2-like receptors may inhibit cytokine production via PP2A and Akt inhibition (Zhang, Jiang et al., 2016). In addition to CCL2, in primary murine splenocytes, peritoneal macrophages and NK cells, dopamine (10−5–10−6M) increased CXCL1 production (Kawano et al., 2018).
In addition to increasing the production of chemokines, dopamine itself can act as a chemoattractant and potentiate chemotaxis and chemokinesis. In neutrophils, high concentrations of dopamine (10−4 and 10−5 M) inhibited IL-8 mediated transendothelial migration (Sookhai et al., 2000). In mature human CD14+CD16+ monocytes, dopamine (10−6M–5 × 10−5M) and SKF38393 (D1-like agonist) increased chemokinesis and transmigration across a model of the blood–brain barrier (Coley et al., 2015; Calderon et al., 2017), suggesting a chemotactic role for D1-like receptors. A role for D1-like receptors, specifically D5, was also shown in murine CD4+ T-cells in the gut, as the formation of a heteromeric CCR9:D5 complex on these cells mediated inflammation-induced T-cell migration into the gut mucosa. These effects were mediated by ERK1/2 signaling and were specific to CCR9, as D5 did not form heteromers with other chemokine receptors (Osorio-Barrios et al., 2021).
D2-like receptor signaling has also been implicated in dopamine-mediated chemotaxis. In mice, D3 stimulation of Tregs reduced CCR9 expression and inhibited the migration of these cells into the lamina propria region of the gut upon intestinal inflammation (Ugalde et al., 2021). In naïve CD8+ T-cells, dopamine (10−7M) potentiated homing to CCL19, CCL21, and CXCL12 through D3 and Gαi-mediated Ca2+ mobilization (Watanabe et al., 2006), and treating peripheral T-cells from head and neck cancer patients with dopamine (10−8M) increased spontaneous migration and migration toward CXCL12 (Saussez et al., 2014). A role for D2-mediated Gαi signaling is supported by experiments using HEK293 cells transfected with D2-receptors. Treating these cells with quinpirole (D2-agonist) mediated a chemotactic response to IL-8 that required the activation of Gαi and Gβγ activity, while Gαs and Gαq receptors did not mediate these effects (Neptune and Bourne, 1997).
In addition to driving immune cell chemotaxis, dopamine can also influence inflammation and immune activity by modulating the movement of bacteria and pathogens. Antagonizing D1- and D2-like receptors blocked liver fluke (Clonorchis sinensis) larva chemotaxis into the bile duct, indicating that dopamine plays a role in the development of clonorchiasis (Dai et al., 2020). High levels of dopamine (5 × 10−5M) also increased the expression of flagellar motility-related genes and the swimming motility of the aquatic bacterium Vibrio harveyi, a Gram-negative bacterium that is a major pathogen of all aquatic organisms. This effect was blocked by chlorpromazine, suggesting that this effect was driven by D2-like receptors, although the effect could be influenced by the many effects of chlorpromazine on nondopaminergic receptors (Yang, Anh et al., 2014). Dopamine has biphasic effects on the chemotaxis of E. coli, repelling these bacteria at concentrations below 10−4M but attracting E. coli at higher concentrations. Although gut concentrations of dopamine are <10−4M at most times, higher concentrations are likely present in the microenvironment around the gut lumen and at the mucous layer where these compounds are secreted, and different foods could also elevate gut dopamine levels (Lopes and Sourjik, 2018; Matt and Gaskill, 2020).
Taken together, these data implicate several signaling pathways in the regulation of dopaminergic chemotaxis and suggest that many of the chemotactic effects of dopamine could be mediated through CCL2, CXCL9, and CXCL12. It is difficult to identify patterns relating to cell type, as D1-like activity promoted chemotaxis in monocytes, T-cells, and E. coli, while D2-like receptors drove chemotaxis in T-cells and other pathogens. This indicates that both D1- and D2-like receptor signaling can play a role in chemotactic activity. Pathways mediated by Gαi, Gβγ, and Akt primarily associated with D2-like receptors, while ERK1/2 and Ca2+ flux could be mediated by either receptor subtype. As with many functions, these effects are likely different in distinct cell types and activation states, but the current data make it difficult to further define this, as both types of receptors act on multiple cell types in both resting and activated states.
5. Dopaminergic Regulation of Oxidative Burst and Nitric Oxide Production
In addition to regulating the production of inflammatory mediators, phagocytic activity, and chemotaxis, dopamine also influences respiratory or oxidative bursts, as well as ROS and NO production. These bursts mediate the rapid release of ROS such as hydrogen peroxide (H2O2) and superoxide anion, which can be formed by the activity of superoxide dismutase. This process is common in phagocytes, such as myeloid cells and granulocytes, to degrade internalized bacteria and other particles as part of the immune response. These bursts can also affect cell signaling (Forman and Torres, 2002; Dahlgren and Karlsson, 1999). In neutrophils, dopamine may reduce respiratory bursts, as dopamine treatment (6.5 × 10−7M) of human neutrophils reduced ROS production (Wenisch et al., 1996), and high concentrations of dopamine (10−4 and 10−5 M) inhibited superoxide anion production in response to N-formylated N-formyl-methionyl-leucyl-phenylalanine stimulation (Yamazaki et al., 1989; Matsuoka, 1990). Similarly, high levels of dopamine (10−4M) and fenoldopam (D1-like agonist) reduced respiratory bursts in neutrophils isolated from the blood of patients with systemic inflammatory response syndrome (Sookhai et al., 1999). Lower concentrations of dopamine (2.61 × 10−7M or 2.61 × 10−10M) had no effect on the formation of H2O2-mediated oxidative bursts in human neutrophils (Trabold et al., 2007).
In myeloid cells such as rodent peritoneal macrophages, antagonizing D2-like receptors with domperidone increased spontaneous oxidative bursts of H2O2 (Carvalho-Freitas et al.,2008, 2011). Additionally, dopamine treatment (10−6–10−8M) of hemocytes from the white shrimp Litopenaeus vannamei significantly reduced respiratory bursts and superoxide dismutase activity (Cheng et al., 2005). These data suggest that dopamine generally inhibits the production of ROS and oxidative bursts in neutrophils, particularly at higher levels. This is supported by recent data showing exposure to dopamine (10−6–10−9M) broadly inhibited neutrophil activity in a D1-dependent manner (Marino et al., 2022). Further, data from hypertension studies showing that the activation of D1-like receptors, particularly D5, reduced the production of ROS in mitochondria through an autophagy-associated mechanism (Yang et al., 2006; Lee et al., 2021). In addition, studies showed that knocking out Drd2 in renal tubule cells increased renal ROS production (Yang, Cuevas et al., 2014) and that Drd2 knockout mice had increased levels of ROS due to aldosterone dysregulation (Armando et al., 2007).
Dopamine has also been shown to dysregulate NO production. NO is important in defense against infectious diseases, tumors, sterile inflammation, and other insults. NO is produced by several types of immune cells, mostly macrophages and granulocytes (Bogdan, 2001). In rodents, treatment with apomorphine (pan-dopamine receptor agonist) increased NO2− and NO3− in dialysate from the hypothalamus (Melis et al., 1996), suggesting that dopamine increases NO production. However, this conclusion is opposed by an array of in vitro studies. In LPS-induced primary rodent microglia and BV-2 cells, dopamine (2 × 10−6M) increased NO synthase (Fan et al., 2018). Inhibiting D1-like receptors with SCH23390 and D2-like receptors with sulpiride inhibited NO production by LPS-stimulated peritoneal macrophages (Hasko et al., 1996). Dopamine also enhanced LPS-mediated NO production in RAW264.7 cells, although only at higher concentrations (5 × 10−5M) and to a much lesser extent than other catecholamines (Chi et al., 2003). However, dopamine decreased the production of NO and NO synthase in LPS-stimulated primary rodent microglia as well as N9 and BV2 murine microglia through D1- and D2-like receptors (Chang and Liu, 2000; Farber et al., 2005; Yoshioka et al., 2016; Wang, Chen et al., 2019). These findings opposed other studies that suggested that dopamine increased NO production, but data from BV2 cells suggest that this could result from cytotoxicity due to the production of dopamine quinones (Beck et al., 2004) and may not be mediated by dopamine receptor signaling. These data suggest more research is needed in this area, but that dopamine receptor signaling may promote the production of NO in myeloid cells, although cytotoxic dopamine quinone formation may have the opposite effect.
6. Signaling Mechanisms Mediating Dopaminergic Immunomodulation
The discrete effects of specific dopamine receptors may also reflect D1- and D2-like receptor signaling through both convergent and divergent signaling cascades, depending on the cell system and dopamine receptors activated. For example, Akt inhibition via PP2A recruitment has been suggested to be central in dopamine-mediated anti-inflammatory effects in isolated cell systems and disease models (Tolstanova et al., 2015; Zhang, Jiang et al., 2016; Han et al., 2017; Wu et al., 2020; Yue et al., 2021). Signaling through Akt regulates a wide range of cellular processes, including cell growth, survival, metabolism, and inflammation (Manning and Toker, 2017). In the immune response, Akt modulates NF-κB activity by regulating IkappaB (IκB) kinase, which phosphorylates IκB to release and activate NF-κB (Dan et al., 2008; Cheng and Kane, 2013; Dorrington and Fraser, 2019). This suggests the dopamine-mediated inhibition of Akt could reduce inflammatory cytokine production. Akt inhibition is generally associated with D2-like receptor signaling (Beaulieu et al., 2004, 2007; Zhang, Jiang et al., 2016; Han et al., 2017; Zhu, Hu et al., 2018; Wu et al., 2020), but D1-like receptors can act on AKT (Beaulieu et al., 2005). The activity of this pathway, and the relative frequencies of D2- and D1-like receptors may explain the anti-inflammatory activity of these receptors in certain systems. Both D1- and D2-like receptors can also activate the phosphatidylinositol 3-kinase/Akt signaling cascade, although the immunologic impact of dopamine-mediated activation of this pathway is not well understood (Zhen et al., 2001; Brami-Cherrier et al., 2002; Nair et al., 2003; Nair and Sealfon, 2003; Iwakura et al., 2008; Mannoury la Cour et al., 2011; Chen, Ruan et al., 2012; Perreault et al., 2013; Radl et al., 2013; Mirones et al., 2014; Yan et al., 2020).
Some of the differences in the effects of dopamine on similar cell types may be due to variations in the capacity of D1-like receptors to activate the canonical D1–Gαs–cAMP signaling pathway. While studies have shown that cAMP signaling has both inflammatory and anti-inflammatory effects, in myeloid cells, cAMP signaling is largely associated with a decrease in inflammation (Serezani et al., 2008; Peters-Golden, 2009; Gerlo et al., 2011). Supporting this hypothesis, dopamine-mediated inhibition of NLRP3 in murine macrophages was linked to cAMP activation, and dopamine-mediated inhibition of NF-κB was associated with D1-mediated activation of PKA, a downstream effector of cAMP, in microglia and other cells lines (Takeuchi and Fukunaga, 2003; Yan et al., 2015; Wang, Chen et al., 2019). In contrast, in hMDM where dopamine activates NF-κB and primes the inflammasome (Nolan et al., 2020), D1-like receptors do not act through the Gαs-cAMP pathway (Nickoloff-Bybel et al., 2019). In T-cells, stimulation of D3 reduces cAMP, which increases naïve CD4+ T-cell activation (Franz et al., 2015), further suggesting that a lack of cAMP/PKA signaling supports dopamine-mediated inflammatory effects.
Differences in the capacity of distinct dopamine receptors to activate the MAPK cascade may also contribute to variations in the effects of dopamine. The MAPK signaling cascade modulates numerous cellular functions, including inflammatory cytokine release, in part via the activation of transcription factors such as activator protein 1 (Karin, 1995; Kaminska et al., 2009; Furler and Uittenbogaart, 2010; Cargnello and Roux, 2011; Kyriakis and Avruch, 2012). Dopamine can activate all members of the MAPK family through both D1- and D2-like receptors, although the precise mechanism is unclear and varies from system to system (Luo et al., 1998; Choi et al., 1999; Zhen et al., 2001; Beom et al., 2004; Wang et al., 2005; Lee et al., 2006; Huang et al., 2012; Franz et al., 2015). Many studies have connected the capacity of dopamine to regulate different members of this family with its effects on inflammation. In naïve CD4+ T-cells, D3 and D5 mediate ERK1/2 phosphorylation and promote the differentiation of inflammatory Th1 cells (Franz et al., 2015). However, in DCs, D5 stimulation downregulated LPS-induced ERK1/2 phosphorylation but did not affect JNK or p38 MAPK phosphorylation. The D5-mediated reduction in ERK1/2 was associated with decreased IL-12 and IL-23 production in specific DC subsets, suggesting an inhibitory effect (Prado et al., 2012). Similarly, in primary microglia and BV-2 microglia, dopamine downregulated ERK phosphorylation, while in resting microglia dopamine increased p38 MAPK activity (Fan et al., 2018). Interestingly, in rat astrocytes, D1-like activation of ERK1/2 was associated with cell migration (Huang et al., 2012), supporting differences in the effects of dopamine-induced MAPK signaling between disparate cell types. Indeed, studies have shown both cell-type and receptor-specific differences in D2-like receptor-mediated ERK activation (Beom et al., 2004; Wang et al., 2005). As with cAMP and Akt signaling, this suggests discrete effects of dopamine receptor subtypes on MAPK activation, which may mediate the effects of dopamine on the inflammatory responses in these cells.
Overall, the data discussed in this section of the review indicate that dopamine influences a wide array of immune functions in both an inflammatory and anti-inflammatory manner. Data on most of these functions is relatively sparse, owing to the large variations in experimental design (species, dopamine/ligand concentration, disease model) and potential cell type– and species-specific differences in dopamine receptor expression and signaling bias. Thus, there is no clear consensus on the roles of distinct dopamine receptor subtypes in mediating these effects. The hypothesis that D1-like receptors are inflammatory while D2-like are anti-inflammatory is not broadly supported by the data, although there are certainly specific cell types or diseases, which will be discussed in depth in the following sections, in which this may be the case. Evidence suggests that dopamine may be more anti-inflammatory in stimulated cells and inflammatory in resting cells, and studies in human cells generally show an inflammatory effect of dopamine, although this may be due to the smaller numbers of studies done in humans relative to other systems. Data also suggest that the effects of specific dopamine receptors may differ between cell types and among the same cell type in different species. Signaling pathways activated by dopamine receptors are also likely to overlap, and the activation of multiple dopamine receptor subtypes could interact and result in opposing, additive, or novel outcomes.
Moving forward, careful evaluation of dopamine receptor expression in each model system, defining both raw receptor expression and the ratios of different dopamine receptors, is important in precisely defining the receptors and signaling mechanisms that mediate a particular effect. Many areas warrant further investigation, including the understanding of dopamine release in immune cells, examination of cell type–specific dopamine receptor signaling and specific immune activity, and delineation of dopamine-mediated inflammation from inflammation mediated by pharmacologic drugs that modify dopamine signaling. Broadly, these data show the importance of dopamine in the regulation of immune function and the need for further studies with carefully considered experimental designs to better understand and leverage dopaminergic immunomodulation for disease treatment. In the subsequent sections, we will discuss how the immunomodulatory effects discussed here impact different organs and diseases, highlighting the potential for dopaminergic immunomodulation in the treatment of a wide array of pathogenic conditions.
IV. Regional and Disease-Specific Effects of Dopamine
A. Central Nervous System
1. Introduction
While further research on the signaling processes and cell type–specific effects induced by dopamine is important for understanding the basic processes that mediate dopaminergic immunomodulation, it is also important to consider the application of this research to specific pathologies. Fluctuations in dopamine and the dopaminergic system influence a wide variety of organ systems and diseases. Prominent among these are numerous neurologic conditions including PD and neuroHIV, neuropsychiatric conditions such as schizophrenia and depression, and developmental neurologic disorders such as ADHD and epilepsy. Many of the studies evaluating the impact of dopamine on these diseases can be found in Table 2. This is not an exhaustive list of neuropathologies that could be influenced by dopamine, and substantial research has been devoted to examining other neurologic conditions, such as MS (Pacheco et al., 2014; Levite et al., 2017) and Alzheimer’s disease (Thomas Broome et al., 2020).
Effects of dopaminergic immunomodulation associated with disease
Anatomic changes in dopaminergic brain regions, the dysregulation of dopamine transmission, and aberrant dopamine receptor activity are key aspects in the development or progression of these diseases (Rangel-Barajas et al., 2015). Changes in dopaminergic activity in these diseases could drive pathogenesis by influencing immune function, as the etiology of many of these conditions is associated with changes in immune function (Block and Hong, 2005; Rogers et al., 2007; Cai et al., 2014; Yirmiya et al., 2015; Zrzavy et al., 2017; Thawkar and Kaur, 2019; Cignarella et al., 2020; Deng et al., 2020; Haque et al., 2020; Kam et al., 2020; Zhang et al., 2020; Bido et al., 2021). This section examines these interactions, highlighting the potential crosstalk between pathologic changes in dopaminergic regulation and dopamine-mediated immunomodulatory changes associated with the development of several neurologic diseases. We also discuss the impact of dopamine-altering therapeutics on these pathologic processes, as research in these areas could guide treatment recommendations, suggest specific targets for drug repurposing and development, and improve overall therapeutic efficacy.
2. Parkinson’s Disease
PD is a progressive neurodegenerative condition that affects more than 6 million people globally (GBD 2016 Parkinson’s Disease Collaborators, 2018). It causes a variety of motor and nonmotor symptoms such as resting tremor, bradykinesia, rigidity, and gait and posture alterations, as well as cognitive impairment, anxiety, depression, sleep disturbances, and pain (Poewe et al., 2017; GBD 2016 Parkinson’s Disease Collaborators, 2018). Current data indicate that PD results in dysfunction and the loss of dopaminergic neurons in the SbN (Hirsch et al., 1988; Damier et al., 1999), which is likely related to the formation of α-synuclein protein aggregates known as Lewy bodies (Schulz-Schaeffer, 2010; Dagra et al., 2021). Elevated levels of α-synuclein are found in the CNS of PD patients (Tokuda et al., 2010; Irwin et al., 2012; Kouli et al., 2020) and can activate immune cells, such as microglia, macrophages, and T-cells, and astrocytes, increasing neuroinflammation, which is central to the progression of PD (Croisier et al., 2005; Thomas et al., 2007; Reynolds et al., 2008; Lee et al., 2010; Lindestam Arlehamn et al., 2020). Interestingly, recent data show that TH is upregulated in monocytes in the blood of PD patients and that TH expression in these cells is driven by TNF-α (Gopinath et al., 2021), suggesting that dopamine activity in peripheral myeloid cells may influence or be useful as a biomarker for PD progression.
In the CNS, microgliosis, astrogliosis and leukocyte infiltration are common histopathological findings in PD (Banati et al., 1998; Knott et al., 1999; Wilson et al., 2019; Kouli et al., 2020), and widespread microglial activation is present in PD patients (McGeer et al., 1988; Banati et al., 1998; Mirza et al., 2000; Imamura et al., 2003, 2005). In both humans and animal models, microglial activity correlates with neuronal death, starting prior to the death of dopaminergic neurons and exerting a neurotoxic effect on dopaminergic neurons that can result in neurodegeneration (Gao et al., 2002; Sugama et al., 2003, 2004;Ouchi et al., 2005; Zhang et al., 2005; Gerhard et al., 2006; Sawada et al., 2007; Kang et al., 2018). Neurotoxicity is mainly mediated by the production of ROS and the secretion of inflammatory cytokines such as TNF-α, IL-6 and IL-1β, which are elevated in the brain, cerebrospinal fluid (CSF), and serum in PD patients (Boka et al., 1994; Mogi et al., 1994; Müller et al., 1998; Imamura et al., 2005; Karpenko et al., 2018). The presence of ROS can exacerbate α-synuclein aggregation (Scudamore and Ciossek, 2018), and α-synuclein itself can activate microglia and astrocytes by binding to TLRs (Fellner et al., 2013; Kim et al., 2013; Daniele et al., 2015; Kouli et al., 2020; Sun et al., 2021) that are increased in PD brains (Shin et al., 2015; Dzamko et al., 2017; Maatouk et al., 2018; Ping et al., 2019). Increased TLR activation drives inflammation through NF-κB (Mogi et al., 2007; Reynolds et al., 2008; Pranski et al., 2013) and activates the inflammasome, which has been increasingly linked to PD pathology (Gordon et al., 2018; von Herrmann et al., 2018). Notably, inhibition of the NLRP3 inflammasome, which is primarily activated in macrophages and microglia (Gold and El Khoury, 2015; Chaurasia et al., 2018; Guermonprez and Helft, 2019; Voet et al., 2019), improves disease progression by ameliorating dopaminergic neurodegeneration, striatal dopamine depletion, the formation of α-synuclein aggregates, and the secretion of TNF-α, IL-6, and IL-1β (Mao et al., 2017; Gordon et al., 2018; Campolo et al., 2019; Lee et al., 2019; Ou et al., 2021).
Disruption of the dopaminergic system that triggers immune activation, particularly activation of the NLRP3 inflammasome, may create a feed-forward cycle and exacerbate the development of PD (Fig. 6). The loss of dopaminergic neurons means these cells are no longer available to regulate the concentration or spatial distribution of dopamine via DAT-mediated uptake, potentially exposing immune cells to aberrant dopamine concentrations that could drive inflammatory activity. These effects could be compounded by changes in the expression of dopamine receptors and other dopaminergic proteins on immune cells. While D1 expression is not altered in treated or untreated PD patients (Rinne et al., 1990; Shinotoh et al., 1993; Laihinen et al., 1994), D2 expression is initially upregulated and then decreases as the disease progresses (Rinne et al., 1990, 1995; Antonini et al., 1997; Kaasinen et al., 2000; Yang, Knight et al., 2021), correlating with PD severity (Antonini et al., 1995). Downregulation of D2 occurs mainly in the striatum of PD patients and not in the SbN, where the bulk of dopaminergic cell death occurs (Yang et al., 2021b), suggesting that dopaminergic neuronal death does not directly cause the decrease in D2. Despite the lack of direct effect on neuronal viability, the age-associated reduction in D2 (Antonini et al., 1993) may be one of the reasons that age is a risk factor for PD.