2

Immunogenetics of Parkinson’s Disease

Itzia Jimenez-Ferrer Maria Swanberg

Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden

Abstract: Inflammation is a key feature of Parkinson’s disease (PD). In postmortem PD brains, microglial activation and enhanced major histocompatibility class II (MHCII) expression are seen concomitant to the accumulation of alpha-synuclein (α-synuclein) and loss of dopaminergic cells in the substantia nigra. Recent findings showed that α-synuclein epitopes can be presented and recognized by T-cells. PD is not a single disorder; rather, it encompasses a range of clinical, epidemiological, and genetic subtypes. Around 10% of the cases have a monogenic origin, and several of the disease-causing mutations are linked to inflammatory processes. The remaining 90% of the cases are complex, where environmental and genetic risk factors synergize to induce PD pathology. To date, 41 genetic loci have been identified in genome-wide association studies as associated with PD risk, and among these, two are within the HLA region, coding for immune genes including MHCII. Thus, genetic and immune findings indicate that the immune system has a role in the etiology of PD. Experimentally, inflammatory stimuli can cause selective nigral cell loss in preclinical models of PD, and MHCII is required to elicit α-synuclein-induced pathology in mice. In this chapter, we focus on immunogenetics, that is, the relation between genetic risk factors and immune processes in PD.

Keywords: Genetics; HLA; Inflammation; MHCII; Parkinson’s disease

Author for correspondence: Maria Swanberg, Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden. Email: Maria.swanberg@med.lu.se

Doi: http://dx.doi.org/10.15586/codonpublications.parkinsonsdisease.2018.ch2

In: Parkinson’s Disease: Pathogenesis and Clinical Aspects. Stoker TB, Greenland JC (Editors). Codon Publications, Brisbane, Australia. ISBN: 978-0-9944381-6-4; Doi: http://dx.doi.org/10.15586/codonpublications.parkinsonsdisease.2018

Copyright: The Authors.

Licence: This open access article is licenced under Creative Commons Attribution 4.0 International (CC BY 4.0). https://creativecommons.org/licenses/by-nc/4.0/

INTRODUCTION

Parkinson’s disease (PD) is an increasingly prevalent and progressively disabling neurodegenerative disease that encompasses a range of clinical, epidemiological, and genetic subtypes (1). The high inter-individual variation in onset, progression, and symptoms is in part due to a complex interplay between genes and environment. According to the latest criteria by the International Parkinson and Movement Disorders Society, PD diagnosis should be based on the presence of general bradykinesia in combination with either rest tremor, rigidity or both (2). Neuropathologically, PD is characterized by loss of nigral dopaminergic neurons that innervate the striatum and pathological accumulation of α-synuclein in Lewy bodies and Lewy neurites (3). In addition to the neurodegenerative phenotype, local neuroinflammation is a hallmark of PD and includes activation of microglia and astrocytes as well as an upregulation of major histocompatibility class II (MHCII) molecules. The inflammatory activation in PD is not only confined to the brain but also involves the peripheral immune system. One example is the increased expression of inflammatory molecules both in the central (4) and peripheral nervous systems (5). At a cellular level, there is an increased infiltration of immune cells into the brain parenchyma and an altered peripheral leukocyte profile in PD (6). The finding that α-synuclein epitopes can be recognized by T-lymphocytes (7) further strengthens the notion that PD is an inflammatory disease, with both innate and adaptive immune responses. Although these findings strongly link inflammation to PD, they do not answer whether inflammation is a cause or consequence of the disease. However, the recent advances in genetic analyses of familial and idiopathic PD strongly support inflammatory processes to play a critical role in disease etiology.

ETIOLOGY OF PARKINSON’S DISEASE

Genetic studies of familial PD have led to the identification of disease-causing mutations in single genes, that is, monogenic forms of PD. Mutations that have been causatively linked to PD are located to the genes encoding α-synuclein (SNCA), leucine-rich repeat kinase 2 (LRRK2), vacuolar protein sorting-associated protein 35 (VPS-35), parkin (PARK2), PTEN-induced putative kinase 1 (PINK1), DJ1 (PARK7), and glucocerebrosidase (GBA) (8) (Figure 1). Although mutations in these genes are rare and only account for <10% of all PD cases (9), they have identified key molecular players and processes in PD etiology. This can be illustrated by SNCA, which is both neuropathologically and genetically linked to PD. Lewy bodies and Lewy neurites containing α-synuclein accumulations are present in both familial and idiopathic PD, and in addition to SNCA mutations and copy number variations (CNVs) linked to dominantly inherited monogenic PD (10), common genetic variants in SNCA are associated with increased risk of developing idiopathic PD (11).

Fig 1

Figure 1 Insights into the genetics of Parkinson’s disease (PD). The largest meta-analyses until now have identified 41 PD-risk loci (12, 13). Candidate genes are annotated for each region that has been significantly associated with PD. For some of the regions, there is more than one candidate gene.

In 90% of PD patients, there is no monogenic inheritance pattern, and the disease is determined as idiopathic. Idiopathic PD is sometimes referred to as sporadic but has a multifactorial etiology, where environmental and genetic factors interact, synergize, and together determine an individual’s susceptibility to disease. The genetics of idiopathic PD is therefore complex, similar to many other common conditions like Alzheimer’s disease, diabetes, and different forms of cancer. In the quest to understand the etiology of idiopathic PD, efforts have been made to identify genetic variants associated with disease risk. These variants include single nucleotide polymorphisms (SNP; the change of a single base pair) and structural variants (microsatellites, minisatellites, insertions, deletions) that, depending on their frequency in the population, are defined as polymorphisms (>1%) or mutations (<1%). Genome-wide association studies (GWAS) are based on the genetic association analysis of SNPs covering the entire genome. Due to the large number of SNPs examined, the analysis is unbiased, but requires large sample sizes. Meta-analyses of several different GWAS have identified 41 PD risk loci (12, 13), each representing common genetic variants conferring an increased risk of developing PD (Figure 1).

In 1983, exposure to the heroin side product 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was identified as the causative agent for severe irreversible parkinsonism in humans and primates (14). MPTP crosses the blood–brain barrier and is converted to MPP+, which accumulates in dopaminergic neurons through dopamine transporters. MPP+ inhibits complex 1 of the electron transport chain, leading to impaired mitochondria and loss of nigral dopaminergic neurons. Although MPTP is not present as an environmental hazard, it pointed out the potential of the molecule to induce parkinsonism and increased interest in environmental risk factors for PD. Exposure to pesticides such as rotenone, paraquat, organophosphates, and pyrethroids has been associated with increased risk of PD in several case-control studies (15). The mechanisms behind the risk increments are not completely understood but, like MPTP, rotenone and paraquat are thought to induce dopaminergic degeneration through oxidative stress and damage. Rotenone acts on complex 1 of the respiratory chain, while paraquat triggers a redox cycle that generates toxic superoxide free radicals.

Environmental and genetic factors are now considered to act in a synergistic manner and modify the risk for idiopathic PD. One such example is genetic variations in glutathione transferase genes which modify the risk conferred by paraquat exposure (16). The risk of PD has also been reported to be increased by head trauma (17) and recurrent CNS infections (18), while moderate amounts of nicotine, caffeine consumption, and the use of non-steroid anti-inflammatory drugs have been reported to reduce the risk for PD (19). Thus, genetic and environmental factors act together to modify PD risk.

IMMUNOGENETICS OF MONOGENIC PD

Advances in the genomics field have generated unprecedented opportunities to define the genetic basis of complex diseases. By applying a genetic strategy, one can discriminate between causes and consequences of a disease. Furthermore, in situations where a specific gene variant affects the response to therapy, knowledge about an individual’s genotype can inform clinical decisions. Immunogenetics specifically studies the relationship between genetics and the immune system, that is, how genetic variants contribute to the inter-individual variation in immune responses.

Autosomal dominant forms

Interestingly, many of the genetic variants linked to monogenic PD also play a critical role in modulating inflammatory responses. Mutations in LRRK2 account for 1–2% of all PD cases (20, 21), but the prevalence varies substantially depending on the population studied. The penetrance is not complete, meaning environmental factors and/or other genes can modulate the disease-causing effect of LRRK2 mutations. LRRK2 encodes a large protein with multiple functions and has a moderate homology to the receptor-interacting protein kinases, a family of kinases with a known role in immunity. Expression of LRRK2 in microglia is induced by pro-inflammatory stimuli and affects microglial activation (22). LRRK2 is also expressed in many other tissues (23), including peripheral immune cells, where its expression is increased by inflammatory mediators such as interferon-γ (IFNγ) and lipopolysaccharide (LPS) (24, 25). Variants at the LRRK2 locus have been reported to confer increased risk of Crohn’s disease (26) and leprosy (27). In idiopathic PD patients, the expression of LRRK2 in B-lymphocytes, T-lymphocytes, and monocytes is increased compared to controls and is positively correlated with cytokine expression in T-lymphocytes (28). LRRK2 is thus strongly linked to immune processes in the CNS and periphery and is a promising therapeutic target for both monogenic and idiopathic PD.

Mutations in the SNCA gene and the presence of the encoded protein, α-synuclein, in Lewy bodies were described in 1997 (29, 30), revealing the functional link between α-synuclein and PD. α-synuclein is a nuclear and presynaptic protein, and its overexpression and aggregation within neuron somas and neurites precedes neurodegeneration of dopaminergic cells. Several animal models have been developed that overexpress human α-synuclein in dopaminergic neurons (31), leading to α-synuclein accumulation, dopaminergic neurodegeneration, and microglial activation (3234). Human macrophages upregulate α-synuclein after LPS stimulation (35), while microglia from mice lacking α-synuclein present a highly activated phenotype in terms of cytokine profile and morphology (36, 37). α-synuclein is a ligand for toll-like receptor 2 (TLR2) on microglia (38), linking α-synuclein to the innate immune system. TLR2 is also present on T-lymphocytes, B-lymphocytes, monocytes, and macrophages, cells that are part of the adaptive immune system. Recently, it was reported that α-synuclein epitopes can be presented on MHC molecules and activate both helper and cytotoxic T-lymphocytes (7). α-synuclein can thus elicit both innate and adaptive immune responses.

Autosomal recessive forms

Parkin, PINK1, and DJ-1 are linked to autosomal juvenile recessive parkinsonism. These three genes are involved in mitochondrial function and oxidative stress and are also coupled to immune responses. Although loss-of-function mutations in the parkin gene (encoding an E3 ubiquitin ligase) cause early loss of dopaminergic neurons in patients, parkin-deficient mice do not display nigrostriatal pathway degeneration unless they are challenged with low dose of LPS (39). The need of an inflammatory stimulus suggests that the loss of parkin function increases the vulnerability of nigral dopaminergic neurons to inflammation-related degeneration or vice versa. Gene expression profiling in PINK1-deficient mice showed that loss of PINK1 altered the expression of immunomodulatory genes in the striatum (40). In addition, systemic LPS treatment induced higher levels of the pro-inflammatory cytokines interleukin (IL)-1β, IL-12, and tumor necrosis factor α (TNFα) in brain homogenates from PINK1-deficient mice compared to wild-type mice. DJ-1 is implicated in mitochondrial function as a regulator of oxidative stress rather than mitophagy (41). In the human brain, DJ-1 is mostly expressed by astrocytes (42), and astrocytes from DJ-1-deficient mice display an augmented response to LPS and produce more inflammatory cytokines such as IL-6, possibly via increased activation of MAPK p38 and JNK (43). Loss-of-function of parkin, PINK1, and DJ-1 thus seem to increase the sensitivity of dopaminergic neurons to degeneration through oxidative stress and pro-inflammatory immune responses.

IMMUNOGENETICS OF IDIOPATHIC PD

The availability of high-throughput technologies has allowed genotyping of hundreds of thousands to millions of SNPs in the human genome in a cost and time-efficient manner. These technological advancements allow large-scale GWAS, which identify associations between genetic variants and a particular trait or disease. In case-control studies, SNP allele frequencies are compared between patients and controls (44). To date, meta-analyses of GWAS have identified 41 risk loci associated with PD (12, 13). One of the challenges of association studies is the identification of the casual variants, which are most likely genetic variants in linkage disequilibrium (LD) with genotyped SNPs. The consequence of LD, that genetic variants located on the same chromosome have a distance-dependent likelihood of a recombination event during meiosis, is that closely located variants often are inherited together. In addition, associated SNPs can be attributed to different candidate genes and biological function depending on the genetic map used and the availability of gene expression data. Most PD-associated variants confer relatively small risk increments, and the majority are found in non-coding regions regulating gene expression. Such variants are also known as expression quantitative trait loci (eQTLs) and can regulate the expression of multiple genes. Allele-dependent expression of immune-related genes has been reported for eQTLs near or in SNCA, LRRK2, HLA-DQB1, and MAPT (45), with antigen presentation being the most enriched regulated process. Below, we discuss the immune functions of HLA (Figure 2) and other risk loci identified for idiopathic PD (Figure 3) in two GWAS meta-analyses (12, 13).

Fig 2

Figure 2 Single nucleotide polymorphisms (SNPs) in the human leukocyte antigen (HLA) locus associated with increased risk for Parkinson’s disease (PD). Map of HLA class I, II, and III regions indicating alleles and SNPs associated with PD. An asterisk (*) denotes that the SNP is acting as an expression quantitative trait locus (eQTL). (Adapted from Ref. 85).

Fig 3

Figure 3 Insights into the immunogenetics of Parkinson’s disease (PD). Schematic illustration of the link between genetic risk factors and immune mechanisms underlying PD development. Genes (indicated in bold italics) represent nominated risk genes for idiopathic and/or monogenic PD.

Antigen presentation

From the 41 risk loci identified by GWAS, two are within the human leukocyte antigen (HLA) region. HLA is one of the most polymorphic regions in the human genome and presents a complex combination of alleles in high LD (Figure 2). HLA class I and class II genes encode MHCI and MHCII molecules that present antigens to CD8+ and CD4+ T-lymphocytes, respectively, and thereby regulate adaptive immune responses. Different HLA alleles encode MHC molecules with different antigen-binding affinity and are associated with numerous disorders, including autoimmune diabetes and rheumatoid arthritis (46). A combined GWAS of PD with type 1 diabetes, Crohn’s disease, ulcerative colitis, rheumatoid arthritis, celiac disease, psoriasis, and multiple sclerosis identified 17 loci shared between PD and these autoimmune disorders (47). Most of the PD risk alleles, including HLA-DQB1, HLA-DRB5, MAPT, and LRRK2, also increased the risk for the autoimmune disorders. Others, including BOLA2, SETD1A, CXCR4, IL12A, and GAK, had opposite effects. The identification of common genetic pathways for PD and autoimmune disorders further strengthens the importance of immunogenetics and immune therapy in PD.

Several studies have found association between SNPs and alleles in the HLA class II region and PD. These are summarized in Tables 1 and 2 and outlined in Figure 2. Using a GWAS approach, Hamza et al. reported a non-coding variant in HLA-DRA (rs3129882) associated with late-onset PD (48). This variant has been reported to be a cis-acting eQTL that correlates significantly with expression levels of HLA-DRA, DRB5, and DQA2 (49, 50). Studies following the GWAS approach, conducted in a Dutch population by the International Parkinson Disease Genomics Consortium, confirmed the association of the HLA class II region (rs4248166 and chr6:32588205, respectively) with PD (51, 52). Another study reported the presence of three HLA class II variants (not in LD) to be significantly associated with PD risk (53). Taken together, several studies confirm the association of the HLA class II region with PD risk and suggest associated variants to be eQTLs, that is, regulating gene transcription. This could provide a functional link to the increased expression of MHCII molecules observed in PD brains and affect the interaction between antigen-presenting cells and lymphocytes.

TABLE 1 Key HLA Haplotypes Associated with PD

HLA allele
Association with PD
Meta-analysis p-value
Meta-analysis Odds ratio (OR)
Reference
B*07:02 Risk 3 × 10−4 1.23 (85)
B*40:01 Protective 2 × 10−3 0.76 (85)
C*03:04 Protective 8 × 10−6 0.72 (85)
C*07:02 Risk 2 × 10−4 1.23 (85)
DRB1*04:04 Protective 4 × 10−5 0.65 (85)
DRB1*15:01 Risk 6 × 10−5 1.26 (85)
DRB4*01 Protective 4 × 10−5 0.83 (85)
DRB5*01 Risk 5 × 10−5 1.25 (85)
DQA1*01:02 Risk 1 × 10−3 1.17 (85)
DQA1*03:01 Protective 1 × 10−6 0.77 (85)
DQB1*03:02 Protective 7 × 10−6 0.74 (85)
DQB1*06:02 Risk 4 × 10−5* 1.26* (86)

Class I and class II HLA alleles associated with PD together with meta-analysis data for p-values and odds ratios. HLA, human leukocyte antigen; PD, Parkinson’s disease.

TABLE 2 SNPs in the HLA Region Associated with PD

SNP
Allele/gene
Tissue
p-value
Effect size
Data base/original article
rs3129882 DRA4 9 × 10−11 1.30* (48)
DRB6 Brain [6.4 × 10−7; 2.4 × 10−7] [0.52; 0.58] Gtex
Whole blood 2.7 × 10−17 0.45 Gtex
DRB5 Hypothalamus 1.6 × 10−5 −0.44 Gtex
Whole blood 6.6 × 10−9 −0.25 Gtex
DQA2 Whole blood 3.3 × 10−5 0.27 Gtex
C4A Whole blood 3.7 × 10−5 −0.24 Gtex
DQB1-AS1 Whole blood 4.4 × 10−5 −0.16 Gtex
DRB9 Whole blood 1.5 × 10−5 0.22 Gtex
rs660895 DRB1-DQA1 8 × 10−7 0.80* (87)
DQA1 Whole blood 1.3 × 10−6 −0.18 Gtex
DQA2 Brain [6.0 × 10−17; 1.6 × 10−9] [0.83; 1.1] Gtex
Substantia nigra 6.2 × 10−9 0.95 Gtex
DQB1 Brain [6.8 × 10−6; 9.7 × 10−6] [−0.55; −0.65] Gtex
Whole blood 3.3 × 10−12 −0.41 Gtex
DQB1-AS1 Whole blood 2.2 × 10−6 −0.24 Gtex
DQB2 Whole blood 7.9 × 10−13 0.52 Gtex
DRB1 Cortex 5.5 × 10−6 0.58 Gtex
Whole blood 5.3 × 10−14 −0.21 Gtex
DRB6 Brain [7.8 × 10−7; 5.4 × 10−6] [0.59; 0.58] Gtex
Whole blood 5.3 × 10−14 −0.21 Gtex
LY6G5B Whole blood 1.4 × 10−5 −0.12 Gtex
rs2395163 DRA/BTNL2 3 × 10−11 0.81* (88)
DAQ1 Whole blood 7.8 × 10−6 −.017 Gtex
DQA2 Brain [1.2 × 10−9; 1.2 × 10−6] [0.73; 0.83] Gtex
Whole blood 8.4 × 10−31 0.89 Gtex
DQB1 Whole blood 7.5 × 10−7 −0.30 Gtex
DQB2 Whole blood 1.0 × 10−8 0.43 Gtex
DRB1 Brain [5.4 × 10−7; 3.8 × 10−5] [−0.45; −0.46] Gtex
Whole blood 1.7 × 10−16 −0.23 Gtex
DRB6 Brain [3.6 × 10−5; 5.9 × 10−6] [0.55; 0.57] Gtex
Whole blood 1.2 × 10−10 0.46 Gtex
rs9275326 DQB1 1.19 × 10−12 0.826* (13)
DQA1 Whole blood 3.9 × 10−5 −0.20 Gtex
DQB1 Whole blood 1.4 × 10−7 −0.41 Gtex
DQA2 Brain [2.5 × 10−9; 4.1 × 10−7] [1.0; 1.1] Gtex
Whole blood 1.9 × 10−20 0.96 Gtex
DRB1 Whole blood 6.1 × 10−8 −0.20 Gtex
DRB6 Brain [1.1 × 10−7; 2.1 × 10−5] [0.79; −0.94] Gtex
Whole blood 1.4 × 10−6 0.45 Gtex
TAP2 Whole blood 67 × 10−8 −0.28 Gtex
rs9268515 4 × 10−4 1.25* (53)
rs4248166 DRA/BTNL2 0.07 1.08* (51)
rs75855844 DRAB5 4 × 10−4 1.25* (13)

Summarized eQTL data from publically available databases (Gtex) for whole blood and brain regions. An asterisk (*) denotes values from meta-analyses. p-values were generated for each variant-gene pair by testing the alternative hypothesis that the slope of a linear regression model between genotype and expression deviates from 0. The effect size of the eQTLs is defined as the slope of the linear regression and is computed as the effect of the alternative allele relative to the reference allele in the human genome reference GRCh37/hg19 (i.e., the eQTL effect allele is the alternative allele). HLA, human leukocyte antigen; PD, Parkinson’s disease; SNP, single nucleotide polymorphism.

T- and B-lymphocyte development

From the 41 PD-risk loci identified by GWAS (Figure 1), ITPKB, PDLMI2, SATB1, and BST1 are involved in T- or B-lymphocyte development. Inositol 1,4,5-trisphosphate 3-kinase B (ITPKB) controls positive selection of T-lymphocytes and modulates Erk activity, an important kinase that regulates extracellular signal response and plays a crucial role in the production of pro-inflammatory cytokines and chemokines. Studies in mice have shown that nonsense mutations in ITPKB attenuate Erk signaling in T-lymphocytes (54) and that ITPKB-deficiency leads to defects in B-lymphocyte survival, developmental alterations of B-lymphocytes, and antigen unresponsiveness in vivo (55). PDLIM2 has been reported to inhibit T-helper 17 (TH17) cell development through signal transducer and activator of transcription 3 (STAT3). PDLIM2 deficiency in mice resulted in the accumulation of STAT3 in the nucleus and enhanced the extent of TH17 cell differentiation, known to have a pathogenic role in inflammatory diseases (56). SATB1 encodes for special AT-rich binding protein 1, a T-lymphocyte-enriched transcription factor and chromatin organizer essential for controlling a large number of genes participating in T-lymphocyte development and activation (57). Moreover, it has been observed that mouse SATB1 coordinates the expression of Th2 cytokine genes (58). BST1 encodes for the leukocyte surface protein CD157 that is upregulated in bone marrow cells from patients diagnosed with rheumatoid arthritis (59) and may facilitate pre-B-lymphocyte growth.

NF-κB and IFNγ-signaling

Four loci reported to be associated with PD relate to the transcription factor NF-κB that regulates a number of immune genes in response to different stimuli. These loci include MCCC1 and DDRGK1 (12) (Figure 1) as well as RIT2 and SCARB2 reported in an earlier GWAS meta-analysis (13). MCCC1 knockdown strongly inhibits induction of IFNs and inflammatory cytokines in response to viral infection (60). It has also been observed that expression patterns of RIT2 and IFNγ are positively correlated in PD brains, indicating that RIT2 may modulate IFNγ signaling (61). Depletion of DDRGK1 dramatically inhibits the expression of NF-κB target genes, suggesting that DDRGK1 plays an important role in regulating the NF-κB signaling pathway through interaction with IκBα (62). SCARB2 is a known receptor for GBA and for enterovirus 71 (EV1) and is highly expressed in human plasmacytoid dendritic cells where it has been reported to regulate the production of type I IFN through TLR9 and IFN regulatory factor 7 (63).

Regulation of inflammation through metabolic pathways

Biallelic mutations in GBA cause Gaucher’s disease, and carriage of one mutated GBA allele substantially increases the risk for PD (64). Although GBA mutations are the single largest risk factor for idiopathic PD, the mechanisms behind the risk increment are not fully understood. There are several immune-related effects of GBA deficiency, including multisystem inflammation, B-lymphocyte hyperproliferation (65), increased levels of pro-inflammatory cytokines (66), microglial activation and astrogliosis (67). Less is known about the role of GPNMB, SREBF1, and ACMSD in conferring increased risk for PD. GPNMB encodes for glycoprotein nonmetastatic melanoma B and is highly expressed in microglia after LPS treatment. Inhibition by GPNMB siRNA dramatically suppressed the expressions of TNF-α, IL-1β, and inducible nitric oxide synthase (iNOS) in activated mouse BV2 cells, indicating a role in microglial activation and pro-inflammatory cytokine release (68). SREBF1 may regulate innate immune responses through its actions on lipid metabolism since it contributes to resolution of pro-inflammatory TLR4 signaling by reprogramming fatty acid metabolism (69). ACMSD has a well-described biological function in the kynurenine pathway, where it regulates and limits the formation of quinolinic acid. Quinolinic acid is an NMDA receptor agonist with excitotoxic properties that can also modulate inflammatory responses. ACMSD could therefore reduce inflammation-induced neurodegeneration (70).

Innate immune response

PD risk-loci linked to innate immune responses include TLR9, IL1R2, and ATP6V0A1. TLR9 is part of the toll-like receptor family and can recognize mitochondrial DNA as an endogenous danger-associated molecular pattern (DAMP) and activate an inflammatory cascade (71). IL-1 receptor type 2 (IL1R2) acts as a decoy receptor for IL1 by competing with IL1R1 for ligands and co-receptors. IL1R2 has been implicated in arthritis, endometriosis, organ transplantation, and Alzheimer’s disease (72). In vitro, the expression of IL-1R2 is suppressed by pro-inflammatory agents like LPS and IFN-γ (73). The ATP6V0A1 gene is expressed in microglia and their precursors and is involved in the acidification of intracellular compartments and the phagosomal fusion, a process that is crucial for phagocytosis (74). In addition to these, many gene variants conferring increased risk for PD act, in some way, on the complement system. These include SNCA, MAPT, GBA, STK39, LRRK2, HLA, GPNM8, GCH1, DDRGK1, SCARB2, FGF20, and SREBF1 (75).

ENVIRONMENTAL FACTORS AFFECTING IMMUNOGENETICS IN PARKINSON’S DISEASE

As mentioned above, the incomplete penetrance of monogenic forms of PD and the complex genetic structure of idiopathic PD suggest the presence of environmental components that modify disease risk. The link between inflammation and PD genetic risk factors described above is strong, but how immunogenetics interacts with environmental factors is a research field still in development. For example, a SNP (rs3129882) in HLA-DRA associated with increased MHCII molecule expression has been reported to significantly increase the risk of PD in synergy with environmental exposure to pyrethroid (76). The use of nonsteroidal anti-inflammatory drugs (NSAIDs) has been suggested to be neuroprotective, with ibuprofen being significantly associated with a reduced risk for PD (77, 78). Any interaction between the effect of NSAIDs and genetic risk factors for PD is, however, not known.

The gastrointestinal tract has an extensive immune and neuronal network and is in direct contact with the external environment. According to the Braak observations (79), the enteric nervous system is affected by α-synuclein pathology before the substantia nigra. It has been proposed that PD pathology actually starts in the gut and propagates through the vagus nerve to reach the substantia nigra; however, this hypothesis remains under debate (80). Several of the genes linked to familial PD or associated with idiopathic PD are also linked to the gastrointestinal tract. As mentioned above, GWAS have identified variants at the LRRK2 locus which are also known to be associated with Crohn’s disease (26), and FGF20 has been associated with colitis and has demonstrated therapeutic activity in experimental models of intestinal inflammation (81). The overlapping susceptibility between inflammatory bowel disease and PD suggests that inflammatory processes in the intestines may promote PD pathology. Patients with PD have also been shown to have an altered gut microbiota pattern compared with controls (82, 83), and there is emerging evidence that the microbiota can influence the development of PD. In α-synuclein-overexpressing mice, microbiota were required for α-synuclein pathology, microglial activation, and motor deficits to occur (84). In addition, transplantation with microbiota from PD patients, but not from control subjects, worsened the physical impairment in the α-synuclein-overexpressing mice. These findings suggest the microbiome not only as a risk factor for PD but also as a potential therapeutic target. How genetic factors contribute to the microbiome and its impact on PD risk remains to be determined.

CONCLUSION

Many of the identified gene mutations linked to monogenic PD and common variants associated with idiopathic PD are involved in immune pathways. There is thus increasing evidence that inflammation has a causative role rather than being a consequence of neurodegeneration in PD. The involved pathways include both innate and adaptive immune responses in the CNS and in the periphery. If the risk for PD is, in part, mediated through immune mechanisms, these are obvious targets for therapeutic intervention. The field of immunogenetics in PD is therefore likely to unravel more of the etiology underlying PD, as well as identifying potential targets for novel treatments.

Acknowledgment: Itzia Jimenez Ferrer Carrillo acknowledges the support from CONACyT through an international scholarship for PhD studies.

Conflict of interest: The authors declare no potential conflict of interest with respect to research, authorship and/or publication of this manuscript.

Copyright and permission statement: To the best of our knowledge, the materials included in this chapter do not violate copyright laws. All original sources have been appropriately acknowledged and/or referenced. Where relevant, appropriate permissions have been obtained from the original copyright holder(s).

REFERENCES

  1. Espay AJ, Brundin P, Lang AE. Precision medicine for disease modification in Parkinson disease. Nat Rev Neurol. 2017;13(2):119–26. http://dx.doi.org/10.1038/nrneurol.2016.196
  2. Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015.30(12):1591–601.
  3. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. Alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc Natl Acad Sci U S A. 1998;95(11):6469–73. http://dx.doi.org/10.1073/pnas.95.11.6469
  4. Mogi M, Harada M, Kondo T, Riederer P, Inagaki H, Minami M, et al. Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci Lett. 1994;180(2):147–50. http://dx.doi.org/10.1016/0304-3940(94)90508-8
  5. Qin XY, Zhang SP, Cao C, Loh YP, Cheng Y. Aberrations in peripheral inflammatory cytokine levels in parkinson disease: A systematic review and meta-analysis. JAMA Neurol. 2016;73(11):1316–24. http://dx.doi.org/10.1001/jamaneurol.2016.2742
  6. Kannarkat GT, Boss JM, Tansey MG. The role of innate and adaptive immunity in Parkinson’s disease. J Parkinsons Dis. 2013;3(4):493–514.
  7. Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, et al. T cells from patients with Parkinson’s disease recognize alpha-synuclein peptides. Nature. 2017;546(7660):656–61. http://dx.doi.org/10.1038/nature22815
  8. Deng H, Wang P, Jankovic J. The genetics of Parkinson disease. Ageing Res Rev. 2017;42:72–85. http://dx.doi.org/10.1016/j.arr.2017.12.007
  9. Ross OA. A prognostic view on the application of individualized genomics in Parkinson’s disease. Curr Genet Med Rep. 2013;1(1):52–57. http://dx.doi.org/10.1007/s40142-012-0003-1
  10. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. Alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302(5646):841. http://dx.doi.org/10.1126/science.1090278
  11. Farrer M, Maraganore DM, Lockhart P, Singleton A, Lesnick TG, de Andrade M, et al. Alpha-Synuclein gene haplotypes are associated with Parkinson’s disease. Hum Mol Genet, 2001;10(17):1847–51. http://dx.doi.org/10.1093/hmg/10.17.1847
  12. Chang D, Nalls MA, Hallgrimsdottir IB, Hunkapiller J, van der Brug M, Cai F, et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat Genet. 2017;49(10):1511–6. http://dx.doi.org/10.1038/ng.3955
  13. Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet. 2014;46(9):989–93. http://dx.doi.org/10.1038/ng.3043
  14. Langston JW, Langston EB, Irwin I. MPTP-induced parkinsonism in human and non-human primates--clinical and experimental aspects. Acta Neurol Scand Suppl. 1984;100:49–54.
  15. Nandipati S, Litvan I. Environmental exposures and Parkinson’s disease. Int J Environ Res Public Health. 2016;13(9). http://dx.doi.org/10.3390/ijerph13090881
  16. Goldman SM, Kamel F, Ross GW, Bhudhikanok GS, Hoppin JA, Korell M, et al. Genetic modification of the association of paraquat and Parkinson’s disease. Mov Disord. 2012;27(13):1652–8. http://dx.doi.org/10.1002/mds.25216
  17. Perry DC, Sturm VE, Peterson MJ, Pieper CF, Bullock T, Boeve BF, et al. Association of traumatic brain injury with subsequent neurological and psychiatric disease: A meta-analysis. J Neurosurg. 2016;124(2):511–26. http://dx.doi.org/10.3171/2015.2.JNS14503
  18. Fang F, Wirdefeldt K, Jacks A, Kamel F, Ye W, Chen H. CNS infections, sepsis and risk of Parkinson’s disease. Int J Epidemiol. 2012;41(4):1042–9. http://dx.doi.org/10.1093/ije/dys052
  19. Kieburtz K, Wunderle KB. Parkinson’s disease: Evidence for environmental risk factors. Mov Disord. 2013;28(1):8–13. http://dx.doi.org/10.1002/mds.25150
  20. Paisan-Ruiz C, Jain S, Evans EW, Gilks WP, Simon J, van der Brug M, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron. 2004;44(4):595–600. http://dx.doi.org/10.1016/j.neuron.2004.10.023
  21. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44(4):601–7. http://dx.doi.org/10.1016/j.neuron.2004.11.005
  22. Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, et al. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci. 2012;32(5):1602–11. http://dx.doi.org/10.1523/JNEUROSCI.5601-11.2012
  23. Biskup S, Moore DJ, Rea A, Lorenz-Deperieux B, Coombes CE, Dawson VL, et al. Dynamic and redundant regulation of LRRK2 and LRRK1 expression. BMC Neurosci. 2007;8:102. http://dx.doi.org/10.1186/1471-2202-8-102
  24. Gardet A, Benita Y, Li C, Sands BE, Ballester I, Stevens C, et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol. 2010;185(9):5577–85. http://dx.doi.org/10.4049/jimmunol.1000548
  25. Hakimi M, Selvanantham T, Swinton E, Padmore RF, Tong Y, Kabbach G, et al. Parkinson’s disease-linked LRRK2 is expressed in circulating and tissue immune cells and upregulated following recognition of microbial structures. J Neural Transm (Vienna). 2011;118(5):795–808. http://dx.doi.org/10.1007/s00702-011-0653-2
  26. Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet. 2008;40(8):955–62. http://dx.doi.org/10.1038/ng.175
  27. Zhang FR, Huang W, Chen SM, Sun LD, Liu H, Li Y, et al. Genomewide association study of leprosy. N Engl J Med. 2009;361(27):2609–18. http://dx.doi.org/10.1056/NEJMoa0903753
  28. Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, et al. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis. 2017;3:11. http://dx.doi.org/10.1038/s41531-017-0010-8
  29. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276(5321):2045–7. http://dx.doi.org/10.1126/science.276.5321.2045
  30. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–40. http://dx.doi.org/10.1038/42166
  31. Decressac M, Mattsson B, Lundblad M, Weikop P, Bjorklund A. Progressive neurodegenerative and behavioural changes induced by AAV-mediated overexpression of alpha-synuclein in midbrain dopamine neurons. Neurobiol Dis. 2012;45(3):939–53. http://dx.doi.org/10.1016/j.nbd.2011.12.013
  32. Sanchez-Guajardo V, Febbraro F, Kirik D, Romero-Ramos M. Microglia acquire distinct activation profiles depending on the degree of alpha-synuclein neuropathology in a rAAV based model of Parkinson’s disease. PLoS One. 2010;5(1):e8784. http://dx.doi.org/10.1371/journal.pone.0008784
  33. Harms AS, Cao S, Rowse AL, Thome AD, Li X, Mangieri LR, et al. MHCII is required for alpha-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J Neurosci. 2013;33(23):9592–600. http://dx.doi.org/10.1523/JNEUROSCI.5610-12.2013
  34. Jimenez-Ferrer I, Jewett M, Tontanahal A, Romero-Ramos M, Swanberg M. Allelic difference in Mhc2ta confers altered microglial activation and susceptibility to alpha-synuclein-induced dopaminergic neurodegeneration. Neurobiol Dis. 2017;106:279–90. http://dx.doi.org/10.1016/j.nbd.2017.07.016
  35. Tanji K, Mori F, Imaizumi T, Yoshida H, Matsumiya T, Tamo W, et al. Upregulation of alpha-synuclein by lipopolysaccharide and interleukin-1 in human macrophages. Pathol Int. 2002;52(9):572–7. http://dx.doi.org/10.1046/j.1440-1827.2002.01385.x
  36. Austin SA, Floden AM, Murphy EJ, Combs CK. Alpha-synuclein expression modulates microglial activation phenotype. J Neurosci. 2006;26(41):10558–63. http://dx.doi.org/10.1523/JNEUROSCI.1799-06.2006
  37. Austin SA, Rojanathammanee L, Golovko MY, Murphy EJ, Combs CK. Lack of alpha-synuclein modulates microglial phenotype in vitro. Neurochem Res. 2011;36(6):994–1004. http://dx.doi.org/10.1007/s11064-011-0439-9
  38. Kim C, Ho DH, Suk JE, You S, Michael S, Kang J, et al. Neuron-released oligomeric alpha-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat Commun. 2013;4:1562. http://dx.doi.org/10.1038/ncomms2534
  39. Frank-Cannon TC, Tran T, Ruhn KA, Martinez TN, Hong J, Marvin M, et al. Parkin deficiency increases vulnerability to inflammation-related nigral degeneration. J Neurosci. 2008;28(43): 10825–34. http://dx.doi.org/10.1523/JNEUROSCI.3001-08.2008
  40. Akundi RS, Huang Z, Eason J, Pandya JD, Zhi L, Cass WA, et al. Increased mitochondrial calcium sensitivity and abnormal expression of innate immunity genes precede dopaminergic defects in Pink1- deficient mice. PLoS One. 2011;6(1):e16038. http://dx.doi.org/10.1371/journal.pone.0016038
  41. Cookson MR. Parkinsonism due to mutations in PINK1, parkin, and DJ-1 and oxidative stress and mitochondrial pathways. Cold Spring Harb Perspect Med. 2012;2(9):a009415. http://dx.doi.org/10.1101/cshperspect.a009415
  42. Bandopadhyay R, Kingsbury AE, Cookson MR, Reid AR, Evans IM, Hope AD, et al. The expression of DJ-1 (PARK7) in normal human CNS and idiopathic Parkinson’s disease. Brain. 2004;127 (Pt 2): 420–30. http://dx.doi.org/10.1093/brain/awh054
  43. Waak J, Weber SS, Waldenmaier A, Gorner K, Alunni-Fabbroni M, Schell H, et al. Regulation of astrocyte inflammatory responses by the Parkinson’s disease-associated gene DJ-1. FASEB J. 2009;23(8):2478–89. http://dx.doi.org/10.1096/fj.08-125153
  44. Kruglyak L. Prospects for whole-genome linkage disequilibrium mapping of common disease genes. Nat Genet. 1999;22(2):139–44. http://dx.doi.org/10.1038/9642
  45. Pierce S, Coetzee GA. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLoS One. 2017;12(4):e0175882. http://dx.doi.org/10.1371/journal.pone.0175882
  46. Miyadera H, Tokunaga K. Associations of human leukocyte antigens with autoimmune diseases: Challenges in identifying the mechanism. J Hum Genet. 2015;60(11):697–702. http://dx.doi.org/10.1038/jhg.2015.100
  47. Witoelar A, Jansen IE, Wang Y, Desikan RS, Gibbs JR, Blauwendraat C, et al. Genome-wide pleiotropy between Parkinson disease and autoimmune diseases. JAMA Neurol. 2017;74(7):780–92. http://dx.doi.org/10.1001/jamaneurol.2017.0469
  48. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet. 2010;42(9):781–5. http://dx.doi.org/10.1038/ng.642
  49. Stranger BE, Nica AC, Forrest MS, Dimas A, Bird CP, Beazley C, et al. Population genomics of human gene expression. Nat Genet. 2007;39(10):1217–24. http://dx.doi.org/10.1038/ng2142
  50. Montgomery SB, Sammeth M, Gutierrez-Arcelus M, Lach RP, Ingle C, Nisbett J, et al. Transcriptome genetics using second generation sequencing in a Caucasian population. Nature. 2010;464(7289): 773–7. http://dx.doi.org/10.1038/nature08903
  51. Simon-Sanchez J, van Hilten JJ, van de Warrenburg B, Post B, Berendse HW, Arepalli S, et al. Genome-wide association study confirms extant PD risk loci among the Dutch. Eur J Hum Genet. 2011;19(6):655–61. http://dx.doi.org/10.1038/ejhg.2010.254
  52. International Parkinson Disease Genomics C, Nalls MA, Plagnol V, Hernandez DG, Sharma M, Sheerin UM, et al. Imputation of sequence variants for identification of genetic risks for Parkinson’s disease: A meta-analysis of genome-wide association studies. Lancet. 2011;377(9766):641–9. http://dx.doi.org/10.1016/S0140-6736(10)62345-8
  53. Hill-Burns EM, Factor SA, Zabetian CP, Thomson G, Payami H. Evidence for more than one Parkinson’s disease-associated variant within the HLA region. PLoS One. 2011;6(11):e27109. http://dx.doi.org/10.1371/journal.pone.0027109
  54. Wen BG, Pletcher MT, Warashina M, Choe SH, Ziaee N, Wiltshire T, et al. Inositol (1,4,5) trisphosphate 3 kinase B controls positive selection of T cells and modulates Erk activity. Proc Natl Acad Sci U S A. 2004;101(15):5604–9. http://dx.doi.org/10.1073/pnas.0306907101
  55. Marechal Y, Pesesse X, Jia Y, Pouillon V, Perez-Morga D, Daniel J, et al. Inositol 1,3,4,5-tetrakisphosphate controls proapoptotic Bim gene expression and survival in B cells. Proc Natl Acad Sci U S A. 2007;104(35):13978–83. http://dx.doi.org/10.1073/pnas.0704312104
  56. Tanaka T, Yamamoto Y, Muromoto R, Ikeda O, Sekine Y, Grusby MJ, et al. PDLIM2 inhibits T helper 17 cell development and granulomatous inflammation through degradation of STAT3. Sci Signal. 2011;4(202):ra85. http://dx.doi.org/10.1126/scisignal.2001637
  57. Alvarez JD, Yasui DH, Niida H, Joh T, Loh DY, Kohwi-Shigematsu T. The MAR-binding protein SATB1 orchestrates temporal and spatial expression of multiple genes during T-cell development. Genes Dev. 2000;14(5):521–35.
  58. Cai S, Lee CC, Kohwi-Shigematsu T. SATB1 packages densely looped, transcriptionally active chromatin for coordinated expression of cytokine genes. Nat Genet. 2006;38(11):1278–88. http://dx.doi.org/10.1038/ng1913
  59. Ortolan E, Vacca P, Capobianco A, Armando E, Crivellin F, Horenstein A, et al. CD157, the Janus of CD38 but with a unique personality. Cell Biochem Funct. 2002;20(4):309–22. http://dx.doi.org/10.1002/cbf.978
  60. Cao Z, Xia Z, Zhou Y, Yang X, Hao H, Peng N, et al. Methylcrotonoyl-CoA carboxylase 1 potentiates RLR-induced NF-kappaB signaling by targeting MAVS complex. Sci Rep. 2016;6:33557. http://dx.doi.org/10.1038/srep33557
  61. Liscovitch N, French L. Differential co-Expression between alpha-Synuclein and IFN-gamma signaling genes across development and in Parkinson’s disease. PLoS One. 2014;9(12):e115029. http://dx.doi.org/10.1371/journal.pone.0115029
  62. Xi P, Ding D, Zhou J, Wang M, Cong YS. DDRGK1 regulates NF-kappaB activity by modulating IkappaBalpha stability. PLoS One. 2013;8(5):e64231. http://dx.doi.org/10.1371/journal.pone.0064231
  63. Guo H, Zhang J, Zhang X, Wang Y, Yu H, Yin X, et al. SCARB2/LIMP-2 regulates IFN production of plasmacytoid dendritic cells by mediating endosomal translocation of TLR9 and nuclear translocation of IRF7. J Immunol. 2015;194(10):4737–49. http://dx.doi.org/10.4049/jimmunol.1402312
  64. Sidransky E, Lopez G. The link between the GBA gene and parkinsonism. Lancet Neurol. 2012;11(11):986–98. http://dx.doi.org/10.1016/S1474-4422(12)70190-4
  65. Mizukami H, Mi Y, Wada R, Kono M, Yamashita T, Liu Y, et al. Systemic inflammation in glucocerebrosidase-deficient mice with minimal glucosylceramide storage. J Clin Invest. 2002;109(9): 1215–21. http://dx.doi.org/10.1172/JCI0214530
  66. Barak V, Acker M, Nisman B, Kalickman I, Abrahamov A, Zimran A, et al. Cytokines in Gaucher’s disease. Eur Cytokine Netw. 1999;10(2):205–10.
  67. Farfel-Becker T, Vitner EB, Pressey SN, Eilam R, Cooper JD, Futerman AH. Spatial and temporal correlation between neuron loss and neuroinflammation in a mouse model of neuronopathic Gaucher disease. Hum Mol Genet. 2011;20(7):1375–86. http://dx.doi.org/10.1093/hmg/ddr019
  68. Shi F, Duan S, Cui J, Yan X, Li H, Wang Y, et al. Induction of matrix metalloproteinase-3 (MMP-3) expression in the microglia by lipopolysaccharide (LPS) via upregulation of glycoprotein nonmetastatic melanoma B (GPNMB) expression. J Mol Neurosci. 2014;54(2):234–42. http://dx.doi.org/10.1007/s12031-014-0280-0
  69. Oishi Y, Spann NJ, Link VM, Muse ED, Strid T, Edillor C, et al. SREBP1 contributes to resolution of pro-inflammatory TLR4 signaling by reprogramming fatty acid metabolism. Cell Metab. 2017;25(2):412–27. http://dx.doi.org/10.1016/j.cmet.2016.11.009
  70. Espey MG, Chernyshev ON, Reinhard JF, Jr., Namboodiri MA, Colton CA. Activated human microglia produce the excitotoxin quinolinic acid. Neuroreport. 1997;8(2):431–4. http://dx.doi.org/10.1097/00001756-199701200-00011
  71. Celardo I, Martins LM, Gandhi S. Unravelling mitochondrial pathways to Parkinson’s disease. Br J Pharmacol. 2014;171(8):1943–57. http://dx.doi.org/10.1111/bph.12433
  72. Peters VA, Joesting JJ, Freund GG. IL-1 receptor 2 (IL-1R2) and its role in immune regulation. Brain Behav Immun. 2013;32:1–8. http://dx.doi.org/10.1016/j.bbi.2012.11.006
  73. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464(7285):104–7. http://dx.doi.org/10.1038/nature08780
  74. Peri F, Nusslein-Volhard C. Live imaging of neuronal degradation by microglia reveals a role for v0-ATPase a1 in phagosomal fusion in vivo. Cell. 2008;133(5):916–27. http://dx.doi.org/10.1016/j.cell.2008.04.037
  75. Dzamko N, Geczy CL, Halliday GM. Inflammation is genetically implicated in Parkinson’s disease. Neuroscience. 2015;302:89–102. http://dx.doi.org/10.1016/j.neuroscience.2014.10.028
  76. Kannarkat GT, Cook DA, Lee JK, Chang J, Chung J, Sandy E, et al. Common genetic variant association with altered HLA expression, synergy with pyrethroid exposure, and risk for Parkinson’s disease: An observational and case-Control Study. NPJ Parkinsons Dis. 2015;1.
  77. Rees K, Stowe R, Patel S, Ives N, Breen K, Clarke CE, Ben-Shlomo Y. Non-steroidal anti-inflammatory drugs as disease-modifying agents for Parkinson’s disease: Evidence from observational studies. Cochrane Database Syst Rev. 2011;11:CD008454. http://dx.doi.org/10.1002/14651858.CD008454.pub2
  78. Gao X, Chen H, Schwarzschild MA, Ascherio A. Use of ibuprofen and risk of Parkinson disease. Neurology. 2011;76(10):863–9. http://dx.doi.org/10.1212/WNL.0b013e31820f2d79
  79. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197–211. http://dx.doi.org/10.1016/S0197-4580(02)00065-9
  80. Lionnet A, Leclair-Visonneau L, Neunlist M, Murayama S, Takao M, Adler CH, et al. Does Parkinson’s disease start in the gut? Acta Neuropathol. 2018;135(1):1–12. http://dx.doi.org/10.1007/s00401-017-1777-8
  81. Jeffers M, McDonald WF, Chillakuru RA, Yang M, Nakase H, Deegler LL, et al. A novel human fibroblast growth factor treats experimental intestinal inflammation. Gastroenterology. 2002;123(4):1151–62. http://dx.doi.org/10.1053/gast.2002.36041
  82. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 2015;30(3):350–8. http://dx.doi.org/10.1002/mds.26069
  83. Shen X, Yang H, Wu Y, Zhang D, Jiang H. Meta-analysis: Association of Helicobacter pylori infection with Parkinson’s diseases. Helicobacter. 2017;22(5). http://dx.doi.org/10.1111/hel.12398
  84. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016; 167(6):1469–80, e12.
  85. Wissemann WT, Hill-Burns EM, Zabetian CP, Factor SA, Patsopoulos N, Hoglund B, et al. Association of Parkinson disease with structural and regulatory variants in the HLA region. Am J Hum Genet. 2013;93(5):984–93. http://dx.doi.org/10.1016/j.ajhg.2013.10.009
  86. Lampe JB, Gossrau G, Herting B, Kempe A, Sommer U, Fussel M, et al. HLA typing and Parkinson’s disease. Eur Neurol. 2003;50(2):64–8. http://dx.doi.org/10.1159/000072500
  87. Ahmed I, Tamouza R, Delord M, Krishnamoorthy R, Tzourio C, Mulot C, et al. Association between Parkinson’s disease and the HLA-DRB1 locus. Mov Disord. 2012;27(9):1104–10. http://dx.doi.org/10.1002/mds.25035
  88. Pankratz N, Beecham GW, DeStefano AL, Dawson TM, Doheny KF, Factor SA, et al. Meta-analysis of Parkinson’s disease: Identification of a novel locus, RIT2. Ann Neurol. 2012;71(3):370–84. http://dx.doi.org/10.1002/ana.22687