Open access peer-reviewed chapter

Mitochondria at the Base of Neuronal Innate Immunity in Alzheimer’s and Parkinson’s Diseases

Written By

A. Raquel Esteves, Diana F. Silva, Daniel Santos, Emanuel Candeias, Filipa Filipe and Sandra M. Cardoso

Submitted: 29 May 2017 Reviewed: 21 November 2017 Published: 29 August 2018

DOI: 10.5772/intechopen.72612

From the Edited Volume

Mitochondrial Diseases

Edited by Eylem Taskin, Celal Guven and Yusuf Sevgiler

Chapter metrics overview

1,376 Chapter Downloads

View Full Metrics

Abstract

Mitochondria are exceptionally primed to play a key role in neuronal cell survival since they are involved in energy production and function as the metabolic center of cells. Several findings provide evidence for the role of mitochondria in neurodegeneration associated with Alzheimer’s and Parkinson’s diseases (AD and PD). Recent data highlight the role of mitochondrial proteins and mitochondrial reactive oxygen species in the intracellular signaling that regulates innate immunity and inflammation. In this chapter, we will discuss the relevance of the interplay between mitochondria and innate immunity, focusing on mitochondrial damage-associated molecular patterns (DAMPs) and how they can activate innate immunity and elicit AD and PD neurodegenerative process.

Keywords

  • mitochondria
  • neuronal innate immunity
  • Alzheimer’s disease
  • Parkinson’s disease
  • damage-associated molecular patterns

1. Introductory remarks

Mitochondria, derived from an ancestral bacterial endosymbiosis, are important cellular organelles in all cell types, but particularly important in the nervous system, since they are the major source of energy for the brain. Mitochondria are essential for neuronal function and neuronal processes, such as calcium (Ca2+) homeostasis, maintenance of plasma membrane potential, apoptosis, axonal and dendritic transport, release and re-uptake of neurotransmitters at synapses, among others [1, 2]. The brain is particularly vulnerable to oxidative stress due to its high lipid content, its high oxygen demand and its low levels of antioxidant defenses. Therefore, any abnormalities in mitochondria function may impact the aging process and also potentiate the onset of age-dependent neurodegenerative disorders [3, 4].

In Alzheimer’s disease (AD) and Parkinson’s disease (PD), it has been described that mitochondrial metabolism and dynamics are affected not only in susceptible brain areas but also in peripheral cell models, namely platelets, fibroblasts and lymphocytes. Additionally, it was shown in AD and PD cellular and animal models that mitochondrial network is highly fragmented. Mitochondrial fission is required to selectively target dysfunctional mitochondria for degradation by the lysosome in a process called mitophagy [5, 6]. Nevertheless, it was recently proven that mitochondrial fission leads to the exposure of the inner membrane phospholipid, cardiolipin, which serves an important defensive function for the elimination of damaged mitochondria [7]. Since cardiolipin is found only in mitochondrial and bacterial membranes, it is considered a mitochondrial-derived damage-associated molecular pattern (DAMP) that is detected by a Nod-like receptor (NLR), the nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3 (NLRP3) inflammasome Nlrp3 [8]. NLR and toll-like receptors (TLR) are patternrecognition receptors that recognize pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide and short-chain fatty acids, and DAMPs that are responsible for the initiation of innate immune responses. NLR and TLR activation trigger the production of proinflammatory cytokines and antimicrobial peptides (AMPs) [9]. So, it is perceived that also neuronal cells are able to mount an innate immune response. Neurons express critical Toll/interleukin-1 receptor (TIR) domain-containing adaptors that transduce signals of TLR, regulating the expression of various cytokines. Indeed, TLR 3 and 7, localized in the neuronal endosomal compartment, play a role in neurite outgrowth. It is assumed that the cytokines produced by neurons may be just enough to recruit and activate local microglia and may not cause global brain inflammation [10].

Overall, mitochondria play a central role in metabolism, thus allowing the maintenance of cellular homeostasis. In this chapter, we will discuss how mitochondria can regulate neuronal innate immunity and how this impact age-related neurodegenerative disorders, such as AD and PD.

Advertisement

2. Alzheimer’s disease hallmarks

AD is one of the most frequent age-related neurodegenerative disorder, characterized by neuronal loss and gradual cognitive demise. It is the major cause of dementia in the elderly [11], predominantly affects more women than men [12], and is expected that the number of people with AD will triple by the year 2050 [13]. Patients with AD show an impaired ability to perform everyday tasks and often experience psychiatric, emotional and personality disturbances [14]. Two well-known abnormal protein aggregates in the brain of the patients, cerebral cortex and hippocampus, characterize AD pathologically: the neuritic plaques that are extracellular and composed of insoluble amyloid β peptides (Aβ) and neurofibrillary tangles that are intracellular aggregates, mostly consisted of phosphorylated tau, a microtubule-associated protein [15]. It is assumed that oligomers can induce toxicity for neurons causing synaptic dysfunction, neuroinflammation and oxidative stress [16, 17].

Several authors have mentioned that mitochondrial dysfunction and oxidative damage occur in the AD brain before the onset of Aβ pathology. Mitochondrial dysfunction was reported in brain neurons, platelets and fibroblasts from AD patients and in transgenic AD mice models. These mitochondrial abnormalities have been reported in neurons and astrocytes, suggesting that both types of cell might be affected in brains of AD patients [18]. For example, it has been described in post-mortem AD brains, a deficit of cytochrome c oxidase (COX) in hippocampus, frontal, temporal, occipital and parietal lobes [3]. Additionally, it is recognized that mitochondrial DNA (mtDNA) is also involved in the mitochondrial dysfunction having a determinant role in AD pathogenesis. When patient’s mtDNA is transferred into mtDNA-deficient cell lines, the originated ‘cybrids’ reproduce the respiratory enzyme deficiency that occurs in the brain and other tissues in AD, suggesting this defect is carried in part by mtDNA abnormalities [19].

Neuroinflammation has been implicated in AD etiology, but its contribution to disease progression is still not yet understood [20]. Astrocytes and microglial cells are the main type of cells involved in inflammatory responses in the central nervous system (CNS) after infection or injury occurs. Indeed, in this process, cellular and molecular immune components, such as cytokines, are important players, which may lead to the activation of glial cells (microglia and astrocytes) [21]. Several studies have described that Aβ, pathogenic infection or cellular debris triggers an initial inflammatory stimulus, which activates the microglia, allowing the maintenance of neuronal plasticity and synaptic connectivity [22]. Data suggest that microglia internalize and degrade Aβ deposits, helping its clearance from the brain. However, during disease process, microglia acquire a ‘toxic’ phenotype due to chronic activation and continue the production of proinflammatory mediators [23]. In animal models and human brain tissue, both neuritic plaques and neurofibrillary tangles colocalize with activated glial cells. Different studies have reported pathological astrogliosis, in both AD patients and transgenic animal models brains, characterized by an increased glial fibrillary acidic protein (GFAP) and distinct cellular hypertrophy, which is correlated somehow with the severity of cognitive impairment in AD patients [24].

2.1. The role of mitochondrial dysfunction in Alzheimer’s disease etiology

Despite its elusive origin, mitochondrial dysfunction is long recognized as a striking feature of sporadic AD, mediating cell pathways that sustain the disorder progression. Brain bioenergetic function is compromised in AD. Images from fluorodeoxyglucose positron emission tomography (FDG-PET) scan show that glucose utilization is significantly lower in AD subjects as compared to age-matched controls in the cortex and the posterior cingulate brain regions [25]. This bioenergetic compromise correlates with decreased COX activities measured in post-mortem brain tissue from AD patients [26]. Mitochondrial deficits in AD have been described not only in the brain but also in peripheral tissues. COX activity was found decreased in platelets and lymphocytes from AD subjects [27, 28, 29, 30]. This COX deficiency correlates with decreased oxygen consumption first described in AD subject’s brain, where PET scans showed decreased cerebral metabolic rate of oxygen (CMRO2) [31]. Mitochondrial respiration is also compromised in peripheral blood mononuclear cells [32], and in cytoplasmic hybrid (cybrid) cell lines [33], generated by the fusion of mitochondrial DNA (mtDNA) depleted cells with platelets from AD subjects [34]. These cell lines elucidated on the relevance of mtDNA in AD pathology, since the main features of the disease are recapitulated [33, 35, 36]. The same observation was made in a number of transgenic mice models that carry mutations linked to AD familial forms [37, 38, 39].

Along with impaired mitochondrial function, it has been widely demonstrated that mitochondria from AD tissues and models have decreased mitochondrial membrane potential (∆Ψmit) [40]. Cumulative evidence consistently showed a positive correlation between ∆Ψmit and reactive oxygen species (ROS) production [41]. In the case of neurodegenerative disorders, such as AD, associated with dysfunctions of the respiratory chain components, lower ∆Ψmit and decreased activity of the respiratory chain are observed with a simultaneous increase in ROS production [42]. The primary ROS in mitochondria is the superoxide radical anion O2–., mainly produced at complexes I and III [43], which is rapidly converted to H2O2 by mitochondrial dismutases, superoxide dismutase (SOD). Regardless the contradictory data on the contribution of COX deficiency to ROS production [44, 45], oxidative damage is an utterly feature of AD, from human samples to cellular and animal models [36, 46, 47, 48]. Evidence show that mitochondrial dysfunction and ROS production are accentuated by Aβ, a 4 kDa protein, derived from a larger protein, amyloid β-protein precursor (βAPP), that is overproduced during AD progression. Aβ interacts with mitochondrial proteins, namely ABAD, causing increased ROS production, mitochondrial dysfunction and neuronal death [49, 50]. These changes in mitochondrial metabolism seem to be related with morphological alteration of mitochondria of AD tissues and models. Electron microscopy images from AD brain tissue show mitochondria with reduced dimensions and disrupted cristae [51]. Similarly, mitochondria from AD subjects transferred to mtDNA depleted cell into cybrids at an ultrastructural level are small and have a swollen-like structure [52], with a fragmented mitochondrial network that correlates with increased mitochondrial content of dynamin-related protein 1 (DRP1) [53] a key protein for mitochondrial division [54]. Concerning mitochondrial content/mass in AD neurons, the matter is not as straight forward [55]. Vulnerable neurons have a decrease in functional mitochondria, but mtDNA is increased [51]. In accordance it was observed, in AD cybrids, an increase in mtDNA content [33]. This increment was first explained as a compensatory response to counteract the loss of mtDNA transcription efficiency [51], but data gathered on the subject point to decreased mitochondria degradation through autophagy (mitophagy), with imprisoned mitochondria within autophagic vacuoles that are accessible for mtDNA detection [53]. A number of studies have shown autophagy dysfunction as a driving force of AD progression, with important impact on Aβ deposition and plaque formation [56, 57, 58, 59, 60]. In human brain samples, it could be observed a massive accumulation of autophagic vacuoles and lysosome-related vesicles, which led to the conclusion of simultaneous induction and impairment of autophagy [56, 61]. Purified autophagic vesicles contain βAPP and the proteases responsible for its cleavage [56]. Aβ peptides are produced by sequential proteolytic processing of βAPP by β-secretase (BACE) and γ-secretase complex (presenilin and nicastrin) [62, 63]. These accumulated vacuoles cause swellings along dystrophic neurites and potentiate Aβ production and aggregation [64], which gradually form the extracellular amyloid plaques, one of the most prominent brain pathological hallmarks of AD. It is reasonable to argue that stimulating autophagy would clear the cell waste materials. Although some contradictory data were published, in opposition of ameliorating Aβ pathology, stimulating autophagy, either chemically or starvation-induced, fails to degrade accumulating Aβ and worsens cell function in in vivo models [65]. The driver of such failure is the microtubule network, along which autophagic vesicles are transported towards lysosomes, for degradation of cell waste. Mitochondrial metabolism failure compromises microtubule proper dynamics. Destabilized microtubule cytoskeleton negatively impacts autophagic vesicles retrograde transport towards lysosomes and promotes microtubule-associated protein Tau to detach and undergo phosphorylation [5]. Tau is the main component of paired helical filaments (PHF) that form neurofibrillary tangles found in AD brains [66] and is a microtubule-associated protein (MAP) that promotes microtubule assembly and stabilization [67, 68, 69]. Ultrastructural analysis performed in AD neurons found that the number and total length of microtubules are decreased in AD subjects [70]. In AD cybrids, microtubule network is disrupted with increased free tubulin content, and this correlates with increased Tau phosphorylation, comparing with control cybrids [53]. Targeting microtubule stability is able to protect cells from Tau and Aβ-induced toxicity and restores autophagy function in a variety of AD models [71, 72].

2.2. Immune response in Alzheimer’s disease

The role of neuroinflammation in AD dates back to 1907, to the original report of Alois Alzheimer, with microglia surrounding Aβ plaques, thus showing a close relation between the pathway and the disease [73]. Twenty-five years after the postulation of Selkoe and Hardy, the amyloid cascade hypothesis is still the main hypothesis for AD pathogenesis. It is a fact that all AD patients undergo progressive Aβ deposition, and moreover, the sequence of major pathogenic events leading to AD proposed by the amyloid cascade hypothesis is perfectly aligned with the dominantly inherited forms of AD. However, different mechanisms have to be considered to explain the development of AD in sporadic cases, which constitute the vast majority of the cases [74]. Even though Aβ peptide and tau protein oligomers are considered the major contributors to disease progression and the deposition of Aβ occurs decades before any other alterations, there are some missing links between the accumulation and oligomerization of Aβ and tau pathology, synaptic dysfunction and cognitive decline [15, 75]. In this follow-up, neuroinflammation is consistently reported to be deregulated in AD and to facilitate disease progression [76, 77]. Indeed, various forms of Aβ oligomers and aggregates are detected by numerous receptors (TLRs), receptor for advanced glycated end-products (RAGE), CD14, CD36, CD47, α6β1 integrin, class A scavenger receptor and NOD-like receptor family pyrin domains (NLRP) that activate innate immunity response (mainly via MAPK/Erk and NF-κB-mediated signaling) [22, 78, 79, 80]. In neurodegenerative diseases, such as AD, the inflammatory response starts by innate immune system activating monocytes (in periphery) and microglial cells, astrocytes and perivascular cells (in the CNS) [81].

Microglia, the resident macrophages of the CNS, play an active role surveying the brain for pathogens and maintaining neuronal plasticity and synaptic connectivity [82]. In AD, stimulation of microglia involves the microglial polarization to a M1 phenotype that triggers the production of proinflammatory cytokines (TNF-α, IL-1, IL-6, IL-12 and IL-18) [83, 84] and chemokines (CCL2, CCR3, CCR5) [85, 86] and is accompanied by impaired phagocytic capacity [87]. Interestingly, deregulation of Aβ clearance from the CNS is a key pathogenic mechanism in pathology progression, whereas microglial phagocytosis activation plays a crucial role (in combination with the endolysosomal pathway, being Aβ enzymatically digested by neprilysin, insulin-degrading enzyme and matrix metalloprotease proteases) and is controlled by two microglial cell surface receptors: TREM2 (positive regulator) and CD33 (negative regulator) [88, 89]. Moreover, caspases are known mediators of apoptosis, but they also regulate inflammation. Upon binding of Aβ to NLRP, there is an inflammasome-dependent activation of caspase-1 that mediates the production of mature IL-1β by cleavage of an inactive pro-IL-1β peptide [90, 91]. Therefore, elevated concentrations of active caspase 1 detected in the brains of patients with AD [92] are in accordance with the increased NLRP3 activation observed in monocytes from AD patients [93]. In addition, mitochondrial DAMPs were shown to increase AD-associated biomarkers, such as App mRNA, APP protein and Aβ1–42 levels, in SH-SY5Y and mice brains [94, 95]. Together, these studies suggest that mitochondria and mitochondrial DAMPs have the potential to promote inflammation in the brain, with important consequences relevant for neurodegenerative disorders such as AD.

Pathological responses of astrocytes include reactive astrogliosis directed at recovery of injured neural tissue and neuroprotection [96]. In AD, astrocytes, like microglia, are a major source of cytokines (TNF-α and IL-1β are readily released upon astrocytic Aβ detection) [97, 98] and chemokines. Indeed CCL4 has been detected in reactive astrocytes near Aβ plaques [99] and has a high capacity to degrade Aβ1–42 in a more efficient way than their microglial counterparts [100]. In addition, post-mortem brains from AD patients are characterized by hypertrophic reactive astrocytes, elevated GFAP and S100B expression surrounding senile plaques [101]. Interestingly, studies have shown that reactive astrogliosis occurs early in the course of pathogenesis and correlates with the severity of cognitive impairment in AD patients [102]. Furthermore, resident microglia and astrocytes in AD have been shown to stimulate inducible nitric oxide synthase (iNOS) and NADPH oxidase [103]. These upregulations lead to the production of high concentrations of ROS (such as nitric oxide, superoxide, hydrogen peroxide, peroxynitrite), which not only further promote microglia activation but also lead to post-translational modifications (nitration, S-nitrosylation, and dityrosine formation), including Aβ nitration leading to a higher propensity to aggregate and seriously suppress hippocampal LTP [103, 104, 105]. Likewise, the complement system is another major constituent of the innate immune system that shows enhanced levels in disease settings. In the brain, activated microglia and astrocytes are responsible for the production of proteins of the complement system, which in turn are associated with Aβ deposits [106]. Additionally, complement receptor 1 (CR1) modulates the impact of the APOE ε4 allele on brain fibrillar amyloid burden [107]. Furthermore, there are other players with neuroinflammatory actions in AD, such as perivascular macrophages promoting Aβ clearance [108], endothelial cells contributing to the transport of Aβ species between the brain and the periphery [109, 110], oligodendrocytes [111] and neurons [112] that contribute to neuroinflammation by expressing complement components.

In the end, the recruited microglia and astrocytes fail to resolve the Aβ insult effectively, resulting in an excessive proinflammatory cytokine and chemokine production, as well as enhancing DAMPs secretion, ultimately leading to deleterious microglial and astrocytic reactivity [113]. This chronic neuroinflammatory environment thus starts a vicious cycle altering APP processing towards a further increase in Aβ production, culminating in neuronal loss and perpetuating inflammation, which with the advance of the disease compromises blood-brain barrier (BBB) permeability, allowing the invasion of peripheral inflammatory cells that exacerbate the deleterious neuroinflammation and facilitate neurodegeneration [114]. Therefore, neuroinflammation in AD was firstly attributed exclusively to these innate immune sensors of Aβ, contributing to the exacerbation of the disease and viewed only as a response, but in reality the pathway is much more complex.

A decade ago, a significant change in this thought was brought by Wyss-Coray who reviewed the hypothesis that inflammation may serve as a cause and driving force for AD [115]. As seen by the significant immune response later on in the disease and as a response to the Aβ accumulation, it is accurate to state that inflammatory pathways are a driving force in AD. However, for a causative role, inflammation should have an early impact or precede the pathogenesis of the disease [81]. In support of inflammation as a primary contributor for the disease, recent genome-wide association studies (GWASs) of sporadic AD cases (or LOAD—late-onset AD) have found associations between AD and genes that are involved in cholesterol metabolism and in innate immunity [116]. Surprisingly, even Selkoe and Hardy drew attention to the importance of the innate immune system in AD on their update on the status of the amyloid hypothesis [74]. Accordingly, three risk genes have been highlighted: TREM2, CD33 and CR1, and all are involved in some way in microglial response, being upregulated during Aβ plaque development [117, 118, 119]. Another important aspect is the timeline involvement of the immune system response in AD’s development. Analyses from both patients with early AD and mild cognitive impairment (MCI), which precedes AD stage, have identified a correlation between clinical symptoms and the presence of inflammatory markers in the cerebrospinal fluid (CSF), suggesting a much early involvement of the immune system in the disease [120, 121]. Noteworthy, a study in wild-type mice found that chronic inflammatory conditions triggered the development of AD-like neuropathology during aging, demonstrating a case where immune response not only precedes fibrillary Aβ plaque deposition and neurofibrillary tangle formations but also is responsible for their induction [122]. Thus, the possibility to manipulate inflammatory pathways, thereby changing the course of the disease, is yet another indication of the role of inflammation as a driving force of AD pathology. The questions we should make previously of that said manipulation are: Which cells and immune molecules should be modulated? And when should modulation occur? As the activation of microglia and the neuroinflammatory environment are constantly changing depending on the stage of the disease, the time window for modulation and for therapeutically potential is very important [81]. Inefficiency in clinical trials with nonsteroidal anti-inflammatory drugs (NSAIDs) in AD could be largely due to wrong timing of intervention [123], since epidemiological and preclinical studies show a reduction up to 80% in the risk of AD onset and decrease in microglial activation and amyloid burden with NSAID use [124, 125].

As aforementioned, there is an uncontrolled production of cytokines and chemokines that may be used as effective tools for inflammatory biomarkers in AD. Early assessment of neuroinflammation in the AD patients may be an important preventive strategy to act before the detrimental aspects of neuroinflammation, thus averting or delaying any cognitive decline [126]. Several studies have investigated the levels of proinflammatory and anti-inflammatory markers in the CSF, plasma and serum of AD patients. IL-1β, TNF-α and IL-6 have been observed to be altered in the three types of samples in AD, although the results vary according to the time point of sampling [126]. Once again, the stage of the disease is a crucial factor for any therapeutic intervention. Moreover, an increase in TGF-β [127] and S100B [128] levels in the CSF from AD compared to controls has also been reported. Regarding blood-based biomarkers of inflammation, α-1-antichymotrypsin (ACT) [129] and C-reactive protein (CRP) [130] have been shown to be increased in AD. Noteworthy, α-2-macroglobulin (α-2 M) [131] and clusterin (or apolipoprotein J) [132] have been implicated in the pathology of AD, with significant increases in patients, showing promising results as potential plasma biomarkers of AD. Interestingly, many of these inflammatory mediators are also altered in MCI subjects. The levels of IL-8, monocyte chemoattractant protein-1 and interferon-γ-inducible protein 10 are found to be increased in CSF, while IL-1β and TNF-α are increased and apolipoprotein A-1 and complement C1 inhibitor are decreased in blood [126]. Besides the detection of neuroinflammatory markers, inflammation may be also monitored through imaging methods. In patients with AD or MCI subjects, increased microglia activation has been detected by PET scans [133].

The induction of neuroinflammatory effects is not restricted to factors of the CNS and can result from systemic influences [125]. On the one hand, traumatic brain injury is an example of a CNS-intrinsic neuroinflammatory condition that facilitates the development of AD pathology [134]; on the other hand,systemic inflammation may be induced from several chronic diseases [135], such as obesity and T2D, all characterized by CNS inflammation and microglia activation [136, 137]. Therefore, in AD, neuroinflammation can cause and drive pathogenesis [22].

Advertisement

3. Parkinson’s disease hallmarks

PD is the most common movement neurodegenerative disorder characterized by numerous motor symptoms, including tremor, bradykinesia, rigidity and postural instability [138]. PD is twice as common in men than in women, and about 2% of the population above the age of 60 is affected by the disease [139]. PD is characterized by the severe loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and by the presence of intracytoplasmatic proteinaceous inclusions called Lewy bodies, which are primarily composed of fibrillary α-synuclein (SNCA), and ubiquitinated proteins within some remaining nigral neurons [140, 141].

Several evidences from autopsy studies showed that multiple processes are involved in cell death, including oxidative stress, mitochondrial dysfunction, neuroinflammation, excitotoxicity and accumulation of misfolded proteins due to proteasomal and autophagic impairment [142].

Data show that mitochondrial deficits occur in PD patient’s brain neurons, platelets and lymphocytes [139],which play a critical role in the loss of dopaminergic neurons [143]. Furthermore, data suggest that mitochondrial dysfunction can be potentiated by defects in mitochondrial biogenesis caused by the deregulation of transcription factors, such as peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α) [144], which levels are decreased in post-mortem brains of PD and in white blood cells [139]. Recent studies in post-mortem PD brain tissue showed that nigrostriatal axon terminals are dysfunctional, which can alter normal axonal transport. Also, the generation of ROS induces the damage of complexes I and III and protein oxidation in mitochondria and in cytoplasmic proteins, leading to mitochondrial dysfunction [145].

Several studies obtained in post-mortem PD brain tissue, human clinical imaging and fluid biomarker have demonstrated that neuroinflammation is a salient feature and probably an essential contributor to PD pathogenesis [145]. Inflammation associated with oxidative stress and cytokine-dependent toxicity has been described and can lead to both innate and adaptive immune responses. Immune responses can act a secondary response to cellular damage and/ or neuronal loss in the affected regions of the nervous system. These mechanisms imply not only a complex crosstalk between the CNS and the peripheral immune system but also interactions between the brain resident immune cells (microglial cells) and other brain cells (neurons, astrocytes, endothelial cells) [146]. Indeed, it has been described that PD brains show microglial activation and lymphocyte infiltration in the areas of degeneration and an increased expression of inflammatory cytokines with alterations in the composition of peripheral immune cells, suggesting the key role of neuroinflammation in PD.

3.1. The role of mitochondrial dysfunction in Parkinson’s disease etiology

Mitochondrial dysfunction relevance in PD was first documented when 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was associated with parkinsonian syndrome in humans [147]. MPTP is able to cross the blood-brain barrier, is metabolized to 1-methyl-4-phenylpyridinium (MPP+) and is uptaken by dopaminergic neurons, inhibiting mitochondrial respiration at complex I [148]. Complex I activity was shown to be decreased in PD brain samples [149], in peripheral tissues namely platelets and lymphocytes [150] and in PD cybrids [151]. The inhibition of complex I, with MPTP and rotenone, is widely used as in vitro and in vivo models of PD since these recapitulate the main features of the disease [152, 153, 154]. Mitochondrial dysfunction in PD tissues and models is also characterized by a decrease in ∆Ψmit [52, 155, 156]. Accordingly, at a functional level, brain bioenergetics is compromised in PD where PET scans showed glucose utilization are decreased in PD individuals in the occipital cortex compared to control individuals [157].

Oxidative damage driven by mitochondria malfunctioning is a prominent aspect in PD [158]. Mitochondrial complex I is one of the most important sites of ROS production in the cell, primarily O2 [159]. The consequences of oxidative damage are such in PD that oxidative stress was proposed as the cause for dopaminergic neurons death in the SNpc [160, 161]. The same authors found in post-mortem samples from PD subjects increased lipid peroxidation whereas glutathione pathway, an antioxidant defense, is impaired [160]. Mitochondria are the main producers and are also the primary targets of ROS. PD brain biopsies revealed complex I itself is oxidatively damaged, which prevents its proper assembly and function [149]. Although it is incontestable that oxidative stress contributes to PD pathology, it is now generally accepted that ROS are a by-product of mitochondrial dysfunction that contributes to worsen cell demise [162].

Familial forms of PD bearing mutations in mitochondrial proteins reinforced the involvement of mitochondrial dysfunction in PD etiology and shed light into the mechanisms leading to neuronal death, unifying both familial and sporadic cases. Rare mutations causing juvenile PD are related to mitochondrial degradation by mitophagy created an opportunity for clarification of the disease mechanisms. The first identified mutation in PARK2 (Parkin), an E3 ubiquitin ligase, cause early onset PD [163]. The second mutation was identified in PARK6, PTEN-induced kinase 1 (PINK1) and a mitochondrial kinase [164]. PINK1 and Parkin act together in a tightly regulated process to target dysfunctional mitochondria for degradation, named mitophagy. This process is crucial for the maintenance of a healthy pool of mitochondria, potentially protecting cells in early stages of mitochondrial dysfunction [165].In healthy mitochondria, PINK1 levels are maintained low as this protein is degraded within mitochondrial matrix after its import from cytosol [166]. When mitochondria lose their membrane potential, PINK1 is stabilized at their surface recruiting Parkin that, in turn, ubiquitinates and targets mitochondria to undergo mitophagy [167, 168, 169]. PD caused by PINK1 and Parkin mutations is not clinically differentiated from idiopathic PD [170]. Morphologically, PINK1 mutations have drastic repercussions in mitochondria from Drosophila melanogaster to mouse models, with larger, swollen and disrupted cristae [171, 172]. In cybrids from sporadic PD subjects, mitochondria also present abnormal structure with enlarged and scarce cristae [52, 173]. Mitochondrial network images show that in PD models it presents a fragmented structure. From PD cybrids [174] to dopaminergic neurons treated with MPTP [175], a number of models show early mitochondrial fragmentation that precedes cell death. Although DRP1 has been implicated in the fragmentation of mitochondria in PD [174], studies point to SNCA directly interacting with mitochondria inducing fragmentation, in a process that does not require DRP1 [176]. Recently, it was found a common mechanism for mitophagy failure, besides Pink1-Parkin axis, that is shared by familial and sporadic PD, with potential of an early biomarker [177]. In fibroblasts isolated from patients that carry PD mutations and idiopathic PD subjects, it was found an impairment in RHOT1 degradation that in turn delays mitochondria immobilization and consequent degradation [178]. RHOT1 is a mitochondrial kinesin adaptor protein that, upon mitochondrial damage, interacts with PINK1 and Parkin to target mitochondria for proteasomal degradation [179]. Consequently, abnormal levels of autophagy markers were found in brain tissue preparations from PD patients, both sporadic and early onset [180, 181]. This impairment in autophagy has been related to the decreased transport along microtubules and fusion of autophagic vesicles with the lysosomes rather than a defect in cell waste recognition by autophagy machinery [173]. Mitochondrial dysfunction is intimately connected to microtubule instability and, thus, autophagy impairment in PD models. In PD cybrids, intracellular transport of autophagosomes and mitochondria is compromised [173]. Accordingly, MPP+-treated cells have disrupted microtubule network and a decrease in mitochondrial trafficking [182]. Also, there are some data pointing that Parkin can bind to microtubules contributing to their stabilization, whereas ablation of Parkin causes reduced microtubule mass [183, 184]. Accumulation of non-degraded mitochondria and other autophagic substrates, such as SNCA aggregates, increments cell demise and contributes to Lewy body-like structure formation. Oxidative stress provoked by mitochondrial malfunctioning is able to induce proteasomal subunit disassembly, leading to the accumulation of degrading substrates, such as ubiquitin [185], contributing to Lewy body formation and cell death. In fact, ubiquitin accumulation, impaired ubiquitin proteasome system (UPS) function and mitochondrial dysfunction have been proposed to be intimately associated [186].

3.2. Immune response in Parkinson’s disease

Despite PD is characterized by a slow and progressive degeneration of dopaminergic neurons in the SNpc, the cause of this neuronal loss is still poorly understood. Nevertheless, neuroinflammatory mechanisms, such as microglial activation, astrogliosis and lymphocytic infiltration have been postulated to contribute to the cascade of events leading to neuronal degeneration [187].

A growing body of evidence suggests a role of autoimmune and neuroinflammatory mechanisms in the etiopathogenesis of PD [188]. Peripheral immune responses can trigger inflammation and exacerbate neurodegeneration in several neurodegenerative disorders including PD. Indeed, peripheral inflammation in early stages of disease appears to accompany the development of preclinical non-motor symptoms, including olfactory and gastrointestinal dysfunction, providing a possible association between autoimmunity and PD [189]. Strikingly, chronic constipation, which occurs many years before the first motor symptoms of PD, is casually linked to peripheral inflammation [190].

Inflammation is a defense mechanism aimed at counteracting with diverse insults. In neurodegenerative disorders, such as PD, inflammation could results from the activation of innate immunity by PAMPs; DAMPs or protein aggregates. Other than the activation of inflammatory responses, there is also the ability of the immune system to detect harmful agents. Mounting evidence indicates that dopaminergic cell death is influenced by the innate immune system and neuroinflammatory processes in PD. Soreq and coworkers described an altered expression of neuroimmune signaling-related transcripts in early stages of PD [191]. Remarkably, epidemiological studies showed that non-steroidal anti-inflammatory drugs, such as ibuprofen lowers the risk of PD further supporting the contribution of inflammation to disease process [192, 193, 194]. Interestingly, the SNpc (main area affected in PD) exhibit high sensitivity to proinflammatory compounds, whereas the hippocampus appears to be more resistant, which can be explained due to the differences in the number of microglial cells between both areas [195]. In fact, numerous evidences that came from experimental PD models suggest that dopaminergic neurons are extremely vulnerable to inflammatory challenge [196, 197]. Moreover, stereotaxic injection of lipopolysaccharide (LPS, a Gram-negative bacteriotoxin that activates microglial cells) into the SNpc induced degeneration of dopaminergic neurons while sparing GABAergic and serotonergic neurons, suggesting selective dopaminergic neurons vulnerability to PAMPs [198].

There are several factors that may be underlying this selectivity. Dying neurons release substances that are recognized by glial cells, activating them, such as dopamine, neuromelanin and SNCA [199]. Dopamine seems to play a role in the inflammatory response induced by LPS, since depletion of this neurotransmitter prevents gliosis and reduces peripheral macrophages infiltration and dopaminergic neuronal death induced by 6-hydroxydopamine (6-OHDA) [200]. Recently, Dominguez-Meijide and colleagues observed that the decrease in dopamine levels observed in early stages of PD promotes neuroinflammation and disease progression via glial renin-angiotensin system exacerbation [201]. Neuromelanin is able to activate microglia cells leading to neuroinflammatory processes and degeneration of dopaminergic neurons [202, 203]. Extracellular and misfolded SNCA prompts microglia activation and production of proinflammatory molecules [204, 205, 206].

Further support for a role of innate immunity activation in PD pathogenesis come from genetic studies showing that polymorphisms in some proinflammatory cytokines may influence the risk of developing PD. Indeed, there is an association between genetic variations in the human leukocyte antigen (HLA) region and sporadic PD [207, 208]. HLA isalso called human MHC molecules, which presentation activates CD+4 T cells and CD+8 cytotoxic lymphocytes. Remarkably, in a GWA study, several susceptibility loci have been identified as strong risk factors that are related to both innate and adaptive immune functions [209]. Moreover, PD-linked genes such as LRRK2 and SNCA are also known to stimulate inflammatory responses and immunological regulation [210]. In fact, Harms and colleagues reported that accumulation of pathological SNCA in PD brain leads to T cell infiltration, microglial activation and increased production of inflammatory cytokines and chemokines [211]. Furthermore, transgenic mice with overexpression of wild-type or mutated SNCA showed an early microglial activation [212, 213]. Beraud and colleagues demonstrated that misfolded SNCA directly activates microglia, inducing production and release of TNFα and increasing expression of Nfr2-dependent antioxidant enzymes [214]. Aggregated and nitrated SNCA also stimulates microglia activation triggering innate and adaptive immune responses [215]. Intranigral injection of SNCA resulted in the upregulation of mRNA expression of proinflammatory cytokines and the expression of endothelial markers of inflammation and microglial activation [216, 217]. Multiple immune cells show high levels of LRRK2 expression [218, 219]. R1441G LRRK2 mutation was shown to increase proinflammatory cytokine release from activated microglial cells [220, 221]. Moreover, LPS-mediated neuroinflammation is attenuated in murine lrrk2-knockdown brain microglia [222].

The first evidence for a neuroinflammatory processes in PD came in 1988 when McGeer and co-workers observed the presence of activated microglial cells and inflammatory macrophages, as well as, proinflammatory cytokines in post-mortem brain samples of the SNpc of PD patients [223]. Similarly, Langston and coworkers reported an accumulation of activated microglia around dopaminergic neurons in post-mortem human brains with MPTP-induced parkinsonism [224]. Later, several authors corroborated this result and further observed the presence of other markers such as HLA-DP, HLA-DQ, HLADR (CR3/43), CD68 (EBM11, a low-density lipoprotein binding glycoprotein, equivalent to macrosialin in mice) and ferritin in the SNpc and putamen [225, 226, 227]. In addition, intercellularadhesion molecule-1-positive glia levels are also increased in the SNpc of PD brains, indicating activation of cells of the innate immune system, in particular, in areas with neuronal loss and extracellular melanin accumulation [228]. Furthermore, Damien and colleagues used glutathione peroxidase as an astrocytic marker and observed that the density of astrocytes in the SNpc is low when compared to the ventral tegmental area. This indicates that vulnerable neurons in patients with PD have less surrounding astroglial cells and as a result reduced detoxification of oxygen-free radicals by glutathione peroxidase [229]. McGeer and colleagues described for the first time the presence of cytotoxic T lymphocytes (CD8+) in the substantia nigra from one patient with PD [223]. Moreover, several reports found alterations in the population of blood T lymphocytes in PD patients [230, 231, 232]. In addition, cytotoxic infiltration of CD8+ and CD4+ T cells into the brain parenchyma of both post-mortem human PD specimens and in the MPTP mouse model of PD was described during the course of neuronal degeneration [233, 234]. Interestingly, these markers were not detected in the red nucleus suggesting that this infiltration is selective for the injured brain areas. Furthermore, these cells were in close contact with blood vessels and near to melanized dopaminergic neurons. These data indicate that cells migrate from the bloodstream and suggest an interaction between the lymphocytes and the dopaminergic neurons during the neurodegenerative process. Hence, alterations in the BBB might occur in the brains of PD patients. Not only during aging but also in PD, a BBB disruption can occur, leading to an invasion of immune cells, peripheral mediators, toxins and elements of adaptive immunity to the brain parenchyma potentiating the degenerative process [235]. Additionally, PD patients have increased permeability of the intestinal epithelial barrier and a chronic gut inflammation characterized by increased expression levels of proinflammatory cytokines and inflammatory markers [236, 237]. Moreover, several studies reported increase in TNFα, β2-microglobulin, epidermal growth factor (EGF), transforming growth factor α (TGFα), TGFβ1 and interleukins 1β, 6 and 2 levels in the striatum of PD patients and increasein TNFα, interleukin 1β and interferon γ levels in the SN of PD patients [238, 239, 240, 241, 242, 243]. Interestingly, dopaminergic neurons express the receptors for these cytokines, suggesting that they are sensitive to these cytokines [244, 245]. Proinflammatory cytokines, such as TNFα, interleukin 1β, and interferon γ, can induce the expression of the inducible form of nitric oxide synthase (iNOS) or cyclo-oxygenase 2 (COX2), which are known to produce toxic reactive species. To corroborate the previous studies, a CD23-mediated increase in iNOS in the SN of PD patients was found. Furthermore, enzymes that are involved in neuroinflammatory processes mediated by oxidative stress, such as NADPH oxidase, COX2 and myeloperoxidase, are also increased in PD patients [239, 246, 247]. This may indicate that the inflammation-derived oxidative stress could contribute to dopaminergic neuronal degeneration.

The results obtained in post-mortem studies were further corroborated by studies carried out in biological fluids (serum or CSF) of patients suffering from PD. Serum samples from PD patients indicated that the expression of certain cytokines such as IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, TNFα, TNFR1 and RANTES is increased [248, 249, 250, 251, 252, 253, 254]. Interestingly, RANTES levels were correlated with the severity and duration of the disease [255]. Additionally, studies analyzing CSF from PD patients reported proinflammatory changes such as the presence of TNFα [238] and interleukin 1β [225, 256, 257] and osteopontin (a member of the integrins family) [258]. Moreover, PET scan analysis also reported the presence of PK-11195 in PD samples, which is indicative of microglia activation [259, 260]. PET analysis using radioligand 11C-PK-11195 corroborated these results in the SNpc of sporadic PD patients within a year from clinical onset [261]. More recently, microglial activation in PD has been observed with PET by using [18F]-FEPPA [262]. Moreover, it was found a significantly increase numbers of T-helper 17 cells and myeloid-derived suppressor cells in peripheral circulation in PD patients compared with controls [263]. This suggests that a microglial-mediated inflammatory process occurs early in PD process.

It has also been demonstrated that mitochondrial toxins, such as 6-OHDA, MPTP and rotenone, trigger an immune reaction in the striatum and SNpc suggesting that a primary damage to the mitochondrial respiratory chain represents, per se, a trigger for microglial activation and neuroinflammatory processes [264, 265, 266, 267]. This reaction includes activation of microglia and infiltration of CD4+ and CD8+ T cells. Rotenone administration was shown to cause microglial activation not only in rodent models [268] but also in human microglial cell lines [269]. Similarly, a significant increase in the number of activated microglial cells was detected in the brain of 6-OHDA rats, at both nigral and striatal areas [233, 270]. Moreover, in the same model CD+3, CD+4 and CD+8 T cells were abundant and migrated from blood vessels into the SNpc [271]. Additionally, in the brains of both monkeys and mice after systemic injection of MPTP, an activated microglia and infiltration of T-lymphocytes has been observed [197]. Microglial activation was also observed in mice that overexpress SNCA [213], in the SNpc and striatum of rats exposed to 6-OHDA [272, 273] and to MPTP [274].

Interestingly, intranigral or systemic injection of LPS in animals can selectively kill dopaminergic neurons [200, 275, 276, 277, 278, 279]. Furthermore, injection of LPS into pregnant female rats led to offspring with less and abnormal dopaminergic neurons and increased levels of TNFα in the striatum when compared to the controls [280]. Remarkably, the offspring in adulthood were also more susceptible to the effects of parkinsonian toxins than were the controls [281, 282]. Furthermore, the injection of other proinflammatory compounds such as thrombin within the SNpc also induced the death of dopaminergic neurons [283, 284]. These studies suggest that microglia-mediated inflammation underlies the neuronal cell death in the SNpc.

As previously mentioned, microglial cells when activated produce and release toxic oxygen-derived and nitrogen-derived products, which rely on the regulated induction of several enzymatic systems such as NADPH oxidase and iNOS.Indeed, the expression of these biocatalytic systems within the SNpc is significantly increased in PD patient’s post-mortem samples as well as in PD animal models [239, 247]. Oxygen and nitrogen-derived products such as NO, O2 and ONOO can directly cross membranes and enter dopaminergic neurons, which can cause oxidative damage in tyrosine hydroxylase decreasing its enzymatic activity and in SNCA promoting its aggregation [285, 286]. Additionally, activated microglia can release inflammatory cytokines and chemokines, such as TNFα, interleukin 1β and interferon,whichcan induce neurotoxicity via a direct mechanism through receptor binding on dopaminergic neurons or an indirect mechanism through glial-cell activation and expression of inflammatory factors. In fact, chronic adenoviral expression of TNFα in the SNpc of rats can cause time-dependent dopaminergic cell death [287].

Advertisement

4. The interplay between mitochondria and innate immunity

In response to microbial infection, the mammalian innate immune system recognizes invading microorganisms and orchestrates a proinflammatory immune response to eliminate the undesired pathogens and infected cells. The sensing of the infection by the innate immune system is mediated by a variety of pattern recognition receptors (PRRs), which recognize molecular patterns conserved among microbial species known as PAMPs. For detailed information regarding the different families of receptors, respective PAMPs recognition, and the intracellular signaling cascades triggered, see reference [288]. Interestingly, even in the absence of microbial infection, PRRs sense and orchestrate inflammatory responses through recognition of intracellular molecules known as DAMPs. DAMPs are endogenous molecules sequestered within cellular compartments of healthy cells, which, upon injury or stress, are released to trigger sterile proinflammatory immune responses.

Recent insights revealed that mitochondria are an important source of DAMPs. Interestingly, upon injury, both mtDNA and N-formylated peptides can act as DAMPs. This is due to the fact that mitochondria and bacteria display some similarities in that both possess circular DNA, N-formylated proteins and are double-membrane structures—evidence used in support of the endosymbiotic theory. mtDNA is similar to bacterial DNA in that it contains CpG motifs, which activate the TLR9 [289, 290]. Moreover, mitochondrial protein synthesis is initiated with the residue N-formyl methionine, similar to bacterial protein synthesis [291]. The resulting bacterial N-formylated peptides are known to act as PAMPs by binding and activating G protein-coupled formyl peptide receptors (FPRs) [292], while the mitochondrial N-formylated peptides act as DAMPs through activation of the formyl peptide receptor 1 [290]. Therefore, upon injury, release of these mitochondrial DAMPs activates the innate immune system, much like bacterial PAMPs, to promote sterile inflammatory responses [290].

Several studies have now described a crucial role for mitochondria in the regulation and activation of the inflammasome, specifically the NLRP3 inflammasome [293]. The inflammasomes are intracellular molecular platforms activated upon cellular infection or sterile stressors, which activate the proinflammatory cytokines, interleukin-1β (IL-1β) and IL-18, to trigger pyroptotic cell death (reviewed in [294, 295]). A variety of insults, resulting from cellular infection or stress, can promote mitochondrial dysfunction and activate the NLRP3 inflammasome [293]; however, the molecular mechanisms underlying the contribution of mitochondria to the activation of the NLRP3 inflammasome have only recently been described. While initial studies showed that mitochondrial dysfunction and mtROS production are required for NLRP3 inflammasome activation [296, 297], further evidence has shown that mtDNA translocation to the cytosol plays an active role in this process [297, 298], where it can directly bind to and activate the NRLP3 inflammasome [298]. In addition, the mitochondrial lipid cardiolipin—a phospholipid located exclusively in mitochondrial inner and bacterial membranes, regarded as evidence for symbiogenesis [299, 300]—is also required for NLRP3 inflammasome activation, by directly binding to NLRP3, downstream of mitochondrial dysfunction [301]. Altogether, mitochondria and mitochondrial DAMPs (such as mtDNA and cardiolipin) play a critical role in NLRP3 inflammasome activation and regulation. Moreover, by sensing mitochondrial DAMPs, the NLRP3 inflammasome plays a critical role in integrating mitochondrial dysfunction in a proinflammatory signaling response, thus explaining the association of mitochondrial damage with inflammatory diseases.

Despite the great number of studies describing mitochondria as a source of DAMPs during inflammation in the periphery, the potential for mitochondrial DAMPs to trigger, or exacerbate, inflammation in the brain is now being explored. In recent studies, this potential was tested by treating different brain cell types with mitochondrial components and measuring markers of inflammation afterwards. Neuronal and microglial cell lines exposed to mitochondrial lysates displayed increased markers of inflammation, with mtDNA being identified as the candidate DAMP responsible for the inflammatory changes [95]. While SH-SY5Y neuronal cells treated with mitochondrial lysates showed increased TNFα mRNA, decreased IκBα protein and increased NF-κB protein, microglial cells treated with mitochondrial lysates showed increased TNFα mRNA, increased IL-8 mRNA and redistribution of NF-κB to the nucleus [95]. In a different study, extracellular recombinant Tfam treatment of different models of human microglia, in combination with IFN-ϒ, was shown to induce secretions that were toxic to SH-SY5Y neuronal cells [302]. Recombinant Tfam treatment induced the expression of proinflammatory cytokines, such as IL-1β, IL-6 and IL-18, supporting the hypothesis that Tfam may also act as a proinflammatory intercellular signaling molecule recognized by brain microglia [302]. Moreover, mice injected with isolated mitochondria into the brain also revealed increased markers of inflammation such as increased Tnfα, increased NF-κB phosphorylation, increased GFAP protein and decreased Trem2 mRNA [94]. Despite these novel findings describing a role for extracellular mitochondrial DAMPs as proinflammatory signaling molecules in the brain, little is known about the mechanisms by which mitochondria act as a transcellular signaling platforms in the CNS. Recent research revealed that neurons and astrocytes can exchange mitochondria as a potential mode of cell-to-cell signaling [303, 304]. Whilean initial study showed that retinal ganglion cell axons can transfer mitochondria to adjacent astrocytes for degradation [303], mitochondria can also be transferred from astrocytes to adjacent neurons during ischemia to amplify cell survival signals [304], thus representing a neuroprotective strategy or a more efficient way to dispose/recycle mitochondria. However, during neurodegeneration, increased disposal of damaged mitochondria by compromised neurons (e.g. due to compromised mitochondrial quality control mechanisms) or its inefficient uptake by the recipient astrocytes (e.g. due to the presence of extracellular protein aggregates) might result in extracellular accumulation of mitochondrial DAMPs and, as a result, exacerbating neuroinflammation. Further research is necessary to test this hypothesis and identify the PRRs in the brain that are responsible for recognizing extracellular mitochondrial DAMPs; nevertheless, these studies suggest that mitochondria play an active role in neuroglial crosstalk during cellular homeostasis and stress.

Advertisement

5. Concluding comments

Although the innate immune system has specialized in the recognition of molecular patterns foreign to the host cells, cellular injury or stress may result in the release of endogenous molecular patterns, which trigger sterile inflammatory responses. Given its bacterial origin, mitochondria display some similarities with bacteria and represent an important source of DAMPs (including lipids, nucleic acids and proteins) with immunostimulatory potential. While under healthy conditions these DAMPs are sequestered within mitochondria, pathological insults resulting in mitochondrial and cellular damage promote the release of these danger signals to cause inflammation mediated by the innate immune system. Recent studies have shown that mitochondrial DAMPs have the potential to mediate inflammatory signaling in the brain; therefore, its contribution to the neuroinflammatory process in neurodegenerative disorders characterized by impaired mitochondrial function represents an emerging and promising field of research (Figure 1).

Figure 1.

Mitochondria are primary targets of cellular peptides, such as Aβ, tau and SNCA, overproduced during AD and PD pathogenesis. Damaged mitochondria are a source of DAMPs that activate the NLRP3 inflammasome and TLRs leading to the intraneuronal production of cytokines. These proinflammatory cytokines are released and activate innate immune response through microglia and astrocytes. This chronic inflammation impacts neurons exacerbating peptides formation and mitochondrial damage.

Further understanding of neuronal innate immunity-induced chronic mild neuroinflammation and its impact on age-related neurodegenerative disorders should focus on new studies addressing not only mitochondrial dysfunction and protein oligomerization but also mild inflammation, nutritional states, among others. The development of new biomarkers focusing on the inflammatory process and the identification of protective inflammatory processes should be pursuit. Additionally, exploiting the effect of mutations, epigenetic and the microbiome on immune-related modifications affecting the AD and PD phenotypes will be of paramount relevance to understand etiology of both diseases.

Advertisement

Acknowledgments

Work in our laboratories is supported by Fundação para a Ciência e a Tecnologia (FCT) and by EU-FEDER funding through the Operational Competitiveness Programme—COMPETE grant UID/NEU/04539/2013 and by Prémio Santa Casa Neurociências Mantero Belard MB-40-2016.

References

  1. 1. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E. Mitochondrial fragmentation in neurodegeneration. Nature Reviews Neuroscience. 2008;9:505-518. DOI: 10.1038/nrn2417
  2. 2. Lobet E, Letesson JJ, Arnould T. Mitochondria: A target for bacteria. Biochemical Pharmacology. 2015;94:173-185. DOI: 10.1016/j.bcp.2015.02.007
  3. 3. Hawking ZL. Alzheimer’s disease: The role of mitochondrial dysfunction and potential new therapies. Bioscience Horizons: The International Journal of Student Research. 2016;9:1-11
  4. 4. Lionaki E, Markaki M, Palikaras K, Tavernarakis N. Mitochondria, autophagy and age-associated neurodegenerative diseases: New insights into a complex interplay. Biochimica et Biophysica Acta. 2015;1847:1412-1423. DOI: 10.1016/j.bbabio.2015.04.010
  5. 5. Silva DF, Esteves AR, Oliveira CR, Cardoso SM. Mitochondria: The common upstream driver of amyloid-beta and tau pathology in Alzheimer's disease. Current Alzheimer Research. 2011;8:563-572
  6. 6. Arduino DM, Esteves AR, Oliveira CR, Cardoso SM. Mitochondrial metabolism modulation: A new therapeutic approach for Parkinson's disease. CNS & Neurological Disorders Drug Targets. 2010;9:105-119
  7. 7. Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, Tyurin VA, Yanamala N, Shrivastava IH, Mohammadyani D, Wang KZQ, Zhu J, Klein-Seetharaman J, Balasubramanian K, Amoscato AA, Borisenko G, Huang Z, Gusdon AM, Cheikhi A, Steer EK, Wang R, Baty C, Watkins S, Bahar I, Bayir H, Kagan VE. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nature Cell Biology. 2013;15:1197-1205. DOI: 10.1038/ncb2837
  8. 8. He Y, Hara H, Nunez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends in Biochemical Sciences. 2016;41:1012-1021. DOI: 10.1016/j.tibs.2016.09.002
  9. 9. Lampron A, Elali A, Rivest S. Innate immunity in the CNS: Redefining the relationship between the CNS and its environment. Neuron. 2013;78:214-232. DOI: 10.1016/j.neuron.2013.04.005
  10. 10. Liu HY, Chen CY, Hsueh YP. Innate immune responses regulate morphogenesis and degeneration: Roles of toll-like receptors and Sarm1 in neurons. Neuroscience Bulletin. 2014;30:645-654. DOI: 10.1007/s12264-014-1445-5
  11. 11. Vinters HV. Emerging concepts in Alzheimer's disease. Annual Review of Pathology: Mechanisms of Disease. 2015;10:291-319. DOI: 10.1146/annurev-pathol-020712-163927
  12. 12. Candeias E, Duarte AI, Sebastião I, Fernandes MA, Plácido AI, Carvalho C, Correia S, Santos RX, Seiça R, Santos MS, Oliveira CR, Moreira PI. Middle-aged diabetic females and males present distinct susceptibility to Alzheimer disease-like pathology. Molecular Neurobiology. 2016;54(8):6471-6489. DOI: 10.1007/s12035-016-0155-1
  13. 13. Huang Y, Mucke L. Alzheimer mechanisms and therapeutic strategies. Cell. 2012;148:1204-1222. DOI: 10.1016/j.cell.2012.02.040
  14. 14. Tarawneh R, Holtzman DM, Tarawneh R, Holtzman DM. The clinical problem of symptomatic Alzheimer disease and mild cognitive impairment. Cold Spring Harbor Perspectives in Medicine. 2012;2:a006148-a006148. DOI: 10.1101/cshperspect.a006148
  15. 15. Jack CR, Holtzman DM. Biomarker modeling of alzheimer's disease. Neuron. 2013;80:1347-1358. DOI: 10.1016/j.neuron.2013.12.003
  16. 16. Irvine GB, El-Agnaf OM, Shankar GM, Walsh DM. Protein aggregation in the brain: The molecular basis for Alzheimer’s and Parkinson’s diseases. Molecular Medicine. 2008;14(7-8):451-64. DOI: 10.2119/2007-00100
  17. 17. Gowrishankar S, Yuan P, Wu Y, Schrag M, Paradise S, Grutzendler J, De Camilli P, Ferguson SM. Massive accumulation of luminal protease-deficient axonal lysosomes at Alzheimer's disease amyloid plaques. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E3699-E3708. DOI: 10.1073/pnas.1510329112
  18. 18. Reddy PH, Beal MF. Amyloid beta, mitochondrial dysfunction and synaptic damage: Implications for cognitive decline in aging and Alzheimer's disease. Trends in Molecular Medicine. 2008;14:45-53. DOI: 10.1016/j.molmed.2007.12.002
  19. 19. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787-795. DOI: 10.1038/nature05292
  20. 20. Walters A, Phillips E, Zheng R. Evidence for neuroinflammation in Alzheimer’s disease. Progress in Neurology and Psychiatry. 2016;20:25-31. DOI: 10.1002/pnp.444
  21. 21. Morales I, Guzmán-Martínez L, Cerda-Troncoso C, Farías GA, Maccioni RB. Neuroinflammation in the pathogenesis of Alzheimer's disease. A rational framework for the search of novel therapeutic approaches. Frontiers in Cellular Neuroscience. 2014;8:1-9. DOI: 10.3389/fncel.2014.00112
  22. 22. Minter MR, Taylor JM, Crack PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer's disease. Journal of Neurochemistry. 2016;136:457-474. DOI: 10.1111/jnc.13411
  23. 23. Hamelin L, Lagarde J, Dorothée G, Leroy C, Labit M, Comley RA, De Souza LC, Corne H, Dauphinot L, Bertoux M, Dubois B, Gervais P, Colliot O, Potier MC, Bottlaender M, Sarazin M. Early and protective microglial activation in Alzheimer's disease: A prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139:1252-1264. DOI: 10.1093/brain/aww017
  24. 24. Bronzuoli MR, Steardo L, Scuderi C. Targeting neuroinflammation in Alzheimer’s disease. Journal of Inflammation Research. 2016:199-208. DOI: 10.2147/JIR.S86958
  25. 25. Swerdlow RH, Koppel S, Weidling I, Hayley C, Ji Y, Wilkins HM. Mitochondria, Cybrids, aging, and Alzheimer's disease. Progress in Molecular Biology and Translational Science. 2017;146:259-302. DOI: 10.1016/bs.pmbts.2016.12.017
  26. 26. Mutisya EM, Bowling AC, Beal MF. Cortical cytochrome oxidase activity is reduced in Alzheimer's disease. Journal of Neurochemistry. 1994;63:2179-2184
  27. 27. Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI, Solaini G. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer's disease. Neurobiology of Aging. 2002;23:371-376
  28. 28. Cardoso SM, Proenca MT, Santos S, Santana I, Oliveira CR. Cytochrome c oxidase is decreased in Alzheimer's disease platelets. Neurobiology of Aging. 2004;25:105-110
  29. 29. Parker WD Jr, Mahr NJ, Filley CM, Parks JK, Hughes D, Young DA, Cullum CM. Reduced platelet cytochrome c oxidase activity in Alzheimer's disease. Neurology. 1994;44:1086-1090
  30. 30. Molina JA, deBustos F, Jimenez-Jimenez FJ, Benito-Leon J, Gasalla T, Orti-Pareja M, Vela L, Bermejo F, Martin MA, Campos Y, Arenas J. Respiratory chain enzyme activities in isolated mitochondria of lymphocytes from patients with Alzheimer's disease. Neurology. 1997;48:636-638
  31. 31. Fukuyama H, Ogawa M, Yamauchi H, Yamaguchi S, Kimura J, Yonekura Y, Konishi J. Altered cerebral energy metabolism in Alzheimer's disease: A PET study. Journal of Nuclear Medicine. 1994;35:1-6
  32. 32. Maynard S, Hejl AM, Dinh TS, Keijzers G, Hansen AM, Desler C, Moreno-Villanueva M, Burkle A, Rasmussen LJ, Waldemar G, Bohr VA. Defective mitochondrial respiration, altered dNTP pools and reduced AP endonuclease 1 activity in peripheral blood mononuclear cells of Alzheimer's disease patients. Aging (Albany NY). 2015;7:793-815. DOI: 10.18632/aging.100810
  33. 33. Silva DF, Selfridge JE, Lu JEL, Roy N, Hutfles L, burns JM, Michaelis EK, Yan S, Cardoso SM, Swerdlow RH. Bioenergetic flux, mitochondrial mass and mitochondrial morphology dynamics in AD and MCI cybrid cell lines. Human Molecular Genetics. 2013;22:3931-3946. DOI: 10.1093/hmg/ddt247
  34. 34. King MP, Attardi G. Human cells lacking mtDNA: Repopulation with exogenous mitochondria by complementation. Science. 1989;246:500-503
  35. 35. Ghosh SS, Swerdlow RH, Miller SW, Sheeman B, Parker WD Jr, Davis RE. Use of cytoplasmic hybrid cell lines for elucidating the role of mitochondrial dysfunction in Alzheimer's disease and Parkinson's disease. Annals of the New York Academy of Sciences. 1999;893:176-191
  36. 36. Silva DF, Santana I, Esteves AR, Baldeiras I, Arduino DM, Oliveira CR, Cardoso SM. Prodromal metabolic phenotype in MCI cybrids: Implications for Alzheimer's disease. Current Alzheimer Research. 2013;10:180-190
  37. 37. Caspersen C, Wang N, Yao J, Sosunov A, Chen X, Lustbader JW, Xu HW, Stern D, McKhann G, Yan SD. Mitochondrial Abeta: A potential focal point for neuronal metabolic dysfunction in Alzheimer's disease. The FASEB Journal. 2005;19:2040-2041. DOI: 10.1096/fj.05-3735fje
  38. 38. Schuh RA, Jackson KC, Schlappal AE, Spangenburg EE, Ward CW, Park JH, Dugger N, Shi GL, Fishman PS. Mitochondrial oxygen consumption deficits in skeletal muscle isolated from an Alzheimer's disease-relevant murine model. BMC Neuroscience. 2014;15:24. DOI: 10.1186/1471-2202-15-24
  39. 39. Chowdhury SR, Djordjevic J, Albensi BC, Fernyhough P. Simultaneous evaluation of substrate-dependent oxygen consumption rates and mitochondrial membrane potential by TMRM and safranin in cortical mitochondria. Bioscience Reports. 2015;36:e00286. DOI: 10.1042/BSR20150244
  40. 40. Onyango IG, Dennis J, Khan SM. Mitochondrial dysfunction in Alzheimer's disease and the rationale for bioenergetics based therapies. Aging & Disease. 2016;7:201-214. DOI: 10.14336/AD.2015.1007
  41. 41. Suski JM, Lebiedzinska M, Bonora M, Pinton P, Duszynski J, Wieckowski MR. Relation between mitochondrial membrane potential and ROS formation. Methods in Molecular Biology. 2012;810:183-205. DOI: 10.1007/978-1-61779-382-0_12
  42. 42. Lebiedzinska M, Karkucinska-Wieckowska A, Giorgi C, Karczmarewicz E, Pronicka E, Pinton P, Duszynski J, Pronicki M, Wieckowski MR. Oxidative stress-dependent p66Shc phosphorylation in skin fibroblasts of children with mitochondrial disorders. Biochimica et Biophysica Acta. 2010;1797:952-960. DOI: 10.1016/j.bbabio.2010.03.005
  43. 43. McLennan HR, Degli Esposti M. The contribution of mitochondrial respiratory complexes to the production of reactive oxygen species. Journal of Bioenergetics and Biomembranes. 2000;32:153-162
  44. 44. Pickrell AM, Fukui H, Moraes CT. The role of cytochrome c oxidase deficiency in ROS and amyloid plaque formation. Journal of Bioenergetics and Biomembranes. 2009;41:453-456. DOI: 10.1007/s10863-009-9245-3
  45. 45. Paradies G, Petrosillo G, Pistolese M, Ruggiero FM. The effect of reactive oxygen species generated from the mitochondrial electron transport chain on the cytochrome c oxidase activity and on the cardiolipin content in bovine heart submitochondrial particles. FEBS Letters. 2000;466:323-326
  46. 46. Lyras L, Cairns NJ, Jenner A, Jenner P, Halliwell B. An assessment of oxidative damage to proteins, lipids, and DNA in brain from patients with Alzheimer's disease. Journal of Neurochemistry. 1997;68:2061-2069
  47. 47. Han BH, Zhou ML, Johnson AW, Singh I, Liao F, Vellimana AK, Nelson JW, Milner E, Cirrito JR, Basak J, Yoo M, Dietrich HH, Holtzman DM, Zipfel GJ. Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and microhemorrhage in aged Tg2576 mice. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E881-E890. DOI: 10.1073/pnas.1414930112
  48. 48. Leutner S, Schindowski K, Frolich L, Maurer K, Kratzsch T, Eckert A, Muller WE. Enhanced ROS-generation in lymphocytes from Alzheimer's patients. Pharmacopsychiatry. 2005;38:312-315. DOI: 10.1055/s-2005-916186
  49. 49. Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, Wang N, Caspersen C, Chen X, Pollak S, Chaney M, Trinchese F, Liu S, Gunn-Moore F, Lue LF, Walker DG, Kuppusamy P, Zewier ZL, Arancio O, Stern D, Yan SS, Wu H. ABAD directly links Abeta to mitochondrial toxicity in Alzheimer's disease. Science. 2004;304:448-452. DOI: 10.1126/science.1091230
  50. 50. Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH. Mitochondria are a direct site of a beta accumulation in Alzheimer's disease neurons: Implications for free radical generation and oxidative damage in disease progression. Human Molecular Genetics. 2006;15:1437-1449. DOI: 10.1093/hmg/ddl066
  51. 51. Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, Johnson AB, Kress Y, Vinters HV, Tabaton M, Shimohama S, Cash AD, Siedlak SL, Harris PL, Jones PK, Petersen RB, Perry G, Smith MA. Mitochondrial abnormalities in Alzheimer's disease. The Journal of Neuroscience. 2001;21:3017-3023
  52. 52. Trimmer PA, Swerdlow RH, Parks JK, Keeney P, Bennett Jr JP, Miller SW, Davis RE, Parker WD Jr. Abnormal mitochondrial morphology in sporadic Parkinson's and Alzheimer's disease cybrid cell lines. Experimental Neurology. 2000;162:37-50. DOI: 10.1006/exnr.2000.7333
  53. 53. Silva DF, Esteves AR, Oliveira CR, Cardoso SM. Mitochondrial metabolism power SIRT2-dependent deficient traffic causing Alzheimer's-disease related pathology. Molecular Neurobiology. 2016. DOI: 10.1007/s12035-016-9951-x
  54. 54. Smirnova E, Griparic L, Shurland DL, van der Bliek AM. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Molecular Biology of the Cell. 2001;12:2245-2256
  55. 55. SilvaDF, SelfridgeJE, Lu JEL, Cardoso SM, Swerdlow RH. Mitochondrial abnormalities in Alzheimer's disease: Possible targets for therapeutic intervention. Advances in Pharmacology.2012;64:83-126. DOI: 10.1016/B978-0-12-394816-8.00003-9
  56. 56. Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, Lee JH, Mohan PS, Mercken M, Farmery MR, Tjernberg LO, Jiang Y, Duff K, Uchiyama Y, Naslund J, Mathews PM, Cataldo AM, Nixon RA. Macroautophagy--a novel Beta-amyloid peptide-generating pathway activated in Alzheimer's disease. The Journal of Cell Biology. 2005;171:87-98. DOI: 10.1083/jcb.200505082
  57. 57. Cardoso SM, Pereira CF, Moreira PI, Arduino DM, Esteves AR, Oliveira CR. Mitochondrial control of autophagic lysosomal pathway in Alzheimer's disease. Experimental Neurology. 2010;223:294-298. DOI: 10.1016/j.expneurol.2009.06.008
  58. 58. Cai Z, Zhou Y, Liu Z, Ke Z, Zhao B. Autophagy dysfunction upregulates beta-amyloid peptides via enhancing the activity of gamma-secretase complex. Neuropsychiatric Disease and Treatment. 2015;11:2091-2099. DOI: 10.2147/NDT.S84755
  59. 59. Nilsson P, Saido TC. Dual roles for autophagy: Degradation and secretion of Alzheimer's disease Abeta peptide. BioEssays. 2014;36:570-578. DOI: 10.1002/bies.201400002
  60. 60. Nilsson P, Loganathan K, Sekiguchi M, Matsuba Y, Hui K, Tsubuki S, Tanaka M, Iwata N, Saito T, Saido TC. Abeta secretion and plaque formation depend on autophagy. Cell Reports. 2013;5:61-69. DOI: 10.1016/j.celrep.2013.08.042
  61. 61. Nixon RA, Wegiel J, Kumar A, WH Y, Peterhoff C, Cataldo A, Cuervo AM. Extensive involvement of autophagy in Alzheimer disease: An immuno-electron microscopy study. Journal of Neuropathology and Experimental Neurology. 2005;64:113-122
  62. 62. Marks N, Berg MJ. BACE and gamma-secretase characterization and their sorting as therapeutic targets to reduce amyloidogenesis. Neurochemical Research. 2010;35:181-210. DOI: 10.1007/s11064-009-0054-1
  63. 63. Yu WH, Kumar A, Peterhoff C, Shapiro Kulnane L, Uchiyama Y, Lamb BT, Cuervo AM, Nixon RA. Autophagic vacuoles are enriched in amyloid precursor protein-secretase activities: Implications for beta-amyloid peptide over-production and localization in Alzheimer's disease. The International Journal of Biochemistry & Cell Biology. 2004;36:2531-2540. DOI: 10.1016/j.biocel.2004.05.010
  64. 64. Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. Journal of Cell Science. 2007;120:4081-4091. DOI: 10.1242/jcs.019265
  65. 65. Chen X, Kondo K, Motoki K, Homma H, Okazawa H. Fasting activates macroautophagy in neurons of Alzheimer's disease mouse model but is insufficient to degrade amyloid-beta. Scientific Reports. 2015;5:12115. DOI: 10.1038/srep12115
  66. 66. Kosik KS, Joachim CL, Selkoe DJ. Microtubule-associated protein tau (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America. 1986;83:4044-4048
  67. 67. Ma RH, Zhang Y, Hong XY, Zhang JF, Wang JZ, Liu GP. Role of microtubule-associated protein tau phosphorylation in Alzheimer's disease. Journal of Huazhong University of Science and Technology. Medical Sciences. 2017;37:307-312. DOI: 10.1007/s11596-017-1732-x
  68. 68. Takemura R, Okabe S, Umeyama T, Kanai Y, Cowan NJ, Hirokawa N. Increased microtubule stability and alpha tubulin acetylation in cells transfected with microtubule-associated proteins MAP1B, MAP2 or tau. Journal of Cell Science. 1992;103(Pt 4):953-964
  69. 69. Michaelis ML, Dobrowsky RT, Li G. Tau neurofibrillary pathology and microtubule stability. Journal of Molecular Neuroscience. 2002;19:289-293. DOI: 10.1385/JMN:19:3:289
  70. 70. Cash AD, Aliev G, Siedlak SL, Nunomura A, Fujioka H, Zhu X, Raina AK, Vinters HV, Tabaton M, Johnson AB, Paula-Barbosa M, Avila J, Jones PK, Castellani RJ, Smith MA, Perry G. Microtubule reduction in Alzheimer's disease and aging is independent of tau filament formation. The American Journal of Pathology. 2003;162:1623-1627
  71. 71. Silva DF, Esteves AR, Arduino DM, Oliveira CR, Cardoso SM. Amyloid-beta-induced mitochondrial dysfunction impairs the autophagic lysosomal pathway in a tubulin dependent pathway. Journal of Alzheimer's Disease. 2011;26:565-581. DOI: 10.3233/JAD-2011-110423
  72. 72. Matsuoka Y, Jouroukhin Y, Gray AJ, Ma L, Hirata-Fukae C, Li HF, Feng L, Lecanu L, Walker BR, Planel E, Arancio O, Gozes I, Aisen PS. A neuronal microtubule-interacting agent, NAPVSIPQ, reduces tau pathology and enhances cognitive function in a mouse model of Alzheimer's disease. The Journal of Pharmacology and Experimental Therapeutics. 2008;325:146-153. DOI: 10.1124/jpet.107.130526
  73. 73. Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer's 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde“. Clinical Anatomy. 1995;8:429-431. DOI: 10.1002/ca.980080612
  74. 74. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Molecular Medicine. 2016;8:595-608. DOI: 10.15252/emmm.201606210
  75. 75. Morris GP, Clark IA, Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer's disease. Acta Neuropathologica Communications. 2014;2:135. DOI: 10.1186/s40478-014-0135-5
  76. 76. Jack CR Jr, Knopman DS, Jagust WJ, Petersen RC, Weiner MW, Aisen PS, Shaw LM, Vemuri P, Wiste HJ, Weigand SD, Lesnick TG, Pankratz VS, Donohue MC, Trojanowski JQ. Tracking pathophysiological processes in Alzheimer's disease: An updated hypothetical model of dynamic biomarkers. Lancet Neurology. 2013;12:207-216. DOI: 10.1016/S1474-4422(12)70291-0
  77. 77. McGeer PL, McGeer EG. The amyloid cascade-inflammatory hypothesis of Alzheimer disease: Implications for therapy. Acta Neuropathologica. 2013;126:479-497. DOI: 10.1007/s00401-013-1177-7
  78. 78. Stewart CR, Stuart LM, Wilkinson K, vanGils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, ElKhoury J, Golenbock DT, Moore KJ. CD36 ligands promote sterile inflammation through assembly of a toll-like receptor 4 and 6 heterodimer. Nature Immunology. 2010;11:155-161. DOI: 10.1038/ni.1836
  79. 79. Koenigsknecht J, Landreth G. Microglial phagocytosis of fibrillar beta-amyloid through a beta1 integrin-dependent mechanism. The Journal of Neuroscience. 2004;24:9838-9846. DOI: 10.1523/JNEUROSCI.2557-04.2004
  80. 80. Bamberger ME, Harris ME, McDonald DR, Husemann J, Landreth GEA. Cell surface receptor complex for fibrillar beta-amyloid mediates microglial activation. The Journal of Neuroscience. 2003;23:2665-2674
  81. 81. Heppner FL, Ransohoff RM, Becher B. Immune attack: The role of inflammation in Alzheimer disease. Nature Reviews. Neuroscience. 2015;16:358-372. DOI: 10.1038/nrn3880
  82. 82. Prokop S, Miller KR, Heppner FL. Microglia actions in Alzheimer's disease. Acta Neuropathologica. 2013;126:461-477
  83. 83. Francois A, Rioux Bilan A, Quellard N, Fernandez B, Janet T, Chassaing D, Paccalin M, Terro F, Page G. Longitudinal follow-up of autophagy and inflammation in brain of APPswePS1dE9 transgenic mice. Journal of Neuroinflammation. 2014;11:139. DOI: 10.1186/s12974-014-0139-x
  84. 84. Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer's disease. Journal of Neuroinflammation. 2005;2:9. DOI: 10.1186/1742-2094-2-9
  85. 85. Xia MQ, Qin SX, LJ W, Mackay CR, Hyman BT. Immunohistochemical study of the beta-chemokine receptors CCR3 and CCR5 and their ligands in normal and Alzheimer's disease brains. The American Journal of Pathology. 1998;153:31-37
  86. 86. Ishizuka K, Kimura T, Igata-yi R, Katsuragi S, Takamatsu J, Miyakawa T. Identification of monocyte chemoattractant protein-1 in senile plaques and reactive microglia of Alzheimer's disease. Psychiatry and Clinical Neurosciences. 1997;51:135-138
  87. 87. Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, Yarasheski KE, Bateman RJ. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science. 2010;330:1774. DOI: 10.1126/science.1197623
  88. 88. Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, Hooli B, Choi SH, Hyman BT, Tanzi RE. Alzheimer's disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78:631-643. DOI: 10.1016/j.neuron.2013.04.014
  89. 89. Lee CY, Landreth GE. The role of microglia in amyloid clearance from the AD brain. Journal of Neural Transmission (Vienna). 2010;117:949-960. DOI: 10.1007/s00702-010-0433-4
  90. 90. Monteleone M, Stow JL, Schroder K. Mechanisms of unconventional secretion of IL-1 family cytokines. Cytokine. 2015;74:213-218. DOI: 10.1016/j.cyto.2015.03.022
  91. 91. Winkler S, Rosen-Wolff A. Caspase-1: An integral regulator of innate immunity. Seminars in Immunopathology. 2015;37:419-427. DOI: 10.1007/s00281-015-0494-4
  92. 92. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674-678. DOI: 10.1038/nature11729
  93. 93. Saresella M, La Rosa F, Piancone F, Zoppis M, Marventano I, Calabrese E, Rainone V, Nemni R, Mancuso R, Clerici M. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer's disease. Molecular Neurodegeneration. 2016;11:23. DOI: 10.1186/s13024-016-0088-1
  94. 94. Wilkins HM, Koppel SJ, Weidling IW, Roy N, Ryan LN, Stanford JA, Swerdlow RH. Extracellular mitochondria and mitochondrial components act as damage-associated molecular pattern molecules in the mouse brain. Journal of Neuroimmune Pharmacology. 2016;11:622-628. DOI: 10.1007/s11481-016-9704-7
  95. 95. Wilkins HM, Carl SM, Weber SG, Ramanujan SA, Festoff BW, Linseman DA, Swerdlow RH. Mitochondrial lysates induce inflammation and Alzheimer's disease-relevant changes in microglial and neuronal cells. Journal of Alzheimer's Disease. 2015;45:305-318. DOI: 10.3233/JAD-142334
  96. 96. Sofroniew MV, Vinters HV. Astrocytes: Biology and pathology. Acta Neuropathologica. 2010;119:7-35. DOI: 10.1007/s00401-009-0619-8
  97. 97. Hennessy E, Griffin EW, Cunningham C. Astrocytes are primed by chronic Neurodegeneration to produce exaggerated chemokine and cell infiltration responses to acute stimulation with the cytokines IL-1beta and TNF-alpha. The Journal of Neuroscience. 2015;35:8411-8422. DOI: 10.1523/JNEUROSCI.2745-14.2015
  98. 98. Carrero I, Gonzalo MR, Martin B, Sanz-Anquela JM, Arevalo-Serrano J, Gonzalo-Ruiz A. Oligomers of beta-amyloid protein (Abeta1-42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain. Experimental Neurology. 2012;236:215-227. DOI: 10.1016/j.expneurol.2012.05.004
  99. 99. Smits HA, Rijsmus A, vanLoon JH, Wat JW, Verhoef J, Boven LA, Nottet HS. Amyloid-beta-induced chemokine production in primary human macrophages and astrocytes. Journal of Neuroimmunology. 2002;127:160-168
  100. 100. Nielsen HM, Mulder SD, Belien JA, Musters RJ, Eikelenboom P, Veerhuis R. Astrocytic a beta 1-42 uptake is determined by a beta-aggregation state and the presence of amyloid-associated proteins. Glia. 2010;58:1235-1246. DOI: 10.1002/glia.21004
  101. 101. Edwards MM, Robinson SR. TNF alpha affects the expression of GFAP and S100B: Implications for Alzheimer's disease. Journal of Neural Transmission (Vienna). 2006;113:1709-1715. DOI: 10.1007/s00702-006-0479-5
  102. 102. Simpson JE, Ince PG. Lace G, Forster G, Shaw PJ, Matthews F, Savva G, Brayne C, Wharton SB, function MRCC, and ageing neuropathology study G. Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain. Neurobiology of Aging. 2010;31:578-590. DOI: 10.1016/j.neurobiolaging.2008.05.015
  103. 103. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP. Neuroinflammation in Alzheimer's disease. Lancet Neurology. 2015;14:388-405. DOI: 10.1016/S1474-4422(15)70016-5
  104. 104. Thiabaud G, Pizzocaro S, Garcia-Serres R, Latour JM, Monzani E, Casella L. Heme binding induces dimerization and nitration of truncated beta-amyloid peptide Abeta16 under oxidative stress. Angewandte Chemie (International Ed. in English). 2013;52:8041-8044. DOI: 10.1002/anie.201302989
  105. 105. Butterfield DA, Reed TT, Perluigi M, De Marco C, Coccia R, Keller JN, Markesbery WR, Sultana R. Elevated levels of 3-nitrotyrosine in brain from subjects with amnestic mild cognitive impairment: Implications for the role of nitration in the progression of Alzheimer's disease. Brain Research. 2007;1148:243-248. DOI: 10.1016/j.brainres.2007.02.084
  106. 106. Wyss-Coray T, Rogers J. Inflammation in Alzheimer disease-a brief review of the basic science and clinical literature. Cold Spring Harbor Perspectives in Medicine. 2012;2:a006346. DOI: 10.1101/cshperspect.a006346
  107. 107. Thambisetty M, An Y, Nalls M, Sojkova J, Swaminathan S, Zhou Y, Singleton AB, Wong DF, Ferrucci L, Saykin AJ, Resnick SM. Baltimore longitudinal study of A, and the Alzheimer's disease neuroimaging I. Effect of complement CR1 on brain amyloid burden during aging and its modification by APOE genotype. Biological Psychiatry. 2013;73:422-428. DOI: 10.1016/j.biopsych.2012.08.015
  108. 108. Hawkes CA, McLaurin J. Selective targeting of perivascular macrophages for clearance of beta-amyloid in cerebral amyloid angiopathy. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:1261-1266. DOI: 10.1073/pnas.0805453106
  109. 109. Sagare AP, Bell RD, Zlokovic BV. Neurovascular dysfunction and faulty amyloid beta-peptide clearance in Alzheimer disease. Cold Spring Harbor Perspectives in Medicine. 2012;2:a011452. DOI: 10.1101/cshperspect.a011452
  110. 110. Vukic V, Callaghan D, Walker D, Lue LF, Liu QY, Couraud PO, Romero IA, Weksler B, Stanimirovic DB, Zhang W. Expression of inflammatory genes induced by beta-amyloid peptides in human brain endothelial cells and in Alzheimer's brain is mediated by the JNK-AP1 signaling pathway. Neurobiology of Disease. 2009;34:95-106. DOI: 10.1016/j.nbd.2008.12.007
  111. 111. Yamada T, Akiyama H, McGeer PL. Complement-activated oligodendroglia: A new pathogenic entity identified by immunostaining with antibodies to human complement proteins C3d and C4d. Neuroscience Letters. 1990;112:161-166
  112. 112. Walker DG, Dalsing-Hernandez JE, Campbell NA, Lue LF. Decreased expression of CD200 and CD200 receptor in Alzheimer's disease: A potential mechanism leading to chronic inflammation. Experimental Neurology. 2009;215:5-19. DOI: 10.1016/j.expneurol.2008.09.003
  113. 113. Latta CH, Brothers HM, Wilcock DM. Neuroinflammation in Alzheimer's disease; a source of heterogeneity and target for personalized therapy. Neuroscience. 2015;302:103-111. DOI: 10.1016/j.neuroscience.2014.09.061
  114. 114. Cai Z, Hussain MD, Yan LJ. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer's disease. The International Journal of Neuroscience. 2014;124:307-321. DOI: 10.3109/00207454.2013.833510
  115. 115. Wyss-Coray T. Inflammation in Alzheimer disease: Driving force, bystander or beneficial response? Nature Medicine. 2006;12:1005-1015. DOI: 10.1038/nm1484
  116. 116. Jones L, Holmans PA, Hamshere ML, Harold D, Moskvina V, Ivanov D, Pocklington A, Abraham R, Hollingworth P, Sims R, Gerrish A, Pahwa JS, Jones N, Stretton A, Morgan AR, Lovestone S, Powell J, Proitsi P, Lupton MK, Brayne C, Rubinsztein DC, Gill M, Lawlor B, Lynch A, Morgan K, Brown KS, Passmore PA, Craig D, McGuinness B, Todd S, Holmes C, Mann D, Smith AD, Love S, Kehoe PG, Mead S, Fox N, Rossor M, Collinge J, Maier W, Jessen F, Schurmann B, Heun R, Kolsch H, van den Bussche H, Heuser I, Peters O, Kornhuber J, Wiltfang J, Dichgans M, Frolich L, Hampel H, Hull M, Rujescu D, Goate AM, Kauwe JS, Cruchaga C, Nowotny P, Morris JC, Mayo K, Livingston G, Bass NJ, Gurling H, McQuillin A, Gwilliam R, Deloukas P, Al-Chalabi A, Shaw CE, Singleton AB, Guerreiro R, Muhleisen TW, Nothen MM, Moebus S, Jockel KH, Klopp N, Wichmann HE, Ruther E, Carrasquillo MM, Pankratz VS, Younkin SG, Hardy J, O’Donovan MC, Owen MJ, Williams J. Genetic evidence implicates the immune system and cholesterol metabolism in the aetiology of Alzheimer's disease. PLoS One. 2010;5:e13950. DOI: 10.1371/journal.pone.0013950
  117. 117. Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A, Alzheimer Disease Neuroimaging Initiative, Morris MC, Evans DA, Johnson K, Sperling RA, Schneider JA, Bennett DA, De Jager PL. CD33 Alzheimer's disease locus: Altered monocyte function and amyloid biology. Nature Neuroscience. 2013;16:848-850. DOI:10.1038/nn.3435
  118. 118. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J, Alzheimer Genetic Analysis Group. TREM2 variants in Alzheimer's disease. The New England Journal of Medicine. 2013;368:117-127. DOI: 10.1056/NEJM-oa1211851
  119. 119. Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, Letenneur L, Bettens K, Berr C, Pasquier F, Fievet N, Barberger-Gateau P, Engelborghs S, De Deyn P, Mateo I, Franck A, Helisalmi S, Porcellini E, Hanon O, European Alzheimer's Disease Initiative I, de Pancorbo MM, Lendon C, Dufouil C, Jaillard C, Leveillard T, Alvarez V, Bosco P, Mancuso M, Panza F, Nacmias B, Bossu P, Piccardi P, Annoni G, Seripa D, Galimberti D, Hannequin D, Licastro F, Soininen H, Ritchie K, Blanche H, Dartigues JF, Tzourio C, Gut I, Van Broeckhoven C, Alperovitch A, Lathrop M, Amouyel P. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nature Genetics. 2009;41:1094-1099. DOI: 10.1038/ng.439
  120. 120. Brosseron F, Krauthausen M, Kummer M, Heneka MT. Body fluid cytokine levels in mild cognitive impairment and Alzheimer's disease: A comparative overview. Molecular Neurobiology. 2014;50:534-544. DOI: 10.1007/s12035-014-8657-1
  121. 121. Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer's disease. Journal of Neurology, Neurosurgery, and Psychiatry. 2003;74:1200-1205
  122. 122. Krstic D, Madhusudan A, Doehner J, Vogel P, Notter T, Imhof C, Manalastas A, Hilfiker M, Pfister S, Schwerdel C, Riether C, Meyer U, Knuesel I. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. Journal of Neuroinflammation. 2012;9:151. DOI: 10.1186/1742-2094-9-151
  123. 123. Group ADC, Bentham P, Gray R, Sellwood E, Hills R, Crome P, Raftery J. Aspirin in Alzheimer's disease (AD2000): A randomised open-label trial. Lancet Neurology. 2008;7:41-49. DOI: 10.1016/S1474-4422(07)70293-4
  124. 124. Szekely CA, Breitner JC, Fitzpatrick AL, Rea TD, Psaty BM, Kuller LH, Zandi PP. NSAID use and dementia risk in the cardiovascular health study: Role of APOE and NSAID type. Neurology. 2008;70:17-24. DOI: 10.1212/01.wnl.0000284596.95156.48
  125. 125. VanItallie TB. Alzheimer's disease: Innate immunity gone awry? Metabolism. 2017;69S:S41-S49. DOI: 10.1016/j.metabol.2017.01.014
  126. 126. Bhamra MS, Ashton NJ. Finding a pathological diagnosis for Alzheimer's disease: Are inflammatory molecules the answer? Electrophoresis. 2012;33:3598-3607. DOI: 10.1002/elps.201200161
  127. 127. Swardfager W, Lanctot K, Rothenburg L, Wong A, Cappell J, Herrmann N. A meta-analysis of cytokines in Alzheimer's disease. Biological Psychiatry. 2010;68:930-941. DOI: 10.1016/j.biopsych.2010.06.012
  128. 128. Petzold A, Jenkins R, Watt HC, Green AJ, Thompson EJ, Keir G, Fox NC, Rossor MN. Cerebrospinal fluid S100B correlates with brain atrophy in Alzheimer's disease. Neuroscience Letters. 2003;336:167-170
  129. 129. Licastro F, Pedrini S, Caputo L, Annoni G, Davis LJ, Ferri C, Casadei V, Grimaldi LM. Increased plasma levels of interleukin-1, interleukin-6 and alpha-1-antichymotrypsin in patients with Alzheimer's disease: Peripheral inflammation or signals from the brain? Journal of Neuroimmunology. 2000;103:97-102
  130. 130. Helmy AA, Naseer MM, Shafie SE, Nada MA. Role of interleukin 6 and alpha-globulins in differentiating Alzheimer and vascular dementias. Neurodegenerative Diseases. 2012;9:81-86. DOI: 10.1159/000329568
  131. 131. Thambisetty M, Hye A, Foy C, Daly E, Glover A, Cooper A, Simmons A, Murphy D, Lovestone S. Proteome-based identification of plasma proteins associated with hippocampal metabolism in early Alzheimer's disease. Journal of Neurology. 2008;255:1712-1720. DOI: 10.1007/s00415-008-0006-8
  132. 132. Schrijvers EM, Koudstaal PJ, Hofman A, Breteler MM. Plasma clusterin and the risk of Alzheimer disease. Journal of the American Medical Association. 2011;305:1322-1326. DOI: 10.1001/jama.2011.381
  133. 133. Yasuno F, Kosaka J, Ota M, Higuchi M, Ito H, Fujimura Y, Nozaki S, Takahashi S, Mizukami K, Asada T, Suhara T. Increased binding of peripheral benzodiazepine receptor in mild cognitive impairment-dementia converters measured by positron emission tomography with [(1)(1)C]DAA1106. Psychiatry Research. 2012;203:67-74. DOI: 10.1016/j.pscychresns.2011.08.013
  134. 134. Djordjevic J, Sabbir MG, Albensi BC. Traumatic brain injury as a risk factor for Alzheimer's disease: Is inflammatory Signaling a key player? Current Alzheimer Research. 2016;13:730-738
  135. 135. Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, Culliford D, Perry VH. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73:768-774. DOI: 10.1212/WNL.0b013e3181b6bb95
  136. 136. Verdile G, Keane KN, Cruzat VF, Medic S, Sabale M, Rowles J, Wijesekara N, Martins RN, Fraser PE, Newsholme P. Inflammation and oxidative stress: The molecular connectivity between insulin resistance, obesity, and Alzheimer's disease. Mediators of Inflammation. 2015;2015:105828. DOI: 10.1155/2015/105828
  137. 137. De Felice FG, Ferreira ST. Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes. 2014;63:2262-2272. DOI: 10.2337/db13-1954
  138. 138. Lotharius J, Brundin P. Pathogenesis of parkinson's disease: Dopamine, vesicles and α-synuclein. Nature Reviews Neuroscience. 2002;3:932-942. DOI: 10.1038/nrn983
  139. 139. Bose A, Beal MF. Mitochondrial dysfunction in Parkinson's disease. Journal of Neurochemistry. 2016;139:216-231. DOI: 10.1111/jnc.13731
  140. 140. Esteves AR, Arduíno DM, Swerdlow RH, Oliveira CR, Cardoso SM. Oxidative stress involvement in α−Synuclein Oligomerization in Parkinson's disease Cybrids. Antioxidants & Redox Signaling. 2009;11:439-448. DOI: 10.1089/ars.2008.2247
  141. 141. McNaught KS, Jenner P. Proteasomal function is impaired in substantia nigra in Parkinson's disease. Neuroscience Letters. 2001;297:191-194
  142. 142. Olanow CW. The pathogenesis of cell death in Parkinson's disease – 2007. Movement Disorders. 2007;22:S335-S342. DOI: 10.1002/mds.21675
  143. 143. Ryan BJ, Hoek S, Fon EA, Wade-martins R. Mitochondrial dysfunction and mitophagy in Parkinson ’ s : From familial to sporadic disease. Trends in Biochemical Sciences. 2015;40:200-210. DOI: 10.1016/j.tibs.2015.02.003
  144. 144. Hu Q, Wang G. Mitochondrial dysfunction in Parkinson’s disease. Translational Neurodegeneration. 2016;5:14-14. DOI: 10.1186/s40035-016-0060-6
  145. 145. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag A-E, Lang AE. Parkinson disease. Nature Reviews Disease Primers. 2017;3:17013-17013. DOI: 10.1038/nrdp.2017.13
  146. 146. Hirsch EVS, Hunot S. Neuroinflammation in Parkinson's disease. Journal of Neuroimmune Pharmacology. 2012:S210-S212. DOI: 10.1007/s11481-009-9176-0
  147. 147. Exner N, Lutz AK, Haass C, Winklhofer KF. Mitochondrial dysfunction in Parkinson's disease: Molecular mechanisms and pathophysiological consequences. The EMBO Journal. 2012;31:3038-3062. DOI: 10.1038/emboj.2012.170
  148. 148. Langston JW, Langston EB, Irwin I. MPTP-induced parkinsonism in human and non-human primates--clinical and experimental aspects. Acta Neurologica Scandinavica. Supplementum. 1984;100:49-54
  149. 149. Keeney PM, Xie J, Capaldi RA, Bennett JP Jr. Parkinson's disease brain mitochondrial complex I has oxidatively damaged subunits and is functionally impaired and misassembled. The Journal of Neuroscience. 2006;26:5256-5264. DOI: 10.1523/JNEUROSCI.0984-06.2006
  150. 150. Yoshino H, Nakagawa-Hattori Y, Kondo T, Mizuno Y. Mitochondrial complex I and II activities of lymphocytes and platelets in Parkinson's disease. Journal of Neural Transmission. Parkinson's Disease and Dementia Section. 1992;4:27-34
  151. 151. Esteves AR, Lu J, Rodova M, Onyango I, Lezi E, Dubinsky R, Lyons KE, Pahwa R, Burns JM, Cardoso SM, Swerdlow RH. Mitochondrial respiration and respiration-associated proteins in cell lines created through Parkinson's subject mitochondrial transfer. Journal of Neurochemistry. 2010;113:674-682. DOI: 10.1111/j.1471-4159.2010.06631.x
  152. 152. Bove J, Prou D, Perier C, Przedborski S. Toxin-induced models of Parkinson's disease. NeuroRx. 2005;2:484-494. DOI: 10.1602/neurorx.2.3.484
  153. 153. Xiong N, Long X, Xiong J, Jia M, Chen C, Huang J, Ghoorah D, Kong X, Lin Z, Wang T. Mitochondrial complex I inhibitor rotenone-induced toxicity and its potential mechanisms in Parkinson's disease models. Critical Reviews in Toxicology. 2012;42:613-632. DOI: 10.3109/10408444.2012.680431
  154. 154. Meredith GE, Rademacher DJ. MPTP mouse models of Parkinson's disease: An update. Journal of Parkinson's Disease. 2011;1:19-33. DOI: 10.3233/JPD-2011-11023
  155. 155. Esteves AR, Domingues AF, Ferreira IL, Januario C, Swerdlow RH, Oliveira CR, Cardoso SM. Mitochondrial function in Parkinson's disease cybrids containing an nt2 neuron-like nuclear background. Mitochondrion. 2008;8:219-228. DOI: 10.1016/j.mito.2008.03.004
  156. 156. Mann VM, Cooper JM, Krige D, Daniel SE, Schapira AH, Marsden CD. Brain, skeletal muscle and platelet homogenate mitochondrial function in Parkinson's disease. Brain. 1992;115(Pt 2):333-342
  157. 157. Bohnen NI, Minoshima S, Giordani B, Frey KA, Kuhl DE. Motor correlates of occipital glucose hypometabolism in Parkinson's disease without dementia. Neurology. 1999;52:541-546
  158. 158. Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurology. 2008;7:97-109. DOI: 10.1016/S1474-4422(07)70327-7
  159. 159. Lenaz G. The mitochondrial production of reactive oxygen species: Mechanisms and implications in human pathology. IUBMB Life. 2001;52:159-164. DOI: 10.1080/15216540152845957
  160. 160. Jenner P, Dexter DT, Sian J, Schapira AH, Marsden CD. Oxidative stress as a cause of nigral cell death in Parkinson's disease and incidental Lewy body disease. The Royal Kings and Queens Parkinson's Disease Research Group. Annals of Neurology. 1992;32(Suppl):S82-S87
  161. 161. Fahn S, Cohen G. The oxidant stress hypothesis in Parkinson's disease: Evidence supporting it. Annals of Neurology. 1992;32:804-812. DOI: 10.1002/ana.410320616
  162. 162. Blesa J, Trigo-Damas I, Quiroga-Varela A, Jackson-Lewis VR. Oxidative stress and Parkinson's disease. Frontiers in Neuroanatomy. 2015;9:91. DOI: 10.3389/fnana.2015.00091
  163. 163. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392:605-608. DOI: 10.1038/33416
  164. 164. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, Gonzalez-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004;304:1158-1160. DOI: 10.1126/science.1096284
  165. 165. Wu W, Xu H, Wang Z, Mao Y, Yuan L, Luo W, Cui Z, Cui T, Wang XL, Shen YH. PINK1-parkin-mediated Mitophagy protects mitochondrial integrity and prevents metabolic stress-induced endothelial injury. PLoS One. 2015;10:e0132499. DOI: 10.1371/journal.pone.0132499
  166. 166. Thomas RE, Andrews LA, Burman JL, Lin WY, Pallanck LJ. PINK1-parkin pathway activity is regulated by degradation of PINK1 in the mitochondrial matrix. PLoS Genetics. 2014;10:e1004279. DOI: 10.1371/journal.pgen.1004279
  167. 167. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate parkin. PLoS Biology. 2010;8:e1000298. DOI: 10.1371/journal.pbio.1000298
  168. 168. Bingol B, Sheng M. Mechanisms of mitophagy: PINK1, parkin, USP30 and beyond. Free Radical Biology & Medicine. 2016;100:210-222. DOI: 10.1016/j.freeradbiomed.2016.04.015
  169. 169. Nguyen TN, Padman BS, Lazarou M. Deciphering the molecular signals of PINK1/parkin Mitophagy. Trends in Cell Biology. 2016;26:733-744. DOI: 10.1016/j.tcb.2016.05.008
  170. 170. Narendra D, Walker JE, Youle R. Mitochondrial quality control mediated by PINK1 and parkin: Links to parkinsonism. Cold Spring Harbor Perspectives in Biology. 2012;4:a011338. DOI: 10.1101/cshperspect.a011338
  171. 171. Kitada T, Pisani A, Porter DR, Yamaguchi H, Tscherter A, Martella G, Bonsi P, Zhang C, Pothos EN, Shen J. Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:11441-11446. DOI: 10.1073/pnas.0702717104
  172. 172. Park J, Lee SB, Lee S, Kim Y, Song S, Kim S, Bae E, Kim J, Shong M, Kim JM, Chung J. Mitochondrial dysfunction in drosophila PINK1 mutants is complemented by parkin. Nature. 2006;441:1157-1161. DOI: 10.1038/nature04788
  173. 173. Arduino DM, Esteves AR, Cortes L, Silva DF, Patel B, Grazina M, Swerdlow RH, Oliveira CR, Cardoso SM. Mitochondrial metabolism in Parkinson's disease impairs quality control autophagy by hampering microtubule-dependent traffic. Human Molecular Genetics. 2012;21:4680-4702. DOI: 10.1093/hmg/dds309
  174. 174. Santos D, Esteves AR, Silva DF, Januario C, Cardoso SM. The impact of mitochondrial fusion and fission modulation in sporadic Parkinson's disease. Molecular Neurobiology. 2015;52:573-586. DOI: 10.1007/s12035-014-8893-4
  175. 175. Wiemerslage L, Ismael S, Lee D. Early alterations of mitochondrial morphology in dopaminergic neurons from Parkinson's disease-like pathology and time-dependent neuroprotection with D2 receptor activation. Mitochondrion. 2016;30:138-147. DOI: 10.1016/j.mito.2016.07.004
  176. 176. Nakamura K, Nemani VM, Azarbal F, Skibinski G, Levy JM, Egami K, Munishkina L, Zhang J, Gardner B, Wakabayashi J, Sesaki H, Cheng Y, Finkbeiner S, Nussbaum RL, Masliah E, Edwards RH. Direct membrane association drives mitochondrial fission by the Parkinson disease-associated protein alpha-synuclein. The Journal of Biological Chemistry. 2011;286:20710-20726. DOI: 10.1074/jbc.M110.213538
  177. 177. Lahiri V, Klionsky DJ. Functional impairment in RHOT1/Miro1 degradation and mitophagy is a shared feature in familial and sporadic Parkinson disease. Autophagy. 2017;13(8):1259-1261. DOI: 10.1080/15548627.2017.1327512
  178. 178. Hsieh CH, Shaltouki A, Gonzalez AE, Bettencourt da Cruz A, Burbulla LF, St Lawrence E, Schule B, Krainc D, palmer TD, Wang X. Functional impairment in miro degradation and mitophagy is a shared feature in familial and sporadic Parkinson's disease. Cell Stem Cell. 2016;19:709-724. DOI: 10.1016/j.stem.2016.08.002
  179. 179. Wang X, Winter D, Ashrafi G, Schlehe J, Wong YL, Selkoe D, Rice S, Steen J, LaVoie MJ, Schwarz TL. PINK1 and parkin target Miro for phosphorylation and degradation to arrest mitochondrial motility. Cell. 2011;147:893-906. DOI: 10.1016/j.cell.2011.10.018
  180. 180. Huang Y, Chegini F, Chua G, Murphy K, Gai W, Halliday GM. Macroautophagy in sporadic and the genetic form of Parkinson's disease with the A53T alpha-synuclein mutation. Transl Neurodegener. 2012;1:2. DOI: 10.1186/2047-9158-1-2
  181. 181. Nixon RA. The role of autophagy in neurodegenerative disease. Nature Medicine. 2013;19:983-997. DOI: 10.1038/nm.3232
  182. 182. Cartelli D, Ronchi C, Maggioni MG, Rodighiero S, Giavini E, Cappelletti G. Microtubule dysfunction precedes transport impairment and mitochondria damage in MPP+ −induced neurodegeneration. Journal of Neurochemistry. 2010;115:247-258. DOI: 10.1111/j.1471-4159.2010.06924.x
  183. 183. Cappelletti G, Casagrande F, Calogero A, De Gregorio C, Pezzoli G, Cartelli D. Linking microtubules to Parkinson's disease: The case of parkin. Biochemical Society Transactions. 2015;43:292-296. DOI: 10.1042/BST20150007
  184. 184. Scarffe LA, Stevens DA, Dawson VL, Dawson TM. Parkin and PINK1: Much more than mitophagy. Trends in Neurosciences. 2014;37:315-324. DOI: 10.1016/j.tins.2014.03.004
  185. 185. Livnat-Levanon N, Kevei E, Kleifeld O, Krutauz D, Segref A, Rinaldi T, Erpapazoglou Z, Cohen M, Reis N, Hoppe T, Glickman MH. Reversible 26S proteasome disassembly upon mitochondrial stress. Cell Reports. 2014;7:1371-1380. DOI: 10.1016/j.celrep.2014.04.030
  186. 186. Ross JM, Olson L, Coppotelli G. Mitochondrial and ubiquitin proteasome system dysfunction in ageing and disease: Two sides of the same coin? International Journal of Molecular Sciences. 2015;16:19458-19476. DOI: 10.3390/ijms160819458
  187. 187. McKenzie JA, Spielman LJ, Pointer CB, Lowry JR, Bajwa E, Lee CW, Klegeris A. Neuroinflammation as a common mechanism associated with the modifiable risk factors for Alzheimer's and Parkinson’s diseases. Current Aging Science. 2017;10(3):158-176. DOI: 10.2174/1874609810666170315113244
  188. 188. Subramaniam SR, Federoff HJ. Targeting microglial activation states as a therapeutic avenue in Parkinson's disease. Frontiers in Aging Neuroscience. 2017;9:176. DOI: 10.3389/fnagi.2017.00176
  189. 189. Hawkes CH, DelTredici K, Braak H. A timeline for Parkinson's disease. Parkinsonism & Related Disorders. 2010;16:79-84. DOI: 10.1016/j.parkreldis.2009.08.007
  190. 190. Savica R, Carlin JM, Grossardt BR, Bower JH, Ahlskog JE, Maraganore DM, Bharucha AE, Rocca WA. Medical records documentation of constipation preceding Parkinson disease: A case-control study. Neurology. 2009;73:1752-1758. DOI: 10.1212/WNL.0b013e3181c34af5
  191. 191. Soreq L, Israel Z, Bergman H, Soreq H. Advanced microarray analysis highlights modified neuro-immune signaling in nucleated blood cells from Parkinson's disease patients. Journal of Neuroimmunology. 2008;201-202:227-236. DOI: 10.1016/j.jneuroim.2008.06.019
  192. 192. 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 of Systematic Reviews. 2011;9:CD008454. DOI: 10.1002/14651858.CD008454.pub2
  193. 193. Chen H, Zhang SM, Hernan MA, Schwarzschild MA, Willett WC, Colditz GA, Speizer FE, Ascherio A. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Archives of Neurology. 2003;60:1059-1064. DOI: 10.1001/archneur.60.8.1059
  194. 194. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson's disease: Systematic review and meta-analysis of observational studies. Drugs & Aging. 2009;26:769-779. DOI: 10.2165/11316780-000000000-00000
  195. 195. Espinosa-Oliva AM, de Pablos RM, Villaran RF, Arguelles S, Venero JL, Machado A, Cano J. Stress is critical for LPS-induced activation of microglia and damage in the rat hippocampus. Neurobiology of Aging. 2011;32:85-102. DOI: 10.1016/j.neurobiolaging.2009.01.012
  196. 196. Pott Godoy MC, Ferrari CC, Pitossi FJ. Nigral neurodegeneration triggered by striatal AdIL-1 administration can be exacerbated by systemic IL-1 expression. Journal of Neuroimmunology. 2010;222:29-39. DOI: 10.1016/j.jneuroim.2010.02.018
  197. 197. McGeer PL, McGeer EG. Glial reactions in Parkinson's disease. Movement Disorders. 2008;23:474-483. DOI: 10.1002/mds.21751
  198. 198. Liu M, Bing G. Lipopolysaccharide animal models for Parkinson's disease. Journal of Parkinson's Disease. 2011;2011:327089. DOI: 10.4061/2011/327089
  199. 199. Herrera AJ, Espinosa-Oliva AM, Carrillo-Jimenez A, Oliva-Martin MJ, Garcia-Revilla J, Garcia-Quintanilla A, dePablos RM, Venero JL. Relevance of chronic stress and the two faces of microglia in Parkinson's disease. Frontiers in Cellular Neuroscience. 2015;9:312. DOI: 10.3389/fncel.2015.00312
  200. 200. dePablos RM, Herrera AJ, Espinosa-Oliva AM, Sarmiento M, Munoz MF, Machado A, Venero JL. Chronic stress enhances microglia activation and exacerbates death of nigral dopaminergic neurons under conditions of inflammation. Journal of Neuroinflammation. 2014;11:34. DOI: 10.1186/1742-2094-11-34
  201. 201. Dominguez-Meijide A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Dopamine modulates astroglial and microglial activity via glial renin-angiotensin system in cultures. Brain, Behavior, and Immunity. 2017;62:277-290. DOI: 10.1016/j.bbi.2017.02.013
  202. 202. Karlsson O, Lindquist NG. Melanin affinity and its possible role in neurodegeneration. Journal of Neural Transmission (Vienna). 2013;120:1623-1630. DOI: 10.1007/s00702-013-1062-5
  203. 203. Viceconte N, Burguillos MA, Herrera AJ, De Pablos RM, Joseph B, Venero JL. Neuromelanin activates proinflammatory microglia through a caspase-8-dependent mechanism. Journal of Neuroinflammation. 2015;12:5. DOI: 10.1186/s12974-014-0228-x
  204. 204. Lema Tome CM, Tyson T, Rey NL, Grathwohl S, Britschgi M, Brundin P. Inflammation and alpha-synuclein's prion-like behavior in Parkinson's disease--is there a link? Molecular Neurobiology. 2013;47:561-574. DOI: 10.1007/s12035-012-8267-8
  205. 205. Klegeris A, Pelech S, Giasson BI, Maguire J, Zhang H, McGeer EG, McGeer PL. Alpha-synuclein activates stress signaling protein kinases in THP-1 cells and microglia. Neurobiology of Aging. 2008;29:739-752. DOI: 10.1016/j.neurobiolaging.2006.11.013
  206. 206. Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, Wilson B, Zhang W, Zhou Y, Hong JS, Zhang J. Aggregated alpha-synuclein activates microglia: A process leading to disease progression in Parkinson's disease. The FASEB Journal. 2005;19:533-542. DOI: 10.1096/fj.04-2751com
  207. 207. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, Kusel VI, Collura R, Roberts J, Griffith A, Samii A, Scott WK, Nutt J, Factor SA, Payami H. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson's disease. Nature Genetics. 2010;42:781-785. DOI: 10.1038/ng.642
  208. 208. International Parkinson Disease Genomics Consortium, Nalls MA, Plagnol V, Hernandez DG, Sharma M, Sheerin UM, Saad M, Simon-Sanchez J, Schulte C, Lesage S, Sveinbjornsdottir S, Stefansson K, Martinez M, Hardy J, Heutink P, Brice A, Gasser T, Singleton AB, Wood NW. Imputation of sequence variants for identification of genetic risks for Parkinson's disease: A meta-analysis of genome-wide association studies. Lancet. 2011;377:641-649. DOI: 10.1016/S0140-6736(10)62345-8
  209. 209. Pihlstrom L, Axelsson G, Bjornara KA, Dizdar N, Fardell C, Forsgren L, Holmberg B, Larsen JP, Linder J, Nissbrandt H, Tysnes OB, Ohman E, Dietrichs E, Toft M. Supportive evidence for 11 loci from genome-wide association studies in Parkinson's disease. Neurobiology of Aging. 1708;2013(34):e1707-e1713. DOI: 10.1016/j.neurobiolaging.2012.10.019
  210. 210. Russo I, Bubacco L, Greggio E. LRRK2 and neuroinflammation: Partners in crime in Parkinson's disease? Journal of Neuroinflammation. 2014;11:52. DOI: 10.1186/1742-2094-11-52
  211. 211. Harms AS, Cao S, Rowse AL, Thome AD, Li X, Mangieri LR, Cron RQ, Shacka JJ, Raman C, Standaert DG. MHCII is required for alpha-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. The Journal of Neuroscience. 2013;33:9592-9600. DOI: 10.1523/JNEUROSCI.5610-12.2013
  212. 212. Su X, Maguire-Zeiss KA, Giuliano R, Prifti L, Venkatesh K, Federoff HJ. Synuclein activates microglia in a model of Parkinson's disease. Neurobiology of Aging. 2008;29:1690-1701. DOI: 10.1016/j.neurobiolaging.2007.04.006
  213. 213. Su X, Federoff HJ, Maguire-Zeiss KA. Mutant alpha-synuclein overexpression mediates early proinflammatory activity. Neurotoxicity Research. 2009;16:238-254. DOI: 10.1007/s12640-009-9053-x
  214. 214. Beraud D, Hathaway HA, Trecki J, Chasovskikh S, Johnson DA, Johnson JA, Federoff HJ, Shimoji M, Mhyre TR, Maguire-Zeiss KA. Microglial activation and antioxidant responses induced by the Parkinson's disease protein alpha-synuclein. Journal of Neuroimmune Pharmacology. 2013;8:94-117. DOI: 10.1007/s11481-012-9401-0
  215. 215. Reynolds AD, Glanzer JG, Kadiu I, Ricardo-Dukelow M, Chaudhuri A, Ciborowski P, Cerny R, Gelman B, Thomas MP, Mosley RL, Gendelman HE. Nitrated alpha-synuclein-activated microglial profiling for Parkinson's disease. Journal of Neurochemistry. 2008;104:1504-1525. DOI: 10.1111/j.1471-4159.2007.05087.x
  216. 216. Couch Y, Alvarez-Erviti L, Sibson NR, Wood MJ, Anthony DC. The acute inflammatory response to intranigral alpha-synuclein differs significantly from intranigral lipopolysaccharide and is exacerbated by peripheral inflammation. Journal of Neuroinflammation. 2011;8:166. DOI: 10.1186/1742-2094-8-166
  217. 217. Rahmani F, Kamalian A, Aarabi MH. New evidence comes to light: How is alpha-synuclein aggregation related to mitochondrial protein import in Parkinson's disease? Movement Disorders. 2017;32:107. DOI: 10.1002/mds.26889
  218. 218. Maekawa T, Kubo M, Yokoyama I, Ohta E, Obata F. Age-dependent and cell-population-restricted LRRK2 expression in normal mouse spleen. Biochemical and Biophysical Research Communications. 2010;392:431-435. DOI: 10.1016/j.bbrc.2010.01.041
  219. 219. Miklossy J, Arai T, Guo JP, Klegeris A, Yu S, McGeer EG, McGeer PL. LRRK2 expression in normal and pathologic human brain and in human cell lines. Journal of Neuropathology and Experimental Neurology. 2006;65:953-963. DOI: 10.1097/01.jnen.0000235121.98052.54
  220. 220. Gillardon F, Schmid R, Draheim H. Parkinson's disease-linked leucine-rich repeat kinase 2(R1441G) mutation increases proinflammatory cytokine release from activated primary microglial cells and resultant neurotoxicity. Neuroscience. 2012;208:41-48. DOI: 10.1016/j.neuroscience.2012.02.001
  221. 221. Lopez de Maturana R, Aguila JC, Sousa A, Vazquez N, Del Rio P, Aiastui A, Gorostidi A, Lopez de Munain A, Sanchez-Pernaute R. Leucine-rich repeat kinase 2 modulates cyclooxygenase 2 and the inflammatory response in idiopathic and genetic Parkinson's disease. Neurobiology of Aging. 2014;35:1116-1124. DOI: 10.1016/j.neurobiolaging.2013.11.018
  222. 222. Kim B, Yang MS, Choi D, Kim JH, Kim HS, Seol W, Choi S, Jou I, Kim EY, Joe EH. Impaired inflammatory responses in murine Lrrk2-knockdown brain microglia. PLoS One. 2012;7:e34693. DOI: 10.1371/journal.pone.0034693
  223. 223. McGeer PL, Itagaki S, Akiyama H, McGeer EG. Rate of cell death in parkinsonism indicates active neuropathological process. Annals of Neurology. 1988;24:574-576. DOI: 10.1002/ana.410240415
  224. 224. Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA, Karluk D. Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure. Annals of Neurology. 1999;46:598-605
  225. 225. Banati RB, Daniel SE, Blunt SB. Glial pathology but absence of apoptotic nigral neurons in long-standing Parkinson's disease. Movement Disorders. 1998;13:221-227. DOI: 10.1002/mds.870130205
  226. 226. Mirza B, Hadberg H, Thomsen P, Moos T. The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson's disease. Neuroscience. 2000;95:425-432
  227. 227. Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, Hashizume Y. Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson's disease brains. Acta Neuropathologica. 2003;106:518-526. DOI: 10.1007/s00401-003-0766-2
  228. 228. Miklossy J, Doudet DD, Schwab C, Yu S, McGeer EG, McGeer PL. Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys. Experimental Neurology. 2006;197:275-283. DOI: 10.1016/j.expneurol.2005.10.034
  229. 229. Damier P, Hirsch EC, Zhang P, Agid Y, Javoy-Agid F. Glutathione peroxidase, glial cells and Parkinson's disease. Neuroscience. 1993;52:1-6
  230. 230. Fiszer U, Mix E, Fredrikson S, Kostulas V, Link H. Parkinson's disease and immunological abnormalities: Increase of HLA-DR expression on monocytes in cerebrospinal fluid and of CD45RO+ T cells in peripheral blood. Acta Neurologica Scandinavica. 1994;90:160-166
  231. 231. Bas J, Calopa M, Mestre M, Mollevi DG, Cutillas B, Ambrosio S, Buendia E. Lymphocyte populations in Parkinson's disease and in rat models of parkinsonism. Journal of Neuroimmunology. 2001;113:146-152
  232. 232. Baba Y, Kuroiwa A, Uitti RJ, Wszolek ZK, Yamada T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism & Related Disorders. 2005;11:493-498. DOI: 10.1016/j.parkreldis.2005.07.005
  233. 233. Brochard V, Combadiere B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, Bonduelle O, Alvarez-Fischer D, Callebert J, Launay JM, Duyckaerts C, Flavell RA, Hirsch EC, Hunot S. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. The Journal of Clinical Investigation. 2009;119:182-192. DOI: 10.1172/JCI36470
  234. 234. Stone DK, Reynolds AD, Mosley RL, Gendelman HE. Innate and adaptive immunity for the pathobiology of Parkinson's disease. Antioxidants & Redox Signaling. 2009;11:2151-2166. DOI: 10.1089/ARS.2009.2460
  235. 235. Cabezas R, Avila M, Gonzalez J, El-Bacha RS, Baez E, Garcia-Segura LM, Jurado Coronel JC, Capani F, Cardona-Gomez GP, Barreto GE. Astrocytic modulation of blood brain barrier: Perspectives on Parkinson's disease. Frontiers in Cellular Neuroscience. 2014;8:211. DOI: 10.3389/fncel.2014.00211
  236. 236. Forsyth CB, Shannon KM, Kordower JH, Voigt RM, Shaikh M, Jaglin JA, Estes JD, Dodiya HB, Keshavarzian A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson's disease. PLoS One. 2011;6:e28032. DOI: 10.1371/journal.pone.0028032
  237. 237. Devos D, Lebouvier T, Lardeux B, Biraud M, Rouaud T, Pouclet H, Coron E, Bruley des Varannes S, Naveilhan P, Nguyen JM, Neunlist M, Derkinderen P. Colonic inflammation in Parkinson's disease. Neurobiology of Disease. 2013;50:42-48. DOI: 10.1016/j.nbd.2012.09.007
  238. 238. Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T. Tumor necrosis factor-alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neuroscience Letters. 1994;165:208-210
  239. 239. Hunot S, Dugas N, Faucheux B, Hartmann A, Tardieu M, Debre P, Agid Y, Dugas B, Hirsch EC. FcepsilonRII/CD23 is expressed in Parkinson's disease and induces, in vitro, production of nitric oxide and tumor necrosis factor-alpha in glial cells. The Journal of Neuroscience. 1999;19:3440-3447
  240. 240. Mogi M, Harada M, Kondo T, Riederer P, Inagaki H, Minami M, Nagatsu T. Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neuroscience Letters. 1994;180:147-150
  241. 241. Mogi M, Harada M, Kondo T, Narabayashi H, Riederer P, Nagatsu T. Transforming growth factor-beta 1 levels are elevated in the striatum and in ventricular cerebrospinal fluid in Parkinson's disease. Neuroscience Letters. 1995;193:129-132
  242. 242. Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T. Interleukin (IL)-1 beta, IL-2, IL-4, IL-6 and transforming growth factor-alpha levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson's disease. Neuroscience Letters. 1996;211:13-16
  243. 243. Mogi M, Kondo T, Mizuno Y, Nagatsu T. p53 protein, interferon-gamma, and NF-kappaB levels are elevated in the parkinsonian brain. Neuroscience Letters. 2007;414:94-97. DOI: 10.1016/j.neulet.2006.12.003
  244. 244. Boka G, Anglade P, Wallach D, Javoy-Agid F, Agid Y, Hirsch EC. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson's disease. Neuroscience Letters. 1994;172:151-154
  245. 245. Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose H, Nagatsu T. Caspase activities and tumor necrosis factor receptor R1 (p55) level are elevated in the substantia nigra from parkinsonian brain. Journal of Neural Transmission (Vienna). 2000;107:335-341. DOI: 10.1007/s007020050028
  246. 246. Knott C, Stern G, Wilkin GP. Inflammatory regulators in Parkinson's disease: iNOS, lipocortin-1, and cyclooxygenases-1 and -2. Molecular and Cellular Neurosciences. 2000;16:724-739. DOI: 10.1006/mcne.2000.0914
  247. 247. Choi DK, Pennathur S, Perier C, Tieu K, Teismann P, Wu DC, Jackson-Lewis V, Vila M, Vonsattel JP, Heinecke JW, Przedborski S. Ablation of the inflammatory enzyme myeloperoxidase mitigates features of Parkinson's disease in mice. The Journal of Neuroscience. 2005;25:6594-6600. DOI: 10.1523/JNEUROSCI.0970-05.2005
  248. 248. Stypula G, Kunert-Radek J, Stepien H, Zylinska K, Pawlikowski M. Evaluation of interleukins, ACTH, cortisol and prolactin concentrations in the blood of patients with parkinson's disease. Neuroimmunomodulation. 1996;3:131-134
  249. 249. Dobbs RJ, Charlett A, Purkiss AG, Dobbs SM, Weller C, Peterson DW. Association of circulating TNF-alpha and IL-6 with ageing and parkinsonism. Acta Neurologica Scandinavica. 1999;100:34-41
  250. 250. Rentzos M, Nikolaou C, Andreadou E, Paraskevas GP, Rombos A, Zoga M, Tsoutsou A, Boufidou F, Kapaki E, Vassilopoulos D. Circulating interleukin-15 and RANTES chemokine in Parkinson's disease. Acta Neurologica Scandinavica. 2007;116:374-379. DOI: 10.1111/j.1600-0404.2007.00894.x
  251. 251. Rentzos M, Nikolaou C, Andreadou E, Paraskevas GP, Rombos A, Zoga M, Tsoutsou A, Boufidou F, Kapaki E, Vassilopoulos D. Circulating interleukin-10 and interleukin-12 in Parkinson's disease. Acta Neurologica Scandinavica. 2009;119:332-337. DOI: 10.1111/j.1600-0404.2008.01103.x
  252. 252. Dufek M, Hamanova M, Lokaj J, Goldemund D, Rektorova I, Michalkova Z, Sheardova K, Rektor I. Serum inflammatory biomarkers in Parkinson's disease. Parkinsonism & Related Disorders. 2009;15:318-320. DOI: 10.1016/j.parkreldis.2008.05.014
  253. 253. Scalzo P, Kummer A, Cardoso F, Teixeira AL. Increased serum levels of soluble tumor necrosis factor-alpha receptor-1 in patients with Parkinson's disease. Journal of Neuroimmunology. 2009;216:122-125. DOI: 10.1016/j.jneuroim.2009.08.001
  254. 254. Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di Nicola M, Onofrj M. Peripheral cytokines profile in Parkinson's disease. Brain, Behavior, and Immunity. 2009;23:55-63. DOI: 10.1016/j.bbi.2008.07.003
  255. 255. Tang P, Chong L, Li X, Liu Y, Liu P, Hou C, Li R. Correlation between serum RANTES levels and the severity of Parkinson's disease. Oxidative Medicine and Cellular Longevity. 2014;2014:208408. DOI: 10.1155/2014/208408
  256. 256. Blum-Degen D, Muller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer's and de novo Parkinson's disease patients. Neuroscience Letters. 1995;202:17-20
  257. 257. Gonzalez-Scarano F, Baltuch G. Microglia as mediators of inflammatory and degenerative diseases. Annual Review of Neuroscience. 1999;22:219-240. DOI: 10.1146/annurev.neuro.22.1.219
  258. 258. Maetzler W, Berg D, Schalamberidze N, Melms A, Schott K, Mueller JC, Liaw L, Gasser T, Nitsch C. Osteopontin is elevated in Parkinson's disease and its absence leads to reduced neurodegeneration in the MPTP model. Neurobiology of Disease. 2007;25:473-482. DOI: 10.1016/j.nbd.2006.10.020
  259. 259. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, Eggert K, Oertel W, Banati RB, Brooks DJ. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson's disease. Neurobiology of Disease. 2006;21:404-412. DOI: 10.1016/j.nbd.2005.08.002
  260. 260. Ouchi Y, Yoshikawa E, Sekine Y, Futatsubashi M, Kanno T, Ogusu T, Torizuka T. Microglial activation and dopamine terminal loss in early Parkinson's disease. Annals of Neurology. 2005;57:168-175. DOI: 10.1002/ana.20338
  261. 261. Iannaccone S, Cerami C, Alessio M, Garibotto V, Panzacchi A, Olivieri S, Gelsomino G, Moresco RM, Perani D. In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson's disease. Parkinsonism & Related Disorders. 2013;19:47-52. DOI: 10.1016/j.parkreldis.2012.07.002
  262. 262. Ghadery C, Koshimori Y, Coakeley S, Harris M, Rusjan P, Kim J, Houle S, Strafella AP. Microglial activation in Parkinson's disease using [18F]-FEPPA. Journal of Neuroinflammation. 2017;14:8. DOI: 10.1186/s12974-016-0778-1
  263. 263. Yang L, Guo C, Zhu J, Feng Y, Chen W, Feng Z, Wang D, Sun S, Lin W, Wang Y. Increased levels of pro-inflammatory and anti-inflammatory cellular responses in Parkinson's disease patients: Search for a disease indicator. Medical Science Monitor. 2017;23:2972-2978
  264. 264. He Y, Appel S, Le W. Minocycline inhibits microglial activation and protects nigral cells after 6-hydroxydopamine injection into mouse striatum. Brain Research. 2001;909:187-193
  265. 265. McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Annals of Neurology. 2003;54:599-604. DOI: 10.1002/ana.10728
  266. 266. Depino AM, Earl C, Kaczmarczyk E, Ferrari C, Besedovsky H, delRey A, Pitossi FJ, Oertel WH. Microglial activation with atypical proinflammatory cytokine expression in a rat model of Parkinson's disease. The European Journal of Neuroscience. 2003;18:2731-2742
  267. 267. Barcia C, Sanchez Bahillo A, Fernandez-Villalba E, Bautista V, Poza YPM, Fernandez-Barreiro A, Hirsch EC, Herrero MT. Evidence of active microglia in substantia nigra pars compacta of parkinsonian monkeys 1 year after MPTP exposure. Glia. 2004;46:402-409. DOI: 10.1002/glia.20015
  268. 268. Sherer TB, Betarbet R, Kim JH, Greenamyre JT. Selective microglial activation in the rat rotenone model of Parkinson's disease. Neuroscience Letters. 2003;341:87-90
  269. 269. Shaikh SB, Nicholson LF. Effects of chronic low dose rotenone treatment on human microglial cells. Molecular Neurodegeneration. 2009;4:55. DOI: 10.1186/1750-1326-4-55
  270. 270. Armentero MT, Levandis G, Nappi G, Bazzini E, Blandini F. Peripheral inflammation and neuroprotection: Systemic pretreatment with complete Freund's adjuvant reduces 6-hydroxydopamine toxicity in a rodent model of Parkinson's disease. Neurobiology of Disease. 2006;24:492-505. DOI: 10.1016/j.nbd.2006.08.016
  271. 271. Wheeler CJ, Seksenyan A, Koronyo Y, Rentsendorj A, Sarayba D, Wu H, Gragg A, Siegel E, Thomas D, Espinosa A, Thompson K, Black K, Koronyo-Hamaoui M, Pechnick R, Irvin DK. T-lymphocyte deficiency exacerbates behavioral deficits in the 6-OHDA unilateral lesion rat model for Parkinson's disease. Journal of Neurology & Neurophysiology. 2014;5(3):209. DOI: 10.4172/2155-9562.1000209
  272. 272. Rodrigues RW, Gomide VC, Chadi G. Astroglial and microglial reaction after a partial nigrostriatal degeneration induced by the striatal injection of different doses of 6-hydroxydopamine. The International Journal of Neuroscience. 2001;109:91-126
  273. 273. Rodrigues RW, Gomide VC, Chadi G. Astroglial and microglial activation in the wistar rat ventral tegmental area after a single striatal injection of 6-hydroxydopamine. The International Journal of Neuroscience. 2004;114:197-216. DOI: 10.1080/00207450490249338
  274. 274. Breidert T, Callebert J, Heneka MT, Landreth G, Launay JM, Hirsch EC. Protective action of the peroxisome proliferator-activated receptor-gamma agonist pioglitazone in a mouse model of Parkinson's disease. Journal of Neurochemistry. 2002;82:615-624
  275. 275. Herrera AJ, Castano A, Venero JL, Cano J, Machado A. The single intranigral injection of LPS as a new model for studying the selective effects of inflammatory reactions on dopaminergic system. Neurobiology of Disease. 2000;7:429-447. DOI: 10.1006/nbdi.2000.0289
  276. 276. Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, Knapp DJ, Crews FT. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia. 2007;55:453-462. DOI: 10.1002/glia.20467
  277. 277. Castano A, Herrera AJ, Cano J, Machado A. Lipopolysaccharide intranigral injection induces inflammatory reaction and damage in nigrostriatal dopaminergic system. Journal of Neurochemistry. 1998;70:1584-1592
  278. 278. Kim WG, Mohney RP, Wilson B, Jeohn GH, Liu B, Hong JS. Regional difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat brain: Role of microglia. The Journal of Neuroscience. 2000;20:6309-6316
  279. 279. Xie X, Luo X, Liu N, Li X, Lou F, Zheng Y, Ren Y. Monocytes, microglia, and CD200-CD200R1 signaling are essential in the transmission of inflammation from the periphery to the central nervous system. Journal of Neurochemistry. 2017;141:222-235. DOI: 10.1111/jnc.13972
  280. 280. Carvey PM, Chang Q, Lipton JW, Ling Z. Prenatal exposure to the bacteriotoxin lipopolysaccharide leads to long-term losses of dopamine neurons in offspring: A potential, new model of Parkinson's disease. Frontiers in Bioscience. 2003;8:s826-s837
  281. 281. Ling Z, Gayle DA, Ma SY, Lipton JW, Tong CW, Hong JS, Carvey PM. Utero bacterial endotoxin exposure causes loss of tyrosine hydroxylase neurons in the postnatal rat midbrain. Movement Disorders. 2002;17:116-124
  282. 282. Ling Z, Chang QA, Tong CW, Leurgans SE, Lipton JW, Carvey PM. Rotenone potentiates dopamine neuron loss in animals exposed to lipopolysaccharide prenatally. Experimental Neurology. 2004;190:373-383. DOI: 10.1016/j.expneurol.2004.08.006
  283. 283. Arguelles S, Herrera AJ, Carreno-Muller E, dePablos RM, Villaran RF, Espinosa-Oliva AM, Machado A, Cano J. Degeneration of dopaminergic neurons induced by thrombin injection in the substantia nigra of the rat is enhanced by dexamethasone: Role of monoamine oxidase enzyme. Neurotoxicology. 2010;31:55-66. DOI: 10.1016/j.neuro.2009.12.001
  284. 284. Carreno-Muller E, Herrera AJ, de Pablos RM, Tomas-Camardiel M, Venero JL, Cano J, Machado A. Thrombin induces in vivo degeneration of nigral dopaminergic neurones along with the activation of microglia. Journal of Neurochemistry. 2003;84:1201-1214
  285. 285. Ara J, Przedborski S, Naini AB, Jackson-Lewis V, Trifiletti RR, Horwitz J, Ischiropoulos H. Inactivation of tyrosine hydroxylase by nitration following exposure to peroxynitrite and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Proceedings of the National Academy of Sciences of the United States of America. 1998;95:7659-7663
  286. 286. Przedborski S, Chen Q, Vila M, Giasson BI, Djaldatti R, Vukosavic S, Souza JM, Jackson-Lewis V, Lee VM, Ischiropoulos H. Oxidative post-translational modifications of alpha-synuclein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. Journal of Neurochemistry. 2001;76:637-640
  287. 287. De Lella Ezcurra AL, Chertoff M, Ferrari C, Graciarena M, Pitossi F. Chronic expression of low levels of tumor necrosis factor-alpha in the substantia nigra elicits progressive neurodegeneration, delayed motor symptoms and microglia/macrophage activation. Neurobiology of Disease. 2010;37:630-640. DOI: 10.1016/j.nbd.2009.11.018
  288. 288. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805-820. DOI: 10.1016/j.cell.2010.01.022
  289. 289. West AP, Koblansky AA, Ghosh S. Recognition and signaling by toll-like receptors. Annual Review of Cell and Developmental Biology. 2006;22:409-437. DOI: 10.1146/annurev.cellbio.21.122303.115827
  290. 290. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464:104-107. DOI: 10.1038/nature08780
  291. 291. Taanman JW. The mitochondrial genome: Structure, transcription, translation and replication. Biochimica et Biophysica Acta. 1999;1410:103-123
  292. 292. Rabiet MJ, Huet E, Boulay F. The N-formyl peptide receptors and the anaphylatoxin C5a receptors: An overview. Biochimie. 2007;89:1089-1106. DOI: 10.1016/j.biochi.2007.02.015
  293. 293. Gurung P, Lukens JR, Kanneganti TD. Mitochondria: Diversity in the regulation of the NLRP3 inflammasome. Trends in Molecular Medicine. 2015;21:193-201. DOI: 10.1016/j.molmed.2014.11.008
  294. 294. Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nature Reviews. Immunology. 2013;13:397-411. DOI: 10.1038/nri3452
  295. 295. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821-832. DOI: 10.1016/j.cell.2010.01.040
  296. 296. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469:221-225. DOI: 10.1038/nature09663
  297. 297. Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nature Immunology. 2011;12:222-230. DOI: 10.1038/ni.1980
  298. 298. Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM, Rentsendorj A, Vargas M, Guerrero C, Wang Y, Fitzgerald KA, Underhill DM, Town T, Arditi M. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36:401-414. DOI: 10.1016/j.immuni.2012.01.009
  299. 299. deAndrade RI, Einicker-Lamas M, Roney Bernardo R, Previatto LM, Mohana-Borges R, Morgado-Diaz JA, Benchimol M. Cardiolipin in hydrogenosomes: Evidence of symbiotic origin. Eukaryotic Cell. 2006;5:784-787. DOI: 10.1128/EC.5.4.784-787.2006
  300. 300. Kutschera U, Niklas KJ. Endosymbiosis, cell evolution, and speciation. Theory in Biosciences. 2005;124:1-24. DOI: 10.1016/j.thbio.2005.04.001
  301. 301. Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, Sadler JJ, Knepper-Adrian V, Han R, Qiao L, Eisenbarth SC, Nauseef WM, Cassel SL, Sutterwala FS. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity. 2013;39:311-323. DOI: 10.1016/j.immuni.2013.08.001
  302. 302. Little JP, Simtchouk S, Schindler SM, Villanueva EB, Gill NE, Walker DG, Wolthers KR, Klegeris A. Mitochondrial transcription factor a (Tfam) is a pro-inflammatory extracellular signaling molecule recognized by brain microglia. Molecular and Cellular Neurosciences. 2014;60:88-96. DOI: 10.1016/j.mcn.2014.04.003
  303. 303. Davis CH, Kim KY, Bushong EA, Mills EA, Boassa D, Shih T, Kinebuchi M, Phan S, Zhou Y, Bihlmeyer NA, Nguyen JV, Jin Y, Ellisman MH, Marsh-Armstrong N. Transcellular degradation of axonal mitochondria. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:9633-9638. DOI: 10.1073/pnas.1404651111
  304. 304. Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, Ji X, Lo EH. Transfer of mitochondria from astrocytes to neurons after stroke. Nature. 2016;535:551-555. DOI: 10.1038/nature18928

Written By

A. Raquel Esteves, Diana F. Silva, Daniel Santos, Emanuel Candeias, Filipa Filipe and Sandra M. Cardoso

Submitted: 29 May 2017 Reviewed: 21 November 2017 Published: 29 August 2018