Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Send Orders for Reprints to reprints@benthamscience.

net

CNS & Neurological Disorders - Drug Targets, 2014, 13, 137-149 137

TRYCAT Pathways Link Peripheral Inflammation, Nicotine, Somatization


and Depression in the Etiology and Course of Parkinson’s Disease
George Anderson*,1 and Michael Maes2

1
CRC, Rm: 30, 57 Laurel Street, Glasgow, Scotland
2
Department of Psychiatry Faculty of Medicine, Pathumwan, Bangkok 10330, Thailand

Abstract: Increased depression, somatization, gut inflammation and wider peripheral inflammation are all associated with the
early stages of Parkinson’s disease (PD). Classically such concurrent conditions have been viewed as “comorbidities”, driven
by high levels of stress in a still poorly understood and treated disorder. Here we review the data on how oxidative and
nitrosative stress in association with immuno-inflammatory responses, drives alteration in tryptophan catabolites, including
kynurenine, kynurenic acid and quinolinic acid that drive not only the ‘comorbidities” of PD but also important processes in
the etiology and course of PD per se. The induction of indoleamine 2,3-dioxygenase, leading to the driving of tryptophan into
neuroregulatory tryptophan catabolite products and away from serotonin and melatonin production, has significant
implications for understanding the role of nicotine, melatonin, and caffeine in regulating PD susceptibility. Tryptophan
catabolite pathway activation will also regulate blood-brain barrier permeability, glia and mast cell reactivity as well as wider
innate and adaptive immune cell responses, all relevant to the course of PD. As such, the “comorbidities” of PD such as
depression, somatization and peripheral inflammatory disorders can all be conceptualized as being an intricate part of the
biological underpinnings of both the etiology and course of PD. As a consequence, the data reviewed here has treatment
implications; relevant to both the course of PD and in the management of L-DOPA induced dyskinesias.
Keywords: Parkinson’s, depression, somatization, tryptophan, indoleamine 2,3-dioxygenase, nicotinic, melatonin, inflammation,
cytokines.

INTRODUCTION Here we look at the role of the tryptophan catabolite


(TRYCAT) pathways in linking the wider aspects of PD,
With no current approved drugs capable of reducing
including somatization, depression and peripheral
disease progression, but only inhibiting the expression of
inflammation, with the induction and course of PD, as well
motor symptoms, the development of treatment that tackles
as in the protection afforded by nicotine and caffeine. This
the underlying neurodegenerative process in Parkinson’s
gives a wider systemic perspective on PD; emphasizing the
disease (PD) is a matter of extensive investigation. The delay role of the immune system in driving alterations centrally,
in effective treatment challenges researchers to better clarify
especially via neuronal regulation by immune and glia cell
the etiology and course of PD.
derived TRYCATs. First we review the data on the wider
Many susceptibility factors have been proposed, aspects of PD presentations and the indicants that these give
including environmental toxin exposure e.g. fungicides, to the underlying biological processes that drive this still
herbicides, pesticides, and metals [1, 2]. Mutations in a poorly understood and managed disorder.
number of genes are associated with both familial and
sporadic forms of PD, including alpha-synuclein, Parkin and PARKINSON’S: SOMATIZATION AND MDD
many others [3-6]. How such environmental and genetic risk
factors drive the etiological and biological underpinnings of Comorbid psychiatric conditions are common in PD [7],
PD, such as oxidative and nitrosative stress (O&NS), having a significant impact not only on quality of life and
mitochondrial dysfunction, peripheral and central resilience [8], but also disease outcome. Much of the focus
inflammation, as well as immune driven changes awaits of psychiatric comorbidity has been on later stages of PD.
clarification. The primary conceptualization of the disorder However, high levels of comorbid somatization (67%),
has linked such risk factors and biological changes directly depression (36%), anxiety (27%) and obsessive-compulsive
to atrophy of dopamine neurons in the substantia nigra (SN). symptoms ((53%) have also been found in the early motor
However, an early biomarker has remained elusive, meaning stages of PD, in non-demented patients [9]. These symptoms
that people only present when considerable damage has are highly intercorrelated and thought driven by adrenergic
already occurred, with symptoms predominantly involving and serotonergic dysfunction, mediated by early
SN atrophy. A wider perspective on the processes involved degeneration in brain stem nuclei, as shown by Braak [10].
in PD may help to not only improve treatment, but to provide Level of depression correlates with global cognitive
an early biomarker allowing preventative intervention. function, suggesting depression as an early marker of wider
brain dysfunction in PD 10]. The overlap of depression and
neurodegenerative processes is highlighted by the 2%
*Address correspondence to this author at the George Anderson, CRC, Rm 30,
57 Laurel St. Glasgow G11 7QT, Scotland; Tel: +447 505 623 759;
decrease in some cognitive measures occurring with every
E-mail: anderson.george@rocketmail.com depressive episode, linking to recent conceptualizations of

1996-3181/14 $58.00+.00 © 2014 Bentham Science Publishers


138 CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 Anderson and Maes

psychiatric conditions as neuroprogressive disorders [11]. associated with an increased risk for other conditions,
Other studies report similar rates of psychopathology in PD including Alzheimer’s, whilst the IL-18 induction of IFN-γ,
[12], with striatal dopamine transporter levels and is the main driver of wider immuno-inflammation. Increased
dopaminergic tone correlating with depressive and anxiety O&NS, evident in PD, contributes to Nod-like receptor
symptoms [13]. Single nucleotide polymorpisms (SNPs) in family containing pyrin domain (Nlrp)3 inflammasome
the circadian clock controlled gene thyrotroph embryonic activation [34]. As such inflammasome activation is likely to
factor (Tef) associate with depression in PD, suggesting interact with PD susceptibility genes in driving immuno-
overlaps with the circadian dysregulation evident in PD [14]. inflammation both centrally and periphery, in co-ordination
Overall psychiatric comorbidities may predate motor with alterations in oxidant status and wider immuno-
symptoms, correlate positively with motor symptoms, inflammation.
negatively with cognition and are linked to changes in
circadian regulation. CNS Inflammation
Increased cyclooxygenase-2 (COX2), IFN-γ, TNF-α and
PARKINSON’S: AUTONOMIC AND BP
IL-1β are found in postmortem PD brains [35]. Increased
A high prevalence of nocturnal nondipping blood vascular endothelial growth factor (VEGF) is evident in PD
pressure (BP), defined as less than a 10% decrease at night and PD models [36]. VEGF increases blood-brain barrier
versus day BP, is evident in PD [15]. Nondipping in this permeability (BBBp) [37], as found in PD [38] and PD
study occurred irrespective of orthostatic hypotension, models [39], including the 1-methyl-4-phenyl-1,2,3,6-
coexisting arterial hypertension or antihypertensive tetrahydropyridine (MPTP) model, where the increased
treatment. Lewy bodies are evident in peripheral and central BBBp is attenuated by caffeine [40]. Increased BBBp allows
structures involved in autonomic regulation [16], and the extravasation of monocytes/macrophages, neutrophils
alterations in the autonomic nervous system, in association and T-cells into the CNS. Increased CD4+ and CD8+ T-cells
with circadian dysregulation, occur in the early stages of PD are found in the SN in PD models, with CD4+ T-cells driving
[10, 17]. Increased TRYCAT pathway activation decreases dopaminergic toxicity [41]. Along with resident glia,
melatonin, often evident in PD [18], contributing to infiltrating leukocytes are a significant source of pro-
nondipping nocturnal BP. inflammatory cytokines, including TNF-α and IL-1β, which
have synergistic effects in the apoptosis of dopamine
PARKINSON’S: INFLAMMATION neurons [42]. The over-expression of α-synuclein in the
CNS increases microglia activation, priming pro-
Serum and CSF levels of proinflammatory cytokines and inflammatory responses, but only in the SN, suggesting
CD4+ and CD8+ Tcells are increased in PD [19-23]. Genetic region specific α-synuclein effects [43]. Peripheral
variations in these cytokine and immune factors associate inflammation enhances such α-synuclein priming of
with PD risk [24, 25]. Chronic users of some nonsterioidal microglia [44]. Also the loss of dopamine and
anti-inflammatory drugs (NSAIDs) show diminished PD norepinephrine (NE) in PD will impact in the CNS more
incidence [26], suggesting a significant role for inflammation widely [45], altering patterned CNS interactions.
in the etiology of PD. Loss of function mutations in Parkin
are associated with PD, with inflammation decreasing Parkin
Peripheral Inflammation
levels not only in neurons but also microglia and
macrophages [27]. Decreased Parkin enhances levels of The first observable manifestations of PD may occur
tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1b and peripherally, with increased levels of α-synuclein being
inducible nitric oxide synthase (iNOS) mRNA in activated present in the intestine [46], leading to the suggestion that
macrophages, suggesting that inflammation mimicks Parkin colonic mucosal biopsy may provide a biomarker of
mutations, and likely interacts with Parkin mutations to premotor PD. Likewise alterations in the intestine occur
regulate levels of inflammatory response. Combinations of prior to dopamine neuron loss, in α-synuclein over-
proinflammatory cytokines, TNF-α, IL-1beta (IL-1β) and expressing mice [47]. Infection with helicobacter pylori (Hp)
interferon-gamma (IFN-γ) also impact on levels and is associated with the course of idiopathic PD [48, 49] and
localization of glia α-synuclein and tau, suggesting wider not necessarily confined to those with current infection [50].
interactions of inflammation with other PD associated The eradication of Hp improves brady/hypokinesia but
proteins [28]. worsens rigidity [51]. Antimicrobial resistance can
complicate Hp symptoms and treatment [52], with failure to
Inflammasome activation leads to IL-1β and IL-18
eradicate aggravating the effects of infection in PD [53].
production and release. Remarkably little data is available on
Bladder dysregulation similarly predates motor dysfunction
the role of the inflammasome in PD, despite the well proven
in PD models [54]. Both bladder and bowel dysregulation
role of IL-1β in peripheral and central inflammation, as well
as the genetic link of IL-18 SNPs to sporadic late onset PD are associated with wider autonomic alterations. Peripheral
inflammation potentiates nigrostriatal dopamine neuron loss
[29]. PINK1, a susceptibility gene for autosomal recessive
in PD models [55-57]. As such peripheral inflammation is an
familial PD, regulates IL-1β activation [30], whilst α-
integral part of PD presentations, relevant to both etiology
synuclein enhances IL-1β induced interferon-γ inducible
and course, as well as the quest for an early premotor
protein-10 (CXCL10), a proinflammatory and neurotoxic
biomarker.
chemokine in astrocytes [31]. Combined SNPs in TNF-α and
IL-1β modulate the susceptibility to PD [32], although not Gastro-intestinal dysregulation commonly occurs early in
all studies show IL-1β SNP links to PD [33]. IL-18 SNPs are PD, with a history of constipation being a significant PD risk
TRYCAT Pathways Link Peripheral Inflammation CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 139

factor [58]. Animal models of gastro-intestinal disorders, in current or previous smokers [75]. Of the thousands of
including ulcerative colitis, potentiate central atrophy in PD chemical products in cigarette smoke, nicotine affords the
models [59]. Such work has led to the suggestion of a predominant protection in PD. Other cigarette smoke factors
particular role for gastro-intestinal inflammation in the also provide protection, including 2,3,6-trimethyl-1,4-
etiology of idiopathic PD [60]. Other gastro-intestinal naphthoquinone (TMN), a monoamine oxidase (MAO) A
changes occur in PD, including increased Lewy body and B inhibitor, which decreases dopamine metabolism and
pathology [61], serum lipopolysaccharide (LPS) binding MAO activation of endogenous neurotoxins [77]. Rasagiline,
protein [62], small intestinal bacterial overgrowth [63] and an MAO B inhibitor and PD treatment, similarly prolongs
gut permeability. The latter correlating with enhanced E. coli the action of dopamine [78].
intestinal mucosa staining, α-synuclein and oxidative stress.
Cigarette smoking also produces 2,3,7,8-tetrachloro-
Increased gut permeability also allows gram-negative dibenzo-p-dioxin (TCDD), which activates the aryl
bacteria to translocate into the blood, leading to a IgM-
hydrocarbon receptor (AHr). Interestingly the AHr, via AHr
mediated immune response [64].
response element III (AHRE-III) directly activates the
As to how these changes drive central alterations awaits tyrosine hydroxylase (TH) gene, increasing TH mRNA and
clarification. LPS leads to the atrophy of SN dopaminergic protein [79], suggesting that activation of the AHr, and its
neurons [65], partly via microglia activation, which is also modulation by cyclic adenosine monophosphate (cAMP),
evident in depression [66]. Ulcerative colitis increases C- phosphodiesterases (PDE) and circadian genes, will directly
reactive protein (CRP) [67], which increases BBBp [68], modulate levels of dopamine and NE produced [80]. The
contributing to central changes. Melatonin is protective in role of the AHr in PD, especially in TH derived dopamine
ulcerative colitis, preventing the CRP increase [67], and NE, will be important to determine. Two TRYCAT
suggesting a role for melatonin regulation of peripheral products, kyn and kynurenic acid (KYNA) are known to
influences on central PD processes. As well as peripherally regulate the AHr. However, different AHr ligands can have
derived cytokines and decreased microglia reactivity differential effects on AHr driven gene induction. As such
threshold [66, 69, 70], peripherally derived kynurenine (kyn) TCDD may have differential effects from kyn or KYNA.
has central consequences in PD, as detailed below. Also cAMP, known to induce KYNA in astrocytes [81], is
an important activator of the AHr, with the transactivation of
Inflammation and Early Development the B2-adrenoceptor induction of cAMP being necessary for
some other ligands to activate the AHr [82]. The cAMP
Given the role of central and peripheral inflammation in regulation of the AHr is co-ordinated with circadian gene
PD, early developmental factors regulating immuno- (Period 1 and 2) induction, suggesting interactions of TCDD,
inflammatory responses will modulate the susceptibility to, kyn and KYNA with cAMP/AHr/circadian genes, including
and course of, PD. Prenatal maternal infection increases the in the regulation of dopamine and NE. That said, smokeless
toxicity to 6-hydroxydopamine (6-OHDA) in adulthood [71]. tobacco also affords protection against PD [83].
Decreased dopamine neurons levels or activity possibly
mediate this [42], given dopaminergic hypoacitivity Nicotine is protective of dopamine neurons to different
increases adult LPS inflammation [72]. Such an early ongoing toxicities, including MPTP, 6-OHDA and
developmental influence on subsequent immuno- aminochrome [84], but not after damage has already
inflammation overlaps to the early developmental priming occurred [85]. Short term administration of a low dose of
for comorbid depression and somatization in the etiology of nicotine improves cognition and anti-oxidant status after 6-
schizophrenia [11]. As well as priming for schizophrenia and OHDA lesions, with cognitive improvements positively
its comorbidities, prenatal viral infection increases the adult correlating with increased anti-oxidant status [86]. Multiple
levels of the Alzheimer’s associated peptide amyloid B [73]. subtypes of nicotinic acetylcholine receptors (nAChr) are
This highlights the relevance of early immune responses to evident in the nigrostriatal pathway, including the α4β2,
adult neurodegenerative processes. Immunolocalization of α4α5β2, α6α4β2β3, and α6β2 β3 subunits, as well as
the influenza virus is evident in SN neurons and homomeric α7 [87]. All may be involved in the protective
macrophages of PD patients [74], suggesting a direct impact effects of nicotine in PD [88], although the α7nAChr is less
of infection in SN dopamine neurons in PD, including densely expressed in SN dopamine neurons, being more
possibly within an early developmental context. evident in SN astrocytes and GABAergic interneurons [89].
Nicotine also has wider cognitive benefits relevant to PD
[90], including increasing long-term potentiation (LTP). Of
PARKINSON’S: PROTECTIVE FACTORS the two phases of hippocampal LTP, early and late, the
A lack of history of cigarette smoking, having a family α7nAChr increases both, whilst α4β2 agonism increases the
member with PD and a history of constipation are the three early phase [91]. This is suggestive of wider cognitive
major risk factors for PD, each more than doubling the risk protection by nicotine in PD.
[58]. A number of protective factors are evident in PD, The SN receives ACh inputs from the pedunculopontine
including nicotine and caffeine [75, 76], giving important nucleus, an area subject to atrophy in PD. Nigrostriatal
clues as to the relevant processes driving PD. damage by MPTP decreases nAChr levels generally, except
the α7nAChr [92], with non-α7nAChr mediating direct
Nicotine protection in D neurons and PD models [93]. The α4β2 also
has anti-inflammatory effects [94]. Nicotine via the
Smoking decreases the incidence of PD, and SNPs in
α7nAChr activates a number of pathways, including the
nicotinic cholinergic receptor gene cluster, CHRNA5-
cAMP, extracellular kinase (ERK)1/2 and
CHRNA3-CHRNB4 modulate the age of development of PD
140 CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 Anderson and Maes

phosphatidylinositol 3-kinase (PI3K)/Akt paths [95]. These The α7nAChr stimulation of RegTcells immuno-
diverse pathway activations decrease apoptosis and increase suppression also has consequences for how peripheral
trophic factor production [96]. Overall this suggests a direct inflammation modulates PD. In the colon, cytokine induced
protective effect of nicotine via non-α7nAChr on D neurons, alterations in the α7nAChr in CD4+ Tcells mediate the
with non-dopamine neuron protection by the α7nAChr. dichotomous response to nicotine in Th1/Th17 versus Th2
driven colitis [113]. IL-4 increases, whereas IL-12 decreases,
Nicotine decreases L-DOPA induced dyskinesias in the
α7nAChr levels. As a consequence, in inflammatory bowel
6-OHDA model by 50% [97], involving the β2 nAChr [98]
disorders, nicotine ameliorates ulcerative colitis, increasing
and partly dependent on dopamine terminal function [99]. In
RegTcells, but worsens Chrohn’s disease, increasing Th17
about 50% of trials nicotine has direct motor symptom
cells. As to whether these cytokines have opposing effects on
benefits, possibly reflecting PD heterogeneity [100].
the α7nAChr in other cells relevant to PD, including
astrocytes and mast cells requires investigation. Overall, as
Non-Neuronal Impacts of Nicotine well as regulating leukocyte extravasation, nicotine will
modulate the peripheral driven immuno-inflammatory
Glia and Mast Cells
responses that interact with the course of PD.
As well as direct nicotinic effects on dopamine neurons,
nicotine effects in other cell types are also thought protective Autoimmunity
in PD, including immune cells, microglia and astrocytes Recent conceptualizations of many disorders, including
[101]. In the MPTP model α7nAChr protection is via the depression have emphasized a role for O&NS induced
inhibition of astrocyte activation [101], decreasing MPP+ or damage in the induction of autoimmune responses, including
LPS induced p38, ERK1/2 and TNF-α. ERK1/2 and p38 are to serotonin [114]. Autoimmunity is relevant in PD [115].
involved in the activation of astrocytes, increasing iNOS, Increased autoantibodies are evident in PD, being
COX2 and cytokine production [102]. An increase in striatal particularly associated with dyskinesia and depression [116].
and SN glial fibrillary acidic protein (GFAP) is evident in Neuromelanin is highly expressed in dopamine neurons,
PD and astrocyte activation is concurrent to neuronal death contributing to α-synuclein induced neurotoxicity [117].
in the MPTP model [103]. α7nAChr activation also High levels of anti-melanin antibodies are evident in PD,
decreases MPTP induced microglia activation [101]. being recognised by dendritic cells [118] and contributing to
Increased levels of activated microglia are found in the SN in autoimmunity [115]. Increased O&NS contributes to the L-
PD post-mortems [104]. DOPA induction of neuromelanin [119], suggesting a role
Cigarette smokers are also less likely to develop some for O&NS in interaction with L-DOPA in the induction of
inflammatory and allergic disorders [105], partly via nicotine autoimmunity. Both neuromelanin and α-synuclein are
effects at the α7, α9, and α10 nAChRs on mast cells [106]. increased in SN dopamine neurons over normal ageing
Nanomolar nicotine concentrations inhibit inflammatory [120], highlighting how ageing associates with PD
effluxes from rat mast cells, including TNF-α and IL-1β, autoimmune responses. α-synuclein increases neuromelanin
driven by decreased nuclear factor kappaBeta (NF-κB) [121], increasing microglia activation by neuromelanin
[106]. Nanomolar effects of nicotine in mast cells are far [122]. Loss of olfaction occurs early in PD, driven by
lower than that required to produce action potentials, autoimmunity [123].
suggesting that the combination of subunits is important in In attenuating central inflammation, nicotine decreases
modulating nicotine sensitivity. autoimmunity [124]. Melatonin decreases α-synuclein and
Mast cells and especially astrocytes are crucial regulators oxidants, suggesting protection against autoimmunity in PD.
of BBBp, including via TNF-α and IL-1β efflux, which Autoimmunity may also have protective effects, as
nicotine inhibits. This suggests that nicotine will evidenced by increased IgG against α-synuclein [125].
significantly impact on BBBp and associated leukocyte
extravasation, important to the CNS degeneration occurring CAFFEINE/ADENOSINE
in PD.
Caffeine consumption decreases PD risk [76], and is
Adaptive and Innate Immunity protective against MPTP, where, like nicotine it decreasing
BBBp [40, 126]. Caffeine decreases melatonin metabolism
As well as modulating immune cells access to the CNS, by cytochrome P450 (CYP)1A2 [127]. As such, caffeine
nicotine directly regulates immune responses. Adaptive and efficacy in PD [128] may be partly mediated by melatonin,
innate immunity activation is evident in PD, with immuno- in turn increasing the α7nAChr [129], potentiating nicotine
suppression by regulatory T cells (RegT) affording effects. Nicotine and caffeine protection in carriers of PD
protection to MPTP [107], including via the inhibition of T- SNP gene susceptibilities [130] may therefore be partly
helper (Th)17 induced damage [108]. α7nAChr stimulation regulated by melatonin.
in murine RegT cells potentiates their immuno-suppressive
capacity [109], highlighting nicotine protection via immune However, caffeine efficacy is also via adenosine A1r and
regulation [88]. Potentiating RegT cell immuno-suppression A2Ar inhibition, supported by the protection afforded by
decreases the damage arising from elevated CD4+ and CD8+ istradefylline, an A2Ar antagonist, when used as an
T cells in PD. The α7nAChr also directly dampens adjunctive to L-DOPA [131]. As well as the role of
proinflammatory production [110], including in T-cells [111] adenosine in the striatum in the course of PD, cortex ecto-5-
and macrophages [112]. nucleotidase (Ecto-5N) increases over ageing, elevating
adenosine, which normally contribute to night-time
TRYCAT Pathways Link Peripheral Inflammation CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 141

tiredness. cAMP increases astrocyte Ecto-5N and adenosine NMDAr inhibition. These effects highlight the role of the
[132]. As indicated above, the cAMP pathway also regulates NMDAr in driving toxicity in these models. However,
the AHr, circadian genes and TH, as well as tryptophan KYNA inhibition of the NMDAr is unlikely to be achieved
catabolism. As such cAMP may have a role in co-ordinating by physiological variations in kyn. Given elevated peripheral
PD processes. inflammation and somatization in PD, it is of note that KAT-
1 is decreased in PD red blood cells, coupled to decreased
PARKINSON’S AND TRYCATs plasma KYNA [141]. Peripheral KYNA is antinociceptive
via G-protein coupled receptor (GPR)-35 activation [142].
TRYCATs As to whether decreased peripheral KYNA reflects an
Tryptophan is the precursor necessary for serotonin and increase in the kyn/KYNA ratio, with links to peripheral
melatonin synthesis. Over 95% of tryptophan is taken down inflammation and somatization, as suggested by increased
the kynurenine pathway, leading to various neuromodulatory neopterin and kyn/tryptophan ratio in PD [143], requires
TRYCATs, including kyn, KYNA and quinolinic acid investigation. Pharmacologically increasing KYNA lessens
(QUIN). TRYCAT production is driven by indoleamine 2,3- L-DOPA induced dyskinesias in monkeys [144], suggesting
dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). central benefits from KYNA analogs that cross the BBB,
IDO is predominantly induced by IFN-γ, although other unlike peripheral KYNA. However, such benefits are via
proinflammatory cytokines, including IL-1β, TNF-α and IL- NMDAr inhibition, and may require adjunctive,
18 also contribute [133]. TDO is increased by cortisol [134] compensatory nicotine or an α7nAChr agonist.
and perhaps cAMP, as indicated by cAMP induction of Peripheral kyn can also be converted to other TRYCATs,
astrocyte KYNA [81]. IDO activation produces all including QUIN, 3-hydroxykyurenine (3-HK), 3-
TRYCATs, whereas astrocyte TDO is limited to kyn and hydroxyanthranillic acid (3-HA) and picolinic acid (PA). At
KYNA production. Kynurenine aminotransferase (KAT) the NMDAr, QUIN is excitotoxic and PA excitatory. Both 3-
catalyzes KYNA from kyn. In the CNS, TDO is HK and 3-HA decrease mitochondrial complexes I and II,
predominantly expressed in astrocytes and some neurons, with 3-HK additionally inhibiting complex IV [145]. This
whereas IDO is expressed in microglia and infiltrating suggests that peripheral inflammation and somatization
leukocytes. induced kyn contributes to wider neuronal susceptibility in
PD, via mitochondrial inhibition by 3-HA and 3-HK.
TRYCATs and Parkinson’s Melatonin increases mitochondrial complexes I, II and IV,
and would be expected to inhibit such 3-HK and 3-HA
Elevated peripheral and central inflammation in PD mitochondrial toxicity [146].
increases IDO, driving down serotonin and melatonin and
contributing to depression and circadian dysregulation. We
recently showed that increased peripheral kyn/KYNA ratio is TRYCATs: Neurogenesis and LTP
specifically associated with somatization, across a number of Astrocyte and mast cell IL-1β induction modulates
disorders, including depression and myalgic BBBp, as well as inhibiting neurogenesis. IL-1β effects are
encephalomyelitis [135, 136]. Somatization and depression via IDO and downstream TRYCAT products, including kyn
levels are high in PD, including in the earliest stages [9], and and kynurenine 3-monooxygenase (KMO) [147]. KMO
can predate PD motor symptoms, sometimes by decades. inactivation prevents IL-1β inhibition of neurogenesis,
This indicates altered TRYCAT levels and patterning. Over suggesting a powerful role for IDO and the TRYCAT
60% of CNS kyn is peripherally derived, being readily pathway in driving inflammation induced decreased
transported over the BBB. In the CNS, kyn is catalyzed by neurogenesis. TDO and IDO activation decreases mast cell
astrocyte and neuronal TDO and KAT into KYNA. KYNA, serotonin production, also important to dentate gyrus
at physiological levels, is an antagonist of the α7nAChr and neurogenesis, as well as learning and mood [148]. Such data
will therefore inhibit the protection afforded by nicotine at emphasizes the importance of the TRYCATs in mediating
the α7nAChr, including in: immuno-inflammatory responses depression/somatization associations with degenerative and
by astrocytes and mast cells, cytokine induced BBBp [137] regenerative processes in PD. TDO activation decreases
and leukocyte extravasation and activation, as well as direct neurogenesis, whilst increasing anxiety [149].
immune cell regulation. Increased KYNA will inhibit these
Nicotine, via the α7nAChr, increases early and late LTP
manifold nicotine protections in PD, contributing to wider
presentations. As such peripheral inflammation, and [91], suggesting that peripheral derived kyn and central
KYNA conversion contributes to PD cognitive deficits, as
associated somatization and depression may not simply be
evidenced in schizophrenia models [150]. As such nicotine’s
incidental comorbidities of PD, but may be intimately
wider cognitive and neuroregenerative benefits [90] will be
involved in its etiology and course.
inhibited by peripheral inflammation induced kyn.
Following 6-OHDA there is a relative loss of KAT-I in Variability in nicotine benefit in PD studies may therefore be
surviving dopamine neurons, although increased KAT-I in a confound derived from somatization and peripheral
astrocytes and microglia [138]. However, the resultant inflammation induced TRYCATs.
increase in glia KYNA is insufficient to inhibit the N-methyl
d-aspartate receptor (NMDAr), but rather will inhibit the TRYCATs and Wider Immuno-Inflammation
α7nAChr, potentially increasing BBBp and neuronal
toxicity. Pre-treatment with high dose L-kynurenine Wider immune changes are evident in PD, linking to
attenuates 6-OHDA and MPTP induced toxicity [139,140], increased inflammation. Increased natural killer (NK) cells
being mediated by conversion to KYNA, with resultant [151], IL-1β, and decreased IL-1RA occur in PD, correlating
142 CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 Anderson and Maes

with levels of fatigue and depression [152]. NK cells in PD PDE2 inhibitors may change the AHr activation and gene
express decreased inhibitory NKG2A receptor levels, inductions by other AHr ligands. Given the role of the AHr
indicative of priming for activation [153]. Regulated on in regulation of TH, dopamine and NE, this will be important
activation normal T-cell expressed and secreted (RANTES) to clarify. PDE inhibitors are an area of active investigation
is also increased in the PD serum, suggesting increased in the treatment of neurodegenerative disorders, including
monocytes, macrophages and Tcells chemoattraction [154]. PD [162].
Increased gamma delta (γδ) Tcells occur in PD blood and
CSF [155]. In vitro, γδTcells and NK cells can be neurotoxic TRYCATs and Stress
via astrocytes [156]. Such inflammatory processes increase
cytokine production, in turn inducing IDO, kyn and KYNA, PD is a stressful condition, with unexpected symptoms
with KYNA inhibiting the α7nAChr benefits in many cell and comorbidities/wider presentations often emerging.
types and processes. Within the animal literature chronic unpredictable mild
stress (CUMS) is known to increase depression, with effects
The α7nAChr is a treatment target for many being mediated by increased peripheral kyn, driving raised
inflammatory conditions, including ulcerative colitis [157], levels of excitotoxic QUIN in the amygdala and striatum
suggesting KYNA regulation of nicotine modulation of coupled to a trend increase in cortex KYNA [163,164].
peripheral inflammatory effects in PD. As such the absolute, Excitatory activity in the amygdala, coupled to KYNA
as well as local, levels of KYNA would be relevant. The inhibition of cortex acetylcholine, dopamine and glutamate
inability of peripheral inflammation to potentiate SN atrophy suggests increased affective processing at the expense of
in PD models, when monocytes are depleted [59], highlights higher order cognition [165]. Stressful life events are major
the importance of increased BBBp and infiltrating susceptibility factors for the emergence of depression,
leukocytes. An increase in infiltrating Tcells in the SN is including in PD [166]. As such stress, as with peripheral
evident in PD patients [158]. inflammation, mediates its effects in the CNS, at least in
part, via conversion by glia into neuronal regulating
TRYCATs and AHr TRYCATs.
Given that different AHr ligands, including kyn, KYNA
and cAMP, can have differential effects on AHr induced Alexithymia
transcriptions, including reactive oxygen species (ROS) but Other PD comorbidities may be similarly related. Up to
also NF-E2-related factor 2 (Nrf-2) induced endogenous 20% of people with PD show symptoms of alexithymia, with
antioxidants, it will be important to determine how different altered emotional recognition and expression leading to
endogenous and exogenous ligands modulate TH levels, and deficits in empathy and the emotional aspects of social
how cAMP and circadian genes interact with this. A role for interactions [167]. In PD patients, alexithymia is associated
the AHr suggests that cigarette smoke, which contains high with cognitive deficits [168], linked to decreased cortex
levels of TCDD, may have impacts on PD processes, as well α7nAChr level and activity [165]. Alexithymia also
as the known effects of nicotine. The interactions of the AHr, associates with increased impulsivity [169], although not
cAMP and circadian genes can be relevant to the course of correlating with measures of depression in PD [170].
peripheral inflammation, including in gastro-intestinal However, alexithymia is classically associated with
disorders [80]. Given the role of such peripheral somatization [171], and it requires investigation as to
inflammatory disorders in PD, and their possible etiological whether alexithymia in PD associates with increased
importance, the regulation of the AHr, peripherally and peripheral kyn/KYNA ratio.
centrally, requires investigation. Developmental exposure to
AHr ligands can impact on TH levels and dopamine
production [159], and may mediate early TRYCATs and Melatonin
developmental/epigenetic driven changes relevant to PD Melatonin affords protection in the MPTP and 6-OHDA
etiology. As such TRYCAT variations may interact with the models [172, 173]. The driving down of melatonin levels,
AHr, circadian genes and cAMP in the regulation of central including pineal but also local glia/leukocyte melatonin in
dopamine and NE, as well as in peripheral and early the SN [18], by enhanced IDO and TDO activation in PD,
developmental PD factors. means not only the loss of melatonin’s potentiation of
Interestingly the activation of the AHr can increase IDO, α7nAChr levels and effects. Melatonin is an anti-oxidant,
leading to TRYCAT induction [160]. This suggests that kyn anti-inflammatory, anti-nociceptive and increases
[161] or KYNA activation of the AHr could induce a mitochondrial oxidative phosphorylation [146]. Melatonin
positive feedback loop via the induction of IDO. Given the also increases activity induced dentate gyrus neurogenesis
important role of TRYCATs in PD, neuronal regulation, and new cell survival [174]. Melatonin receptors are
excitotoxicity and BBBp, it will be important to determine if decreased in the SN in PD, suggesting a loss of local
this occurs and what are the feedback paths that inhibit this melatonin effects, given that melatonin increases its own
kyn/KYNA-AHr loop. It may be that the prolonged receptors. Decreased melatonin will contribute to the sleep
activation of cAMP paths leading to PDEs is relevant to the disturbance and circadian dysregulation common in PD [18].
inhibition of such a putative positive feedback loop. PDE2 Melatonin receptors are negatively coupled to adenylyl
can bind to one of the cytosolic factors that associate with cyclase, leading to cAMP inhibition. As such melatonin may
the AHr, the hepatitis B virus X-activating protein 2 (XAP2), inhibit any cAMP induction of TDO/KYNA, as well as
preventing AHr nuclear translocation and transcription regulating AHr activation and circadian gene inductions. As
inductions. In modulating cAMP regulation of the AHr, such, variations in melatonin can be intimately associated
TRYCAT Pathways Link Peripheral Inflammation CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 143

with TRYCAT regulation and will impact on many aspects activation increases subventricular zone neurogenesis via the
of PD and its associated presentations. induction of fibroblast growth factor-2 [186], whereas
chronic nicotine decreases new neuron survival in the
The regulation of astrocyte melatonin production,
dentate gyrus [187]. As to whether differential regulation of
including by available serotonin and MAOA/B, will be
important to determine in PD. neurogenesis or the lack of optimized temporal activation of
the α7nAChr is relevant to some of the mixed results
regarding melatonin efficacy in PD models requires
TREATMENT IMPLICATIONS investigation.
Melatonin
Caffeine
Melatonin’s inhibition of oxidative stress, inflammation,
α-synuclein and nociception, coupled to its increase in Caffeine decreases the risk of PD [76] and also decreases
mitochondrial oxidative phosphorylation, neurogenesis and melatonin metabolism by CYP1A2 [127]. As such, some of
new cell survival, as well as its induction of the α7nAChr the efficacy of caffeine in the treatment of PD [128] may be
[129], all suggest benefits in PD. Melatonin would decrease mediated by its influence on melatonin and nictotine at the
the high levels (88%) of nocturnal non-dipping hypertension α7nAChr [129]. Caffeine, like nicotine at the α7nAChr,
in PD [15]. Melatonin also decreases ageing associated decreases BBBp [126]. The effects of caffeine in PD may
decrements in neurogenesis [175]. Melatonin decreases then be linked to alterations in the TRYCATs, as well as via
nigrostriatal neurodegeneration, behavioural motor deficits adenosine receptors.
and mitochondrial dysfunction, as well as maintaining TH
and dopamine transporter levels [172]. In comparison to L- Vitamin D
DOPA, melatonin treatment can improve performance on
motor tasks, with no dyskinesias evident [176]. Importantly Hypovitaminosis D associates with neurodegeneration,
melatonin decreases α-synuclein induced neurotoxicity, including PD [188], leading to vitamin D being proposed as
whilst inhibiting α-synuclein protofibril formation, an adjunctive treatment for PD [189]. Vitamin D is also a
oligomerization, and secondary structure transitions [177], significant regulator of neurogenesis, both in the dentate
including SN α-synuclein induced by amphetamine [178]. gyrus and subventricular zone, generally decreasing
proliferation but increasing the neuron/astrocyte ratio [190,
Sleep dysregulation is common in PD and in animal 191]. Vitamin D is also a significant immune regulator, and
models contributes to the disease process [179]. Melatonin, a therefore a decrease in vitamin D is likely to contribute to
key circadian entrainer, improves sleep regulation [18]. the role of peripheral and central immuno-inflammation in
Melatonin has anti-depressant effects, including potentiation PD. As to how vitamin D interacts with TRYCATs requires
buspirone efficacy [180]. N-acetylserotonin, the immediate investigation.
precursor of melatonin, has similar beneficial effects,
including via the activation of tyrosine kinase receptor-beta,
the brain-derived neurotrophic factor receptor [181]. Anti-Inflammatories
Melatonin’s protection in ulcerative colitis, preventing the Chronic use of NSAIDs associates with decreased PD
CRP rise and subsequent BBBp [67], highlights how the incidence [26], suggesting a significant role for inflammation
prevention of inflammatory processes in peripheral disorders in the etiology of PD. However, later data shows that the
will modulate the etiology and course of PD. protection afforded by NSAIDs is mediated primarily by
ibuprofen [192], supported by a meta-analysis of all NSAID
Nicotine and Melatonin Combined studies [193]. Meloxicam, an oxicam NSAID, affords
protection in the MPTP model, increasing TH levels and
Nicotine affords some benefits in PD, including on motor prosurvival pathway activation, independent of COX
symptoms in some studies. The lack of clarity as to how it is inhibition [194]. The NSAIDs, tolmetin and sulindac,
best administered (patches, gum, intermittent versus chronic decrease levels of hepatic TDO, suggesting an impact on
etc) seems somewhat amiss, and requires clarification. Also peripheral TRYCATs [195]. Although increasing
it will be important to determine the most beneficial dose of hippocampal serotonin, these NSAIDs also decrease striatal
melatonin for the induction of the α7nAChr in PD [129], dopamine levels; with potential adverse effects in PD.
especially in astrocytes and mast cells, and therefore on NSAID use may also inhibit antidepressant response [196].
BBBp. Melatonin modulates the LPS regulation of the No evidence suggests a benefit from aspirin.
α7nAChr mRNA in astrocytes [182]. As to whether a
combination of melatonin and nicotine/α7nAChr agonist Some of these studies are confounded by increased
would prevent or delay the development of PD, is hard to NSAID use in the years immediate to a PD diagnosis [197],
test, given the lack of an early biomarker. However, it is where increased peripheral inflammation may be intimately
likely that such a combination would, at the very least, have associated with PD. Such data therefore confounds
immediate benefit in offsetting L-DOPA induced dyskinesias preventative effects with impacts on the early course of
[183]. degenerative processes, and possibly with indirect, non-
identifiable early course PD symptoms. As a consequence an
However, not all studies using PD models have found increase in PD incidence has been associated with NSAID
benefits of melatonin [184,185], raising questions as to use in some studies [197]. Gastric dysregulation is
whether variations in experimental protocol, including heightened in depression, with NSAIDs exacerbating gastric
detrimental effects of very high levels of melatonin, mediate disorders, including acid secretion and ulcer formation [198].
such lack of protection. Intermittent nicotinic receptor Given the putative role of gastro-intestinal disorders in PD,
144 CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 Anderson and Maes

this could be another route for NSAID interactions with PD IFN-γ = Interferon-gamma
etiology and course.
IL = Interleukin
COX-2 inhibitors may also increase neuroinflammation,
KAT = Kynurenine aminotransferase
Th1 responses and lipid peroxidation, as well as decreasing
antioxidants and damaging mitochondria [199]. COX-2 Kyn = Kynurenine
inhibitors may therefore negatively modulate the course of KYNA = Kynurenic acid
PD and depression. Importantly for PD, COX-2 inhibitors
may potentiate bacterial translocation via enhanced Th1 LPS = Lipopolysaccharide
effects. Overall, the conceptualization of PD in the context LTP = Long term potentiation
of wider immuno-inflammatory processes, does not lead
simply to treatment with classical anti-inflammatories. MAO = Monoamine oxidase
MPTP = 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
PDE Inhibitors NE = Norepinephrine
PDE inhibitors are being extensively investigated for NK = Natural killer cells
their efficacy in the treatment of a range of
neurodegenerative disorders, including PD [162], affording NMDAr = N-methyl d-aspartate receptor
protection in the MPTP model [200] and on BBBp [201]. NSAID = Non-steroidal anti-inflammatory drug
PDE4 inhibitors will also increase night-time melatonin
levels [202]. PDE2A inhibitors are under extensive research O&NS = Oxidative and nitrosative stress
in the treatment of inflammatory processes [203], which as PD = Parkinson’s disease
indicated above would have significance in PD and therefore
PDE = Phosphodiesterase inhibitor
in peripherally derived TRYCATs, as well as on AHr
regulation. QUIN = Quinolinic acid
How the various treatments highlighted would interact RegT = Regulatory T cells
with TRYCATs and biological processes in PD will be
SN = Substantia nigra
important to determine. A number of future research
directions are suggested by including TRYCATs in the SNP = Single nucleotide polymorphism
etiology and course of PD. Perhaps the most fruitful to TCDD = 2,3,7,8-tetrachlorodibenzo-p-dioxin
clarify will be the role of local melatonin production by SN
glia. TDO = Tryptophan 2,3-dioxygenase
Th = T-helper
CONCLUSIONS
TH = Tyrosine hydroxylase
The role of altered TRYCATs in PD in driving the TNF-α = Tumor necrosis factor-alpha
effects of inflammation, stress/CUMS and somatization has
implications for the conceptualization and treatment of PD. TRYCAT = Tryptophan catabolites
Peripheral inflammation and somatization may not be simple
comorbidities, but rather an intimate part of the processes CONFLICT OF INTEREST
driving the etiology and course of PD. A combination of
melatonin, nicotine/α7nAChr agonist, vitamin D and Neither author has any conflicts of interest to declare.
caffeine are likely to have interactive benefits at different
sites and cells, both peripherally and centrally. ACKNOWLEDGEMENTS
Declared none.
LIST OF ABBREVIATIONS
3-HA = 3-Hydroxyanthranillic acid REFERENCES
3-HK = 3-Hydroxykyurenine [1] Wang A, Costello S, Cockburn M, et al. Parkinson’s disease risk
from ambient exposure to pesticides. Eur J Epidemiol 2011; 26:
6-OHDA = 6-Hydroxydopamine 547-55.
[2] Wirdefeldt K, Adami HO, Cole P, Trichopoulos D, Mandel J.
α7nAChr = Alpha 7 nicotine acetylcholine receptor Epidemiology and etiology of Parkinson’s disease: a review of the
evidence. Eur J Epidemiol 2011; 26(Suppl 1): S1-58.
AHr = Aryl hydrocarbon receptor [3] Feng LR, Maguire-Zeiss KA. Gene therapy in Parkinson’s disease:
BBBp = Blood brain barrier permeability rationale and current status. CNS Drugs 2010; 24: 177-92.
[4] Bonifati V, Rizzu P, van Baren MJ, et al. Mutations in the DJ-1
cAMP = Cyclic adenosine monophosphate gene associated with autosomal recessive early-onset parkinsonism.
Science 2003; 299: 256-9.
COX = Cyclo-oxygenase [5] Strauss KM, Martins LM, Plun-Favreau H, et al. Loss of function
mutations in the gene encoding Omi/HtrA2 in Parkinson’s disease.
CRP = C-reactive protein Hum Mol Genet 2005; 14: 2099-111.
ERK = Extracellular regulated kinase [6] Paisan-Ruiz C, Jain S, Evans EW, et al. Cloning of the gene
containing mutations that cause PARK8-linked Parkinson’s
IDO = Indoleamine 2,3-dioxygenase disease. Neuron 2004; 44: 595-600.
TRYCAT Pathways Link Peripheral Inflammation CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 145

[7] Aarsland D, Brønnick K, Alves G, et al. The spectrum of regulation of TRAF6 and TAK1 Cell Mol Life Sci 2012; 69(19):
neuropsychiatric symptoms in patients with early untreated 3301-15.
Parkinson's disease. J Neurol Neurosurg Psychiatry 2009; 80(8): [31] Tousi NS, Buck DJ, Curtis JT, Davis RL. α-Synuclein potentiates
928-30. interleukin-1β-induced CXCL10 expression in human A172
[8] Robottom BJ, Gruber-Baldini AL, Anderson KE, et al. What astrocytoma cells. Neurosci Lett 2012; 507(2): 133-6.
determines resilience in patients with Parkinson's disease? [32] Pascale E, Passarelli E, Purcaro C, et al. Lack of association
Parkinsonism Relat Disord 2012; 18(2): 174-7. between IL-1β, TNF-α, and IL-10 gene polymorphisms and
[9] Bugalho P, da Silva JA, Cargaleiro I, Serra M, Neto B. Psychiatric sporadic Parkinson's disease in an Italian cohort. Acta Neurol
symptoms screening in the early stages of Parkinson's disease. J Scand 2011; 124(3): 176-81.
Neurol 2012; 259(1): 124-31. [33] Liu GJ, Feng RN, Luo C, Bi S. Lack of association between
[10] Braak H, Del Tredici K, Rub U, et al. Staging of brain pathology interleukin-1 alpha, beta polymorphisms and Parkinson's disease.
related to sporadic Parkinson’s disease. Neurobiol Aging 2003; Neurosci Lett 2010; 480(2): 158-61.
24(2): 197-211. [34] Rubartelli A. Redox control of NLRP3 inflammasome activation in
[11] Anderson G, Maes M, Berk M. Schizophrenia is primed for an health and disease. J Leukoc Biol 2012; 92(5): 951-8.
increased expression of depression through activation of immuno- [35] Mogi M, Harada M, Narabayashi H, et al. Interleukin (IL)-1β, IL-
inflammatory, oxidative and nitrosative stress, and tryptophan 2: IL-4: IL-6 and transforming growth factor-α levels are elevated
catabolite pathways. Prog Neuropsychopharmacol Biol Psychiatry in ventricular cerebrospinal fluid in juvenile parkinsonism and
2013; 42: 101-14. Parkinson’s disease. Neurosci Lett 1996; 211(1): 13-6.
[12] Siri C, Cilia R, De Gaspari D, et al. Psychiatric symptoms in [36] Yasuda T, Fukuda-Tani M, Nihira T, et al. Correlation between
Parkinson's disease assessed with the SCL-90R self-reported levels of pigment epithelium-derived factor and vascular
questionnaire. Neurol Sci 2010; 31(1): 35-40. endothelial growth factor in the striatum of patients with
[13] Ceravolo R, Frosini D, Poletti M, et al. Mild affective symptoms in Parkinson’s disease. Exp Neurol 2007; 206(2): 308-17.
de novo Parkinson's disease patients: relationship with [37] Argaw AT, Asp L, Zhang J, et al. Astrocyte-derived VEGF-A
dopaminergic dysfunction. Eur J Neurol 2013, 20(3), 480-5. drives blood-brain barrier disruption in CNS inflammatory disease.
[14] Hua P, Liu W, Kuo SH, et al. Association of Tef polymorphism J Clin Invest 2012; 122(7): 2454-68.
with depression in Parkinson disease. Mov Disord 2012; 27(13): [38] Rite I, Machado A, Cano J, Venero JL. Blood-brain barrier
1694-7. disruption induces in vivo degeneration of nigral dopaminergic
[15] Sommer S, Aral-Becher B, Jost W. Nondipping in Parkinson's neurons. J Neurochem 2007; 101(6): 1567-82.
disease. Parkinsons Dis 2011; 2011; 897586. [39] Zhao C, Ling Z, Newman MB, Bhatia A, Carvey PM. TNF-α
[16] Ohama E, Ikuta F. Parkinson’s disease: distribution of lewy bodies knockout and minocycline treatment attenuates blood-brain barrier
and monoamine neuron system. Acta Neuropathol 1976; 34(4): leakage in MPTP-treated mice. Neurobiol Dis 2007; 26(1): 36-46.
311-9. [40] Chen X, Lan X, Roche I, Liu R, Geiger JD. Caffeine protects
[17] Niwa F, Kuriyama N, Nakagawa M, Imanishi J. Circadian rhythm against MPTP-induced blood-brain barrier dysfunction in mouse
of rest activity and autonomic nervous system activity at different striatum. J Neurochem 2008; 107(4): 1147-57.
stages in Parkinson's disease. Auton Neurosci 2011; 165(2): 195- [41] Brochard V, Combadiere B, Prigent A, et al. Infiltration of CD4+
200. lymphocytes into the brain contributes to neurodegeneration in a
[18] Srinivasan V, Cardinali DP, Srinivasan US, et al. Therapeutic mouse model of Parkinson disease. J Clin Invest 2009; 119(1):
potential of melatonin and its analogs in Parkinson’s disease: focus 182-92.
on sleep and neuroprotection. Ther Adv Neurol Disord 2011; 4(5): [42] Carvey PM, Chen EY, Lipton JW, Tong CW, Chang QA, Ling ZD.
297-317. Intra-parenchymal injection of tumor necrosis factor-α and
[19] Dobbs RJ, Charlett A, Purkiss AG, et al. Association of circulating interleukin 1-β produces dopamine neuron loss in the rat. J Neural
TNF- α and IL-6 with ageing and parkinsonism. Acta Neurol Scand Transm 2005; 112(5): 601-12.
1999; 100(1): 34-41. [43] Watson MB, Richter F, Lee SK, et al. Regionally-specific
[20] Blum-Degena D, Muller T, Kuhn W, et al. Interleukin-1β and microglial activation in young mice over-expressing human
interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s wildtype alpha-synuclein. Exp Neurol 2012; 237(2): 318-34.
and de novo Parkinson’s disease patients. Neurosci Lett 1995; [44] Couch Y, Alvarez-Erviti L, Sibson NR, Wood MJ, Anthony DC.
202(1-2): 17-20. The acute inflammatory response to intranigral α-synuclein differs
[21] Reale M, Iarlori C, Thomas A, et al. Peripheral cytokines profile in significantly from intranigral lipopolysaccharide and is exacerbated
Parkinson’s disease. Br Beh Immun 2009; 23(1): 55-63. by peripheral inflammation. J Neuroinflamm: 2011; 8: 166.
[22] Hisanaga K, Asagi M, Itoyama Y, Iwasaki Y. Increase in peripheral [45] Domenger D, Dea D, Theroux L, et al. The MPTP neurotoxic
CD4 bright+ CD8 dull+T cells in Parkinson disease. Arch Neurol lesion model of Parkinson's disease activates the apolipoprotein E
2001; 58(10): 1580-3. cascade in the mouse brain. Exp Neurol 2012; 233(1): 513-22.
[23] Bas J, Calopa M, Mestre M, et al. Lymphocyte populations in [46] Shannon KM, Keshavarzian A, Dodiya HB, Jakate S, Kordower
Parkinson’s disease and in rat models of parkinsonism. J JH. Is alpha-synuclein in the colon a biomarker for premotor
Neuroimmunol 2001; 113(1): 146-52. Parkinson's disease? Evidence from 3 cases. Mov Disord 2012;
[24] Hamza TH, Zabetian CP, Tenesa A, et al. Common genetic 27(6): 716-9.
variation in the HLA region is associated with late- onset sporadic [47] Wang L, Magen I, Yuan PQ, et al. Mice overexpressing wild-type
Parkinson’s disease. Nat Genet 2010; 42(9): 781-5. human alpha-synuclein display alterations in colonic myenteric
[25] Wahner AD, Sinsheimer JS, Bronstein JM, Ritz B. Inflammatory ganglia and defecation. Neurogastroenterol Motil 2012; 24(9):
cytokine gene polymorphisms and increased risk of Parkinson e425-36.
disease. Arch Neurol 2007; 64(6): 836-40. [48] Dobbs RJ, Dobbs SM, Weller C, et al. Role of chronic infection
[26] Chen H, Jacobs E, Schwarzschild MA, et al. A Nonsteroidal and inflammation in the gastrointestinal tract in the etiology and
antiinflammatory drug use and the risk for Parkinson’s disease. pathogenesis of idiopathic parkinsonism. Part 1: eradication of
Annal Neurol 2005; 58(6): 963-7. Helicobacter in the cachexia of idiopathic parkinsonism.
[27] Tran TA, Nguyen AD, Chang J, et al. Lipopolysaccharide and Helicobacter 2005; 10(4): 267-725.
tumor necrosis factor regulate Parkin expression via nuclear factor- [49] Dobbs RJ, Dobbs SM, Weller C, et al. Helicobacter hypothesis for
kappa B. PLoS One 2011; 6(8): e23660. idiopathic parkinsonism: before and beyond Helicobacter 2008;
[28] Bick RJ, Poindexter BJ, Kott MM, et al. Cytokines disrupt 13(5): 309-22.
intracellular patterns of Parkinson's disease-associated proteins [50] Weller C, Charlett A, Oxlade NL, et al. Role of chronic infection
alpha-synuclein, tau and ubiquitin in cultured glial cells. Brain Res and inflammation in the gastrointestinal tract in the etiology and
2008; 1217: 203-12. pathogenesis of idiopathic parkinsonism. Part 3: predicted
[29] Xu X, Li D, He Q, et al. Interleukin-18 promoter polymorphisms probability and gradients of severity of idiopathic parkinsonism
and risk of Parkinson's disease in a Han Chinese population. Br Res based on H. pylori antibody profile. Helicobacter 2005; 10(4): 288-
2011; 1381: 90-4. 97.
[30] Lee HJ, Jang SH, Kim H, Yoon JH, Chung KC. PINK1 stimulates
interleukin-1β-mediated inflammatory signaling via the positive
146 CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 Anderson and Maes

[51] Dobbs RJ, Charlett A, Dobbs SM, et al. Towards defining a [72] Engler H, Doenlen R, Riether C, et al. Time-dependent alterations
rigidity-associated pathogenic pathway in idiopathic parkinsonism. of peripheral immune parameters after nigrostriatal dopamine
Neurodegener Dis 2012; 10(1-4): 183-6. depletion in a rat model of Parkinson's disease. Brain Behav
[52] McMahon BJ, Hennessy TW, Bensler JM, et al. The relationship Immun 2009; 23(4): 518-26.
among previous antimicrobial use, antimicrobial resistance, and [73] Krstic D, Madhusudan A, Doehner J, et al. Systemic immune
treatment outcomes for Helicobacter pylori infections. Ann Intern challenges trigger and drive Alzheimer-like neuropathology in
Med 2003; 139(6): 463-9. mice. J Neuroinflamm 2012; 9: 151.
[53] Bjarnason IT, Charlett A, Dobbs RJ, et al. Role of chronic infection [74] Rohn TT, Catlin LW. Immunolocalization of influenza A virus and
and inflammation in the gastrointestinal tract in the etiology and markers of inflammation in the human Parkinson's disease brain.
pathogenesis of idiopathic parkinsonism. Part 2: response of facets PLoS One 2011; 6(5): e20495.
of clinical idiopathic parkinsonism to Helicobacter pylori [75] Greenbaum L, Rigbi A, Lipshtat N, et al. Association of nicotine
eradication. A randomized, double-blind, placebo-controlled dependence susceptibility gene, CHRNA5: with Parkinson's
efficacy study. Helicobacter 2005; 10(4): 276-87. disease age at onset: Gene and smoking status interaction.
[54] Hamill RW, Tompkins JD, Girard BM, et al. Autonomic Parkinsonism Relat Disord 2013; 19(1): 72-6.
dysfunction and plasticity in micturition reflexes in human α- [76] Palacios N, Gao X, McCullough ML, et al. Caffeine and risk of
synuclein mice. Dev Neurobiol 2012; 72(6): 918-36. Parkinson's disease in a large cohort of men and women. Mov
[55] Machado A, Herrera AJ, Venero JL, et al. Peripheral inflammation Disord 2012; 27(10): 1276-82.
increases the damage in animal models of nigrostriatal [77] Castagnoli K, Petzer JB, Steyn SJ, van der Schyf CJ, Castagnoli N
dopaminergic neurodegeneration: possible implication in Jr. Inhibition of human MAO-A and MAO-B by a compound
Parkinson's disease incidence. Parkinsons Dis 2011; 2011: 393769. isolated from flue-cured tobacco leaves and its neuroprotective
[56] Hernández-Romero MC, Delgado-Cortés, MJP, Sarmiento M, et al. properties in the MPTP mouse model of neurodegeneration.
Peripheral inflammation increases the deleterious effect of CNS Inflammopharmacology 2003; 11: 183-8.
inflammation on the nigrostriatal dopaminergic system. [78] Weinreb O, Amit T, Bar-Am O, Youdim MB. Rasagiline: a novel
Neurotoxicol 2012; 33(3): 347-60. anti-Parkinsonian monoamine oxidase-B inhibitor with
[57] Godoy MCP, Tarelli R, Ferrari CC, Sarchi MI, Pitossi FJ. Central neuroprotective activity. Prog Neurobiol 2010; 92: 330-44.
and systemic IL-1 exacerbates neuro degeneration and motor [79] Akahoshi E, Yoshimura S, Uruno S, Ishihara-Sugano M. Effect of
symptoms in a model of Parkinson’s disease. Brain 2008; 131(7): dioxins on regulation of tyrosine hydroxylase gene expression by
1880-94. aryl hydrocarbon receptor: a neurotoxicology study. Environ
[58] Noyce AJ, Bestwick JP, Silveira-Moriyama L, et al. Meta-analysis Health 2009; 8: 24.
of early nonmotor features and risk factors for Parkinson disease. [80] Anderson G, Beischlag TV, Vinciguerra M, Mazzoccoli G. The
Ann Neurol, 2012; 72(6): 893-901. circadian clock circuitry and the AHR signaling pathway in
[59] Villaran RF, Espinosa-Oliva AM, Sarmiento M, et al. Ulcerative physiology and pathology. Biochem Pharmacol 2013; 85(10):
colitis exacerbates lipopolysaccharide-induced damage to the nigral 1405-16.
dopaminergic system: potential risk factor in Parkinson’s disease. J [81] Luchowska E, Kloc R, Olajossy B, et al. Beta-adrenergic
Neurochem 2010; 114(6): 1687-700. enhancement of brain kynurenic acid production mediated via
[60] Weller C, Oxlade N, Dobbs SM, et al. Role of inflammation in cAMP-related protein kinase A signalling. Prog
gastrointestinal tract in aetiology and pathogenesis of idiopathic Neuropsychopharmacol Biol Psychiatry 2009; 33(3): 519-29.
parkinsonism FEMS Immunol Med Microbiol 2005; 44(2): 129-35. [82] Mayati A, Levoin N, Paris H, et al. Induction of intracellular
[61] Devos D, Lebouvier T, Lardeux B, et al. Colonic inflammation in calcium concentration by environmental benzo(a)pyrene involves a
Parkinson's disease. Neurobiol Dis 2012; 50C, 42-8. β2-adrenergic receptor/adenylyl cyclase/Epac-1/inositol 1:4:5-
[62] Forsyth CB, Shannon KM, Kordower JH, et al. Increased intestinal trisphosphate pathway in endothelial cells. J Biol Chem 2012; 287:
permeability correlates with sigmoid mucosa alpha-synuclein 4041-52.
staining and endotoxin exposure markers in early Parkinson's [83] O’Reilly EJ, McCullough ML, Chao A, et al. Smokeless tobacco
disease. PLoS One 2011; 6(12): e28032. use and the risk of Parkinson’s disease mortality. Mov Disord
[63] Gabrielli M, Bonazzi P, Scarpellini E, et al. Prevalence of small 2005; 1383-4.
intestinal bacterial overgrowth in Parkinson's disease. Mov Disord [84] Muñoz P, Huenchuguala S, Paris I, et al. Segura-Aguilar J, Tizabi
2011; 26(5): 889-92. Y. Protective effects of nicotine against aminochrome-induced
[64] Maes M, Kubera M, Leunis JC. The gut-brain barrier in major toxicity in substantia nigra derived cells: implications for
depression: intestinal mucosal dysfunction with an increased Parkinson's disease. Neurotox Res 2012; 22(2): 177-80.
translocation of LPS from gram negative enterobacte-ria (leaky [85] Huang LZ, Parameswaran N, Bordia T, McIntosh JM, Quik M.
gut) plays a role in the inflammatory pathophysiology of Nicotine is neuroprotective when administered before but not after
depression. Neurol Endocrinol Lett 2008; 29(1): 117-24. nigrostriatal damage in rats and monkeys. J Neurochem 2009; 109:
[65] Hunter RL, Dragicevic N, Seifert K, et al. Inflammation induces 826-37.
mitochondrial dysfunction and dopaminergic neurodegeneration in [86] Ciobica A, Padurariu M, Hritcu L. The effects of short-term
the nigrostriatal system. J Neurochem 2007; 100(5): 1375-86. nicotine administration on behavioral and oxidative stress
[66] Maes M, Kubera M, Obuchowiczwa E, Goehler L, Brzeszcz J. deficiencies induced in a rat model of Parkinson's disease.
Depression’s multiple comorbidities explained by Psychiatr Danub 2012; 24(2): 194-205.
(neuro)inflammatory and oxidative & nitrosative stress pathways. [87] Quik M, Wonnacott S. {alpha}6{beta}2and
Neuroendocrinol Lett. 2011; 32(1): 7-24. {alpha}4{beta}2Nicotinic acetylcholine receptors as drug targets
[67] Chojnacki C, Wisniewska-Jarosinska M, Walecka-Kapica E, et al. for Parkinson’s disease. Pharmacol Rev 2011; 63: 938-66.
Evaluation of melatonin effectiveness in the adjuvant treatment of [88] Quik M, Perez XA, Bordia T. Nicotine as a potential
ulcerative colitis. J Physiol Pharmacol 2011; 62(3): 327-34. neuroprotective agent for Parkinson's disease. Mov Disord 2012;
[68] Hsuchou H, Kastin AJ, Mishra PK, Pan W. C-Reactive Protein 27(8): 947-57.
Increases BBB Permeability: Implications for Obesity and [89] Poisik OV, Shen JX, Jones S, Yakel JL. Functional alpha7-
Neuroinflammation. Cell Physiol Biochem 2012; 30(5): 1109-19. containing nicotinic acetylcholine receptors localize to cell bodies
[69] Gao HM, Liu B, Zhang W, Hong JS. Synergistic dopaminergic and proximal dendrites in the rat substantia nigra pars reticulata. J
neurotoxicity of MPTP and inflammogen lipopolysaccharide: Physiol 2008; 586(5): 1365-7138.
relevance to the etiology of Parkinson’s disease. FASEB J 2003; [90] Holmes AD, Copland DA, Silburn PA, Chenery HJ. Nicotine
17(13): 1957-9. effects on general semantic priming in Parkinson's disease. Exp
[70] Depino AM, Earl C, Kaczmarczyk E, et al. Microglial activation Clin Psychopharmacol 2011; 19(3): 215-23.
with atypical proinflammatory cytokine expression in a rat model [91] Kroker KS, Rast G, Rosenbrock H. Differential effects of subtype-
of Parkinson’s disease. Eur J Neurosc 2003; 18(10): 2731-42. specific nicotinic acetylcholine receptor agonists on early and late
[71] Ling ZD, Chang Q, Lipton JW, Tong CW, Landers TM, Carvey hippocampal LTP. Eur J Pharmacol 2011; 671(1-3): 26-32.
PM. Combined toxicity of prenatal bacterial endotoxin exposure [92] Quik M, Sum JD, Whiteaker P, et al. Differential declines in
and postnatal 6-hydroxydopamine in the adult rat midbrain. striatal nicotinic receptor subtype function after nigrostriatal
Neuroscience 2004; 124(3): 619-28. damage in mice. Mol Pharmacol 2003; 63(5): 1169-79.
TRYCAT Pathways Link Peripheral Inflammation CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 147

[93] Jeyarasasingam G, Tompkins L, Quik M. Stimulation of non- dichotomous response to nicotine in murine models of Th1/Th17-
alpha7 nicotinic receptors partially protects dopaminergic neurons versus Th2-mediated colitis. J Immunol 2011; 187(5): 2677-87.
from 1-methyl-4-phenylpyridinium-induced toxicity in culture. [114] Maes M, Ringe K, Kubera M, et al. Increased autoimmune activity
Neuroscience 2002; 109(2): 275-85. against 5-HT: a key component of depression that is associated
[94] Hosur V, Loring RH. alpha4beta2 nicotinic receptors partially with inflammation and activation of cell-mediated immunity, and
mediate anti-inflammatory effects through Janus kinase 2-signal with severity and staging of depression. J Affect Disord 2012;
transducer and activator of transcription 3 but not calcium or cAMP 136(3): 386-92.
signaling. Mol Pharmacol 2011; 79: 167-74. [115] Koutsilieri E, Lutz MB, Scheller C. Autoimmunity, dendritic cells
[95] Kawamata J, Shimohama S. Stimulating nicotinic receptors trigger and relevance for Parkinson's disease. J Neural Transm 2013;
multiple pathways attenuating cytotoxicity in models of 120(1): 75-81.
Alzheimer’s and Parkinson’s diseases. J Alzheimers Dis 2011; [116] Benkler M, Agmon-Levin N, Hassin-Baer S, et al. Immunology,
24(Suppl 2): 95-109. autoimmunity, and autoantibodies in Parkinson's disease. Clin Rev
[96] Belluardo N, Mudo G, Blum M, Fuxe K. Central nicotinic Allergy Immunol 2012; 42(2): 164-71.
receptors, neurotrophic factors and neuroprotection. Behav Br Res [117] Li J, Yang J, Zhao P, et al. Neuromelanin enhances the toxicity of
2000; 113: 21-34. α-synuclein in SK-N-SH cells. J Neural Transm 2012; 119(6): 685-
[97] Quik M, Cox H, Parameswaran N, et al. Nicotine reduces 91.
levodopa-induced dyskinesias in lesioned monkeys. Ann Neurol [118] Oberländer U, Pletinckx K, Döhler A, et al. Neuromelanin is an
2007; 62: 588-96. immune stimulator for dendritic cells in vitro. BMC Neurosci 2011;
[98] Huang L, Grady SR, Quik M. Nicotine reduces L-dopa-induced 12: 116.
dyskinesias by acting at {beta}2 nicotinic receptors. J Pharmacol [119] He AY, Qiu LJ, Gao Y, et al. The role of oxidative stress in
Exp Ther 2011; 338: 932-41. neuromelanin synthesis in PC12 cells. Neuroscience 2011; 189: 43-
[99] Quik M, Mallela A, Chin M, et al. Nicotine-mediated improvement 50.
in l-dopa-induced dyskinesias in MPTP-lesioned monkeys is [120] Xuan Q, Xu SL, Lu DH, et al. Increased expression of α-synuclein
dependent on dopamine nerve terminal function. Neurobiol Dis in aged human brain associated with neuromelanin accumulation. J
2013; 50: 30-41. Neural Transm 2011; 118(11): 1575-83.
[100]. Thiriez C, Villafane G, Grapin F, et al. Can nicotine be used [121] Pan T, Zhu J, Hwu WJ, Jankovic J. The role of alpha-synuclein in
medicinally in Parkinson’s disease? Expert Rev Clin Pharmacol melanin synthesis in melanoma and dopaminergic neuronal cells.
2011; 4: 429-36. PLoS One 2012; 7(9): e45183.
[101] Liu Y, Hu J, Wu J, et al. α7 nicotinic acetylcholine receptor- [122] Zhang W, Zecca L, Wilson B, et al. Human neuromelanin: an
mediated neuroprotection against dopaminergic neuron loss in an endogenous microglial activator for dopaminergic neuron death.
MPTP mouse model via inhibition of astrocyte activation. J Front Biosci (Elite Ed), 2013; 5: 1-11.
Neuroinflamm 2012; 9: 98. [123] Benkler M, Agmon-Levin N, Shoenfeld Y. Parkinson's disease,
[102] Choi SH, Joe EH, Kim SU, Jin BK. Thrombin-induced microglial autoimmunity, and olfaction. Int J Neurosci 2009; 119(12): 2133-
activation produces degeneration of nigral dopaminergic neurons in 43.
vivo. J Neurosci. 2003; 23: 5877-86. [124] Shi FD, Piao WH, Kuo YP, et al. Nicotinic attenuation of central
[103] Yang YJ, Zhang S, Ding JH, Zhou F, Hu G. Iptakalim protects nervous system inflammation and autoimmunity. J Immunol 2009;
against MPP+-induced degeneration of dopaminergic neurons in 182: 1730-9.
association with astrocyte activation. Int J Neuropsychopharmacol [125] Yanamandra K, Gruden MA, Casaite V, et al. α-synuclein reactive
2009; 12: 317-27. antibodies as diagnostic biomarkers in blood sera of Parkinson's
[104] Hirsch EC, Hunot S, Damier P, Faucheux B. Glial cells and disease patients. PLoS One 2011; 6(4): e18513.
inflammation in Parkinson’s disease: a role in neurodegeneration? [126] Chen X, Ghribi O, Geiger JD. Caffeine protects against disruptions
Annal Neurol 1998; 44(3)(Suppl1): S115-20. of the blood-brain barrier in animal models of Alzheimer's and
[105] Linneberg A, Nielsen NH, Madsen F, et al. Factors related to Parkinson's diseases. J Alzheimers Dis 2010; 20(Suppl1): S127-41.
allergic sensitization to aeroallergens in a cross-sectional study in [127] Ursing C, Wikner J, Brismar K, Röjdmark S. Caffeine raises the
adults: The Copenhagen Allergy Study. Clin Exp Allergy 2001; 31: serum melatonin level in healthy subjects: an indication of
1409-1417. melatonin metabolism by cytochrome P450(CYP)1A2. J
[106] Mishra NC, Rirsimaah J, Boyd RT, et al. Nicotine inhibits Fc Endocrinol Invest 2003; 26(5): 403-6.
epsilon RI-induced cysteinyl leukotrienes and cytokine production [128] Postuma RB, Lang AE, Munhoz RP, et al. Caffeine for treatment
without affecting mast cell degranulation through alpha 7/alpha of Parkinson disease: a randomized controlled trial. Neurology
9/alpha 10-nicotinic receptors. J Immunol 2010; 185(1): 588-96. 2012; 79(7): 651-8.
[107] Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. [129] Markus RP, Silva CL, Franco DG, Barbosa EM Jr, Ferreira ZS. Is
Neuroprotective activities of CD4+CD25+ regulatory T cells in an modulation of nicotinic acetylcholine receptors by melatonin
animal model of Parkinson’s disease. J Leukocyte Biol 2007; relevant for therapy with cholinergic drugs? Pharmacol Ther 2010;
82(5): 1083-94. 126(3): 251-62.
[108] Reynolds AD, Stone DK, Hutter JAL, et al. Regulatory T cells [130] Gao J, Nalls MA, Shi M, et al. An exploratory analysis on gene-
attenuate Th17 cell-mediated nigrostriatal dopaminergic environment interactions for Parkinson disease. Neurobiol Aging
neurodegeneration in a model of Parkinson’s disease. J Immunol 2012; 33(10): 2528.e1-e6.
2010; 184(5): 2261-71. [131] Chen W, Wang H, Wei H, Gu S, Wei H. Istradefylline, an
[109] Wang DW, Zhou RB, Yao YM, et al. Stimulation of α7 nicotinic adenosine A(2A) receptor antagonist, for patients with Parkinson's
acetylcholine receptor by nicotine increases suppressive capacity of Disease: A meta-analysis. J Neurol Sci 2013; 324(1-2): 21-8.
naturally occurring CD4+CD25+ regulatory T cells in mice in vitro. [132] Amano S, Hazama F, Kawai J, Sasahara M. Increased 5'-
J Pharmacol Exp Ther 2010; 335(3): 553-61. nucleotidase activity induced by dibutyryl cyclic AMP treatment of
[110] de Jonge WJ, Ulloa L. The alpha7 nicotinic acetylcholine receptor cultured glial cells. Br Res 1990; 506(2): 254-8.
as a pharmacological target for inflammation. Br J Pharmacol [133] Liebau C, Baltzer AW, Schmidt S, et al. Interleukin-12 and
2007; 151: 915-29. interleukin-18 induce indoleamine 2:3-dioxygenase (IDO) activity
[111] Razani-Boroujerdi S, Boyd RT, Davila-Garcia MI, et al. T cells in human osteosarcoma cell lines independently from interferon-
express alpha7-nicotinic acetylcholine receptor subunits that gamma. Anticancer Res 2002; 22(2A): 931-6.
require a functional TCR and leukocyte-specific protein tyrosine [134] Ren S, Correia MA. Heme: a regulator of rat hepatic tryptophan
kinase for nicotine-induced Ca2+ response. J Immunol 2007; 179: 2:3- dioxygenase. Arch Biochem Biophys 2000; 377(1): 195-203.
2889-98. [135] Maes M, Rief W. Diagnostic classifications in depression and
[112] Wang H, Yu M, Ochani M, et al. Nicotinic acetylcholine receptor somatization should include biomarkers, such as disorders in the
alpha7 subunit is an essential regulator of inflammation. Nature tryptophan catabolite (TRYCAT) pathway. Psychiatry Res 2012;
2003; 421: 384-8. 196: 243-9.
[113] Galitovskiy V, Qian J, Chernyavsky AI, et al. Cytokine-induced [136] Anderson G, Maes M, Berk M. Biological Underpinnings of the
alterations of α7 nicotinic receptor in colonic CD4 T cells mediate Commonalities in Depression, Somatization, and Chronic Fatigue
Syndrome. Med Hypotheses 2012; 78: 752-6.
148 CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 Anderson and Maes

[137] Didier N, Romero IA, Créminon C, et al. Secretion of interleukin- [159] Tanida T, Warita K, Ishihara K, et al. Fetal and neonatal exposure
1beta by astrocytes mediates endothelin-1 and tumour necrosis to three typical environmental chemicals with different
factor-alpha effects on human brain microvascular endothelial cell mechanisms of action: mixed exposure to phenol, phthalate, and
permeability. J Neurochem 2003; 86(1): 246-54. dioxin cancels the effects of sole exposure on mouse midbrain
[138] Knyihár-Csillik E, Chadaide Z, Mihály A, et al. Effect of 6- dopaminergic nuclei. Toxicol Lett 2009; 189(1): 40-7.
hydroxydopamine treatment on kynurenine aminotransferase-I [160] Vogel CF, Goth SR, Dong B, Pessah IN, Matsumura F. Aryl
(KAT-I) immunoreactivity of neurons and glial cells in the rat hydrocarbon receptor signaling mediates expression of indoleamine
substantia nigra. Acta Neuropathol 2006; 112(2): 127-37. 2:3-dioxygenase. Biochem Biophys Res Commun 2008; 375(3):
[139] Silva-Adaya D, Pérez-De La Cruz V, Villeda-Hernández J, et al. 331-5.
Protective effect of L-kynurenine and probenecid on 6- [161] Mezrich JD, Fechner JH, Zhang X, et al. An interaction between
hydroxydopamine-induced striatal toxicity in rats: implications of kynurenine and the aryl hydrocarbon receptor can generate
modulating kynurenate as a protective strategy Neurotoxicol regulatory T cells. J Immunol 2010; 185(6): 3190-8.
Teratol 2011; 33(2): 303-12. [162] Bollen E, Prickaerts J. Phosphodiesterases in neurodegenerative
[140] Lee do Y, Lee KS, Lee HJ, et al. Kynurenic acid attenuates disorders. IUBMB Life 2012; 64(12): 965-70.
MPP(+)-induced dopaminergic neuronal cell death via a Bax- [163] Laugeray A, Launay JM, Callebert J, et al. Peripheral and cerebral
mediated mitochondrial pathway. Eur J Cell Biol 2008; 87(6): 389-97. metabolic abnormalities of the tryptophan-kynurenine pathway in a
[141] Hartai Z, Klivenyi P, Janaky T, et al. Kynurenine metabolism in murine model of major depression. Behav Br Res 2010; 210(1): 84-
plasma and in red blood cells in Parkinson's disease. J Neurol Sci 91.
2005; 239(1): 31-5. [164] Laugeray A, Launay JM, Callebert J, et al. Evidence for a key role
[142] Cosi C, Mannaioni G, Cozzi A, et al. G-protein coupled receptor of the peripheral kynurenine pathway in the modulation of anxiety-
35 (GPR35) activation and inflammatory pain: Studies on the and depression-like behaviours in mice: Focus on individual
antinociceptive effects of kynurenic acid and zaprinast. differences. Pharmacol Biochem Behav 2011; 98: 161-8.
Neuropharmacology 2011; 60: 1227-31. [165] Zmarowski A, Wu HQ, Brooks JM, et al. Astrocyte-derived
[143] Widner B, Leblhuber F, Fuchs D. Increased neopterin production kynurenic acid modulates basal and evoked cortical acetylcholine
and tryptophan degradation in advanced Parkinson's disease. J erelease. Eur J Neurosci. 2009; 29(3): 529-38.
Neural Transm 2002; 109(2): 181-9. [166] Rod NH, Bordelon Y, Thompson A, Marcotte E, Ritz B. Major life
[144] Samadi P, Grégoire L, Rassoulpour A, et al. Effect of kynurenine events and development of major depression in Parkinson's disease
3-hydroxylase inhibition on the dyskinetic and antiparkinsonian patients. Eur J Neurol 2013; 20(4): 663-70.
responses to levodopa in Parkinsonian monkeys. Mov Disord 2005; [167] Decety J, Moriguchi Y. The empathic brain and its dysfunction in
20(7): 792-802. psychiatric populations: implications for intervention across
[145] Schuck PF, Tonin A, da Costa Ferreira G, et al. Kynurenines different clinical conditions. Biopsychosoc Med 2007; 1: 22.
impair energy metabolism in rat cerebral cortex. Cell Mol [168] Bogdanova Y, Cronin-Golomb A. Alexithymia and apathy in
Neurobiol 2007; 27(1): 147-60. Parkinson's disease: Neurocognitive correlates. Behav Neurol
[146] Martín M, Macías M, León J, et al. Melatonin increases the activity 2013; 27(4): 535-45.
of the oxidative phosphorylation enzymes and the production of [169] Poletti M, Frosini D, Pagni C, et al. Alexithymia is associated with
ATP in rat brain and liver mitochondria. Int J Biochem Cell Biol impulsivity in newly-diagnosed, drug-naïve patients with
2002; 34(4): 348-57. Parkinson's disease: an affective risk factor for the development of
[147] Zunszain PA, Anacker C, Cattaneo A, et al. Interleukin-1β: a new impulse-control disorders? J Neuropsychiatry Clin Neurosci 2012;
regulator of the kynurenine pathway affecting human hippocampal 24(4): E36-7.
neurogenesis. Neuropsychopharmacology 2012; 37(4): 939-49. [170] Assogna F, Palmer K, Pontieri FE, et al. Alexithymia is a non-
[148] Nautiyal KM, Dailey CA, Jahn JL, et al. Serotonin of mast cell motor symptom of Parkinson disease. Am J Geriatr Psychiatry
origin contributes to hippocampal function. Eur J Neurosci 2012; 2012; 20(2): 133-41.
36(3): 2347-59. [171] Kirmayer LJ, Dao THT, Smith A. Somatization and
[149] Kanai M, Funakoshi H, Takahashi H, et al. Tryptophan 2:3- psychologization: understanding cultural idioms of distress. In:
dioxygenase is a key modulator of physiological neurogenesis and Clinical Methods in Transcultural Psychiatry, Okpaku S, Ed.;
anxiety-related behavior in mice. Mol Brain 2009; 2: 8. American Psychiatric Press: Washington DC 1998; pp. 233-65.
[150] Potter MC, Elmer GI, Bergeron R, et al. Reduction of endogenous [172] Patki G, Lau YS. Melatonin protects against neurobehavioral and
kynurenic acid formation enhances extracellular glutamate, mitochondrial deficits in a chronic mouse model of Parkinson’s
hippocampal plasticity, and cognitive behavior. Neuropsycho- disease. Pharmacol Biochem Behav 2011; 99(4): 704-11.
pharmacology 2010; 35(8): 1734-42. [173] Sharma R, McMillan CR, Tenn CC, Niles LP. Physiological
[151] Niwa F, Kuriyama N, Nakagawa M, Imanishi J. Effects of neuroprotection by melatonin in a 6-hydroxydopamine model of
peripheral lymphocyte subpopulations and the clinical correlation Parkinson's disease. Brain Res 2006; 1068: 230-6.
with Parkinson's disease. Geriatr Gerontol Int 2012; 12(1): 102-7. [174] Liu J, Somera-Molina KC, Hudson RL, Dubocovich ML.
[152] Katsarou Z, Bostantjopoulou S, Hatzizisi O, et al. Immune factors Melatonin potentiates running wheel-induced neurogenesis in the
or depression? Fatigue correlates in Parkinson's disease. Rev dentate gyrus of adult C3H/HeN mice hippocampus. J Pineal Res
Neurol 2007; 45(12): 725-8. 2013; 54(2): 222-31.
[153] Mihara T, Nakashima M, Kuroiwa A, et al. Natural killer cells of [175] Ramírez-Rodríguez G, Vega-Rivera NM, Benítez-King G, Castro-
Parkinson's disease patients are set up for activation: a possible role García M, Ortíz-López L. Melatonin supplementation delays the
for innate immunity in the pathogenesis of this disease. decline of adult hippocampal neurogenesis during normal aging of
Parkinsonism Relat Disord 2008; 14(1): 46-51. mice Neurosci Lett 2012; 530(1): 53-8.
[154] Rentzos M, Nikolaou C, Andreadou E, et al. Circulating [176] Gutierrez-Valdez AL, Anaya-Martínez V, Ordoñez-Librado JL, et
interleukin-15 and RANTES chemokine in Parkinson's disease. al. Effect of chronic L-dopa or melatonin treatments after
Acta Neurol Scand 2007; 116(6): 374-9. dopamine deafferentation in rats: dyskinesia, motor performance,
[155] Fiszer U, Mix E, Fredrikson S, et al. Gamma delta+ T cells are and cytological analysis. ISRN Neurol 2012; 2012: 360379.
increased in patients with Parkinson's disease. J Neurol Sci 1994; [177] Ono K, Mochizuki H, Ikeda T, et al. Effect of melatonin on α-
121(1): 39-45. synuclein self-assembly and cytotoxicity. Neurobiol Aging 2012;
[156] Darlington PJ, Podjaski C, Horn KE, et al. Innate immune- 33(9): 2172-85.
mediated neuronal injury consequent to loss of astrocytes. J [178] Sae-Ung K, Uéda K, Govitrapong P, Phansuwan-Pujito P.
Neuropathol Exp Neurol 2008; 67(6): 590-9. Melatonin reduces the expression of alpha-synuclein in the
[157] Bencherif M, Lippiello PM, Lucas R, Marrero MB. Alpha7 dopamine containing neuronal regions of amphetamine-treated
nicotinic receptors as novel therapeutic targets for inflammation- postnatal rats. J Pineal Res 2012; 52(1): 128-37.
based diseases. Cell Mol Life Sci 2011; 68(6): 931-49. [179] Rothman SM, Mattson MP. Sleep disturbances in Alzheimer's and
[158] McGeer PL, Itagaki S, Akiyama H, McGeer EG. Rate of cell death Parkinson's diseases. Neuromol Med 2012; 14(3): 194-204.
in parkinsonism indicates active neuropathological process. Ann [180] Fava M, Targum SD, Nierenberg AA, et al. An exploratory study
Neurol 1988; 24(4): 574-6. of combination buspirone and melatonin SR in Major Depressive
TRYCAT Pathways Link Peripheral Inflammation CNS & Neurological Disorders - Drug Targets, 2014, Vol. 13, No. 1 149

Disorder (MDD): A possible role for neurogenesis in drug [192] Gao X, Chen H, Schwarzschild MA, Ascherio A. Use of ibuprofen
discovery. J Psychiatr Res 2012; 46(12): 1553-63. and risk of Parkinson disease. Neurology 2011; 76(10): 863-9.
[181] Tosini G, Ye K, Iuvone PM. N-acetylserotonin: neuroprotect-ion, [193] Rees K, Stowe R, Patel S, Ives N, Breen K, Clarke CE, Ben-
neurogenesis, and the sleepy brain. Neuroscientist 2012; 18(6): Shlomo Y. Non-steroidal anti-inflammatory drugs as disease-
645-53. modifying agents for Parkinson's disease: evidence from obser-
[182] Niranjan R, Nath C, Shukla R. Melatonin attenuated mediators of vational studies. Cochr Database Syst Rev 2011; (11): CD008454.
neuroinflammation and alpha-7 nicotinic acetylcholine receptor [194] Tasaki Y, Yamamoto J, Omura T, et al. Meloxicam ameliorates
mRNA expression in lipopolysaccharide (LPS) stimulated rat motor dysfunction and dopaminergic neurodegeneration by
astrocytoma cells, C6. Free Radic Res 2012; 46(9): 1167-77. maintaining Akt-signaling in a mouse Parkinson's disease model.
[183] Huang LZ, Campos C, Ly J, Ivy Carroll F, Quik M. Nicotinic Neurosci Lett 2012; 521(1): 15-9.
receptor agonists decrease L-dopa-induced dyskinesias most [195] Dairam A, Antunes EM, Saravanan KS, Daya S. Non-steroidal
effectively in partially lesioned parkinsonian rats. Neuropharmaco- anti-inflammatory agents, tolmetin and sulindac, inhibit liver
logy 2011; 60(6): 861-8. tryptophan 2:3-dioxygenase activity and alter brain neurotrans-
[184] Morgan WW, Nelson JF. Chronic administration of pharmacol- mitter levels. Life Sci 2006; 79(24): 2269-74.
ogical levels of melatonin does not ameliorate the MPTP-induced [196] Gallagher PJ, Castro V, Fava M, et al. Antidepressant response in
degeneration of the nigrostriatal pathway. Brain Res 2001; 921: patients with major depression exposed to NSAIDs: a pharmaco-
115-21. vigilance study. Am J Psychiatry 2012; 169(10): 1065-72.
[185] Tapias V, Cannon JR, Greenamyre JT. Melatonin treatment [197] Driver JA, Logroscino G, Lu L, Gaziano JM, Kurth T. Use of non-
potentiates neurodegeneration in a rat rotenone Parkinson's disease steroidal anti-inflammatory drugs and risk of Parkinson's disease:
model. J Neurosci Res 2010; 88: 420-7. nested case-control study. Br Med J 2011; 342: d198.
[186] Mudò G, Belluardo N, Mauro A, Fuxe K. Acute intermittent [198] Padol IT, Wang C, Hunt RH. Altered physiology of acid secretion
nicotine treatment induces fibroblast growth factor-2 in the in depression-prone Flinders rats results in exacerbated NSAID and
subventricular zone of the adult rat brain and enhances neuronal stress-induced gastric damage. Neurogastroenterol Motil 2012;
precursor cell proliferation. Neuroscience 2007; 145(2): 470-83. 24(2): 154-63.
[187] Wei Z, Belal C, Tu W, et al. Chronic nicotine administration [199] Maes M. Targeting cyclooxygenase-2 in depression is not a viable
impairs activation of cyclic AMP-response element binding protein therapeutic approach and may even aggravate the pathophysiology
and survival of newborn cells in the dentate gyrus. Stem Cells Dev underpinning depression. Metab Brain Dis 2012; 27(4): 405-13.
2012; 21(3): 411-22. [200] Yang L, Calingasan NY, Lorenzo BJ, Beal MF. Attenuation of
[188] Knekt P, Kilkkinen A, Rissanen H, Marniemi J, Sääksjärvi K, MPTP neurotoxicity by rolipram, a specific inhibitor of
Heliövaara M. Serum vitamin D and the risk of Parkinson disease. phosphodiesterase IV. Exp Neurol 2008; 211(1): 311-4.
Arch Neurol 2010; 67(7): 808-11. [201] Staines DR, Brenu EW, Marshall-Gradisnik S. Postulated
[189] Santos CM. New agents promote neuroprotection in Parkinson's vasoactive neuropeptide immunopathology affecting the blood-
disease models. CNS Neurol Disord Drug Targets 2012; 11(4): brain/blood-spinal barrier in certain neuropsychiatric fatigue-
410-8. related conditions: A role for phosphodiesterase inhibitors in
[190] Cui X, McGrath JJ, Burne TH, Mackay-Sim A, Eyles DW. treatment? Neuropsychiatr Dis Treat 2009; 5: 81-9.
Maternal vitamin D depletion alters neurogenesis in the developing [202] Kim JS, Bailey MJ, Ho AK, et al. Daily rhythm in pineal
rat brain. Int J Dev Neurosci 2007; 25(4): 227-32. phosphodiesterase (PDE) activity reflects adrenergic/3',5'-cyclic
[191] Zhu Y, Zhou R, Yang R, et al. Abnormal neurogenesis in the adenosine 5'-monophosphate induction of the PDE4B2 variant.
dentate gyrus of adult mice lacking 1:25-dihydroxy vitamin D3 Endocrinology 2007; 148(4): 1475-85.
(1:25-(OH)2 D3). Hippocampus 2012; 22(3): 421-33. [203] Sierksma AS, Rutten K, Sydlik S, et al. Chronic phosphodiesterase
type 2 inhibition improves memory in the APPswe/PS1dE9 mouse
model of Alzheimer's disease. Neuropharmacology 2013; 64: 124-36.

Received: February 20, 2013 Revised: May 25, 2013 Accepted: May 29, 2013

PMID: 23844687

You might also like