Next Article in Journal
FDG and Non-FDG Radiopharmaceuticals for PET Imaging in Invasive Lobular Breast Carcinoma
Next Article in Special Issue
The Effects of Serping1 siRNA in α-Synuclein Regulation in MPTP-Induced Parkinson’s Disease
Previous Article in Journal
Dynamics of Gene Expression Profiling and Identification of High-Risk Patients for Severe COVID-19
Previous Article in Special Issue
The Effectiveness and Toxicity of Frameless CyberKnife Based Radiosurgery for Parkinson’s Disease—Phase II Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of microRNAs on Mitochondrial Function and Immunity: Relevance to Parkinson’s Disease

by
Beatriz F. S. Guedes
1,
Sandra Morais Cardoso
1,2 and
Ana Raquel Esteves
1,3,*
1
CNC–Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
2
Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
3
IIIUC–Institute for Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(5), 1349; https://doi.org/10.3390/biomedicines11051349
Submission received: 23 March 2023 / Revised: 21 April 2023 / Accepted: 28 April 2023 / Published: 3 May 2023

Abstract

:
Parkinson’s Disease (PD), the second most common neurodegenerative disorder, is characterised by the severe loss of dopaminergic neurons in the Substantia Nigra pars compacta (SNpc) and by the presence of Lewy bodies. PD is diagnosed upon the onset of motor symptoms, such as bradykinesia, resting tremor, rigidity, and postural instability. It is currently accepted that motor symptoms are preceded by non-motor features, such as gastrointestinal dysfunction. In fact, it has been proposed that PD might start in the gut and spread to the central nervous system. Growing evidence reports that the gut microbiota, which has been found to be altered in PD patients, influences the function of the central and enteric nervous systems. Altered expression of microRNAs (miRNAs) in PD patients has also been reported, many of which regulate key pathological mechanisms involved in PD pathogenesis, such as mitochondrial dysfunction and immunity. It remains unknown how gut microbiota regulates brain function; however, miRNAs have been highlighted as important players. Remarkably, numerous studies have depicted the ability of miRNAs to modulate and be regulated by the host’s gut microbiota. In this review, we summarize the experimental and clinical studies implicating mitochondrial dysfunction and immunity in PD. Moreover, we gather recent data on miRNA involvement in these two processes. Ultimately, we discuss the reciprocal crosstalk between gut microbiota and miRNAs. Studying the bidirectional interaction of gut microbiome–miRNA might elucidate the aetiology and pathogenesis of gut-first PD, which could lead to the application of miRNAs as potential biomarkers or therapeutical targets for PD.

1. Introduction

Parkinson’s Disease

Parkinson’s disease (PD) was first described by James Parkinson more than two centuries ago, in “An Essay on the Shaking Palsy” in 1817. PD is the second most common progressive neurodegenerative disorder, after Alzheimer’s Disease, with more than 10 million people affected worldwide in 2021. Its incidence and prevalence increase steadily with age (median age at onset is 60 years old), and men are 1.5 times more likely to suffer from the disease than women [1].
PD is clinically characterised by the development of numerous motor symptoms, including bradykinesia, resting tremor, rigidity, and postural instability [2]. These motor symptoms develop as a consequence of the severe loss of dopaminergic neurons in the Substantia Nigra pars compacta (SNpc), and only emerge when striatal dopamine levels are decreased by 60–70% as a result of the degeneration of 40–60% of neurons in the SNpc [3]. The presence of intracytoplasmic protein inclusions of α-synuclein, known as Lewy bodies (LBs), is another important pathological hallmark of PD. Furthermore, it is believed that PD progression might also affect non-dopaminergic pathways prior to the onset of nigral neurodegeneration, leading to the manifestation of several non-motor symptoms that appear at a prodromic stage, up to 15–20 years before the onset of motor symptoms (Figure 1) [2]. Based on various population studies, a multitude of non-motor symptoms have been commonly associated with prodromal PD, namely, hyposmia, constipation, depression, dysautonomia, and rapid eye movement (REM) sleep behaviour disorder (RBD) (Figure 1) [2,3].
Since the discovery of LBs, it has been clear that a variety of other neuronal populations were affected by these proteinaceous inclusions, and not only the neurons in the SNpc [4]. In order to determine whether the pathology affects the nigral and extranigral structures simultaneously, Braak and colleagues conducted various studies, which culminated in the formulation of a six-stage hypothesis for the progression of Lewy pathology (LP) in sporadic PD [5]. According to Braak’s hypothesis, PD pathogenesis might initiate in non-dopaminergic areas, such as the olfactory bulb and the enteric nervous system (ENS), years and even decades before spreading, via the olfactory tract and the vagal nerve, respectively, towards the central nervous system (CNS), eventually reaching the SNpc and triggering the emergence of motor symptoms [6]. The earliest stages of LP (before SNpc involvement) have been linked to the prodromal non-motor symptoms of PD, such as olfactory impairment and gastrointestinal dysfunctions [7], since consistent evidence has demonstrated the presence of α-synuclein aggregates in the gut and in the neurons of the olfactory bulb during the prodromal phase, before the clinical diagnosis of PD [6,8]. Nonetheless, studies showed that some PD cases cannot be staged by the Braak staging system, as some patients presented with evident LB pathology first in the locus coeruleus, SNpc, and the amygdala, and then in the dorsal motor nucleus of the vagus (DMV) and gut, representing a rostro-caudal route of α-synuclein transmission from the CNS to the ENS [9]. This neuropathological evidence gave rise to two distinct hypotheses: body-first PD vs. brain-first PD. In the body-first subtype of PD, LP begins in the enteric or autonomic nervous system and spreads to the CNS via the vagal nerve, which aligns with Braak’s proposed staging [10]. These patients display more autonomic symptoms and a longer prodromal phase, with the development of non-motor symptoms such as gastrointestinal dysfunction, RBD, and hyposmia [9]. The opposite is true for the brain-first subtype of PD, where brain pathology presumably initiates in the amygdala or in closely connected areas such as the olfactory bulb [9]. These patients present with fewer autonomic symptoms and a shorter prodromal phase, with less frequent development of non-motor symptoms such as RBD or hyposmia [9]. Present evidence reveals that most PD cases follow the body-first subtype [10].
Despite all the efforts and contributions of the scientific community, the primary cause of sporadic PD remains largely unknown; however, it has been hypothesised that the disease has a multifactorial aetiology, resulting from the interaction between environmental, genetic, and age-associated factors [8]. Several environmental and lifestyle risk factors have been linked to PD, including environmental toxins, pesticide and heavy metals exposure, traumatic lesions, and bacterial or viral infections [11]. An interesting report from 2022 evaluated the correlation between some risk factors, including coffee consumption, cigarette smoking, and physical activity, and clinical symptoms [12]. Remarkably, coffee consumption is positively correlated with milder motor symptoms, whereas physical exercise is positively correlated with milder non-motor symptoms.
Moreover, while most cases occur in a sporadic manner, approximately 5–10% of PD cases are of genetic origin [11]. The era of PD genetics started when several families seemed to exhibit a Mendelian inheritance pattern (dominant or recessive) of Parkinson’s, suggesting a genetic cause of the disease [2]. In 1997, the first PD-associated mutation was discovered in the SNCA gene [13], originating a highly productive period of gene hunting that resulted in the identification of several PD-related genes, with either autosomal dominant (e.g., SNCA, LRRK2, VPS35) or autosomal recessive (e.g., PRKN, PINK1, DJ1) modes of inheritance [14]. Although genetic PD corresponds to a small fraction of all cases, focusing the research on genetic forms has proved to be valuable to understanding the pathophysiology of PD, since some of the proteins encoded by PD-associated genes are involved in key neuropathological mechanisms linked to the development and progression of both sporadic and familial PD [15]; these include α-synuclein aggregation and accumulation, mitochondrial dysfunction, proteasomal and autophagic impairment, oxidative stress, and neuroinflammation [2].
For several decades, numerous epidemiological studies have consistently shown that increased PD prevalence positively correlates with increasing age, highlighting aging as one of the most important contributing factors for PD. Indeed, the aging process is strongly associated with mitochondrial dysfunction, increased oxidative stress, neuroinflammation, and impairment of protein clearance (which facilitates α-synuclein accumulation even in the normal ageing brain [16]), all of which have also been considered pathological hallmarks of PD [4,17].
James Parkinson optimistically stated that “there appears to be sufficient reason for hoping that some remedial process may ere long be discovered, by which, at least, the progress of the disease may be stopped” [18]. Over 200 years later, no cure or neuroprotective therapy has been discovered for PD; nevertheless, researchers have been making huge progress in understanding neurodegeneration in PD, which will bring us closer to finding new effective disease-modifying therapies [8]. The currently available diagnostic methods are not able to predict the onset of the disease early on; therefore, the existing pharmacological and neurosurgical treatments are symptomatic, in order to ameliorate motor and non-motor symptoms and attempt to slow down the progression of the neurodegenerative process, but are not able to fully stop it [1].
The relationship between microRNAs (miRNAs) and PD has been intensively studied for several years, leading to the hypothesis that miRNAs might play an important role in the pathogenesis and progression of PD. Numerous miRNAs found to be dysregulated in PD were shown to target key molecular mechanisms linked to the pathogenesis and progression of PD, namely, α-synuclein aggregation and accumulation, mitochondrial dysfunction, oxidative stress, and inflammation. Herein, we will summarize evidence regarding mitochondrial dysfunction and inflammation involvement in PD and, in addition, will review the impact of miRNA dysregulation in these two important molecular mechanisms implicated in PD pathophysiology.
To carry out this state-of-the-art review, which aims for a comprehensive search of the current literature, we performed a methodical search of the literature using the database PubMed. Relevant articles were searched without year limitations. The search strategy consisted of a combination of the keywords “Parkinson’s disease” together with (AND) “mitochondrial dysfunction” (OR) “inflammation” (OR) “innate and adaptive immunity”; “Parkinson’s disease” together with (AND) “miRNA” (AND) “mitochondrial dysfunction” (OR) “inflammation” (OR) “innate and adaptive immunity” (OR) and (AND) “gut microbiota”. The present review includes an extensive collection and evaluation of the published studies (1245 papers), where 210 were used, and evaluations were independently performed by two authors (Guedes BFS and Esteves AR).

2. Mitochondrial Dysfunction in Parkinson’s Disease

Mitochondria are dynamic membrane-bound organelles known to play a critical role in multiple cellular functions, including ATP production, Ca2+ homeostasis, ROS generation, and programmed cell death [19]. The first evidence that mitochondrial dysfunction is a key player in PD pathogenesis dates back to 1983, when Langston and colleagues [20] reported several cases of a parkinsonian syndrome in humans caused by the use of a synthetic drug containing 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). These events rapidly received the attention of the scientific community, leading to a better understanding of how MPTP caused parkinsonism. The lipophilic nature of MPTP allows it to cross the blood–brain barrier and, once inside the brain, it is metabolised to 1-methyl-4-phenylpyridinium (MPP+), which is taken up by dopaminergic neurons through the dopamine transporter (DAT), inhibiting complex I activity from the mitochondrial electron transport chain (ETC) and resulting in energy depletion and oxidative stress [19]. Mitochondrial dysfunction associated with PD pathogenesis can result from ETC dysfunction, impaired mitochondrial biogenesis, increased oxidative stress, defective mitophagy, altered mitochondrial dynamics, compromised trafficking, and aberrant Ca2+ homeostasis [21]. The complex interplay between these compromised cellular functions leads to progressive cellular dysfunction that ultimately results in dopaminergic neurodegeneration that underlies PD pathogenesis.
The hypothesis that PD can be triggered by mitochondrial dysfunction [22] was validated when decreased activity of complex I was found in PD brain samples [23,24,25,26], platelets [27,28,29,30], lymphocytes [30], fibroblasts [31,32], skeletal muscle [33,34,35], and PD cytoplasmic hybrids (cybrids) [36]. Cybrids are created through transfer of platelet mitochondria from either PD or control subjects to mitochondrial DNA (mtDNA)-depleted recipient cells (rho0 cells). The end result are cybrid cell lines that express the nuclear genes of the recipient rho0 cell line and the mitochondrial genes of the platelet donor [37]. In addition to MPP+, other toxins and pesticides (like rotenone and paraquat) that impair mitochondrial complex I activity also cause parkinsonism symptomatology and dopaminergic neuron loss in animals [38]. Mitochondrial dysfunction may be potentiated by impaired mitochondrial biogenesis, a complex process essential for the maintenance of a healthy mitochondrial network [19]. Mitochondrial respiration, mitochondrial antioxidant defence, as well as mitochondrial biogenesis are regulated by PGC-1α (peroxisome proliferator-activated receptor-gamma coactivator-1alpha), which is significantly reduced in diverse PD models [39,40,41,42], therefore adversely affecting cellular bioenergetics. On the other hand, PGC-1α overexpression has been shown to alleviate α-synuclein oligomerization [39] and protect dopaminergic neurons [43].
The production of ROS through mitochondrial respiration is physiologic; however, disruption of respiratory chain complexes causes excessive production of ROS and leads to oxidative stress, which is detrimental to cells [44]. Remarkably, mitochondria are, simultaneously, the main source and the primary targets of ROS. Under normal conditions, antioxidant proteins like superoxide dismutase (SOD) and glutathione (GSH) prevent ROS levels from rising too high, but malfunction of these defence mechanisms leads to oxidative stress. Consequently, high ROS levels damage the components of the respiratory chain, as well as mtDNA. Complex I was found to be oxidatively damaged in PD brain biopsies, which compromises its appropriate assembly and function [25], leading to further inhibition and greater ROS production. In addition, post-mortem samples from PD patients displayed increased lipid peroxidation with glutathione pathway impairment [45]. ROS possess the ability to damage mtDNA by causing single- and double-strand breaks that, when repaired inefficiently, originate mtDNA deletion mutations that affect the genes coding for essential proteins involved in the mitochondrial respiratory chain [44]. The mitochondrial genome is packaged in nucleoids by the mitochondrial transcription factor A (TFAM), in order to protect it from oxidative insults; however, TFAM deficiency has been observed in dopaminergic neurons of sporadic PD subjects [46,47], suggesting an enhanced exposure of mtDNA to ROS damage in PD. Several studies have detected accumulation of deletions in the mtDNA of aged SNpc neurons of both healthy and PD individuals [46,48,49,50]. Additionally, mtDNA copy numbers have been found to increase with age in healthy individuals [50], allowing the neurons to adapt to mitochondrial dysfunction and maintain a healthy population of mitochondria. However, failure of such regulatory mechanisms of mtDNA copy numbers has been found in PD patients, leading to mtDNA copy number depletion [46,48,50,51], despite accumulating mutations. Evidence has shown that oxidative stress is linked to dopamine metabolism, justifying the selective neurodegeneration of dopaminergic neurons [52]. Auto-oxidation of dopamine generates free radicals and active quinones, which in turn interact with ROS scavengers, respiratory chain complexes, and proteins involved in mitophagy [52], contributing to increased levels of ROS and defective mitophagy processes. Indeed, autophagic alterations occur in PD, as demonstrated by an increased number of autophagosomes observed in post-mortem PD patient brains [53]. Moreover, substantia nigra neurons from PD patients display abnormal mitochondria within autophagosomes, indicating defects in mitophagy [54]. Mitophagy alterations were also observed in several PD models, including toxin-induced and genetic models [55,56]. Most interestingly, PD-associated gene mutations such as PINK1 and Parkin are known to be involved in this selective process of mitochondrial degradation [57]. Remarkably, α-synuclein is also a known target of autophagic degradation [58].
Oxidative damage has also been implicated in impaired proteasomal ubiquitination and degradation of proteins, facilitating the accumulation of aggregated α-synuclein in the form of LBs [59]. α-synuclein accumulation has been noted to induce redox imbalance and mitochondrial fragmentation in in vitro and in vivo models of PD [60,61]. Moreover, α-synuclein has been proven to build up inside mitochondria, damaging complex I of the ETC, resulting in mitochondrial dysfunction and oxidative stress [62,63,64].
Oxidative stress unquestionably contributes to PD pathology, and it is now acknowledged that ROS are a result of mitochondrial dysfunction and further contribute to exacerbating cell death [65]. The interaction between these numerous mechanisms associated with mitochondrial impairment forms a positive feedback loop that drives cellular dysfunction, resulting in the development and progression of PD.
Importantly, as previously mentioned, men have a higher risk of developing PD than women, and mitochondria seem to play a significant role in this fact. Interestingly, brain mitochondria from female mice and post-mortem samples have higher electron transport chain activity and lower oxidative stress [66]. A recent transcriptomic study observed that male PD patients show increases in oxidative stress, inflammation, and angiogenesis when compared to females [67].

3. Innate and Adaptive Immunity Activation in Parkinson’s Disease

The immune system is the result of the complex interplay between innate and adaptive immunity. The innate immune system, which is the first line of defence, is ancient, highly conserved, and nonspecific. It consists of tissue-resident macrophages, dendritic cells, monocytes, granulocytes, and neutrophils. Interestingly, in addition to immune cells, there is a plethora of non-immune cells capable of inducing innate immune responses, such as epithelial cells, epidermal keratinocytes, mesenchymal cells, stromal cells, and neurons [68]. A significant amount of evidence has demonstrated the expression of pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), on intestinal epithelial cells and neurons, suggesting that these non-immune cells possess adequate machinery to act against pathogens through the activation of innate immunity checkpoints [69]. Remarkably, the expression of TLR4 has been found to be altered in inflammatory bowel diseases patients [70]. Colonic epithelial cells isolated from these patients were shown to secrete Interleukin (IL)-1β, produced through activation of the inflammasome [69]. Likewise, it has been demonstrated that neurons express TLR3 and TLR4 [71,72] and also produce inflammatory cytokines, such as IL-6, tumour necrosis factor alpha (TNF-α), and interferon, that may mediate innate immunity in the absence of microglial cells [73]. Furthermore, evidence shows that neurons can activate NF-κB-dependent nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome under ischemic conditions without microglia contribution [74]. Moreover, work from our group demonstrated that both enriched mesencephalic and cortical neuronal cultures exposed to BMAA, a bacterial neurotoxin, activated the NLRP3 inflammasome, accompanied by the release of IL-1β, indicating that neurons can initiate this activation without microglia contribution [71,72].
Overall, the innate immune system provides a rapid line of defence against threats to the host, while adaptive immunity is highly specific and is able to recognize and memorize a specific pathogen and effectively produce an immune response against the previously encountered pathogen [75]. The innate and adaptive immune systems work very closely to produce effective and protective immune responses. However, under pathological conditions, the immune system’s unbalanced activation could lead to tissue damage [75]. Antigen-presenting cells (APCs), such as macrophages and dendritic cells, are the ones responsible for sensing danger and initiating inflammatory processes, upon activation of PRRs by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), and through antigen presentation to T cells. Microglia, the main tissue-resident macrophages, and therefore the predominant APCs of the brain, are the most abundant population of innate immune cells in the CNS, and during CNS inflammation they further contribute to the inflammatory process.
The involvement of the immune system in PD has been hypothesised since it was considered a multisystemic disorder, and evidence has grown significantly to support the hypothesis that (neuro)inflammation initiates and/or drives the progression of PD and that it occurs long before symptomatology becomes apparent. In 1988, McGeer and colleagues showed for the first time the presence of HLA-DR+ (a component of the human MHCII) reactive microglia in the post-mortem SNpc of PD patients [76]. Similarly, another group reported increased levels of activated microglia around dopaminergic neurons in MPTP-induced parkinsonism in post-mortem human brains [77]. Additionally, several groups demonstrated the existence of a pro-inflammatory phenotype or innate immunity activation of microglia through histological studies [78,79,80,81,82,83] and positron emission tomography (PET) imaging studies [84,85,86,87,88]. Extracellular α-synuclein can directly activate microglia early on, as shown in transgenic mice overexpressing wild-type or mutated α-synuclein [89,90]. Additional studies demonstrated that α-synuclein directly promotes microglia activation [91,92,93,94], inducing production and release of pro-inflammatory cytokines [91,92] and increasing expression of antioxidant enzymes [92]. Moreover, in addition to microglial activation, intranigral injection of α-synuclein also resulted in the upregulation of mRNA expression of major pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and the expression of endothelial markers of inflammation [94]. Furthermore, mitochondrial toxins, such as MPTP, 6-OHDA, and rotenone, have the ability to initiate an immune reaction in the striatum and SNpc, showing that specific damage to the ETC can trigger microglial activation and neuroinflammation [95,96,97,98]. Rotenone and 6-OHDA were depicted to activate microglia in mice [99], human microglial cell lines [100], and in rats [101,102,103].
Innate and adaptive immune responses are indeed activated in the CNS of PD patients and are known to cause upregulation of inflammatory responses. There is considerable evidence that inflammatory cytokines are secreted in the brain of PD subjects, as shown by increased levels of TNF-α in both the caudate and putamen [104]. Moreover, several studies performed on the cerebrospinal fluid (CSF) of PD patients revealed higher levels of IL-1β [105,106,107,108], IL-6 [105,106,107,109], and TNF-α [104,108,109]. Furthermore, higher levels of inflammatory cytokines are also found in the blood (the major pathway for immune cell circulation throughout the body) of PD patients, including IL-1β [110,111,112,113,114], IL-6 [110,112,115], TNF-α [112,116,117,118], and IFN-γ [117,118,119]. Together, these results validate the hypothesis that PD is indeed a multisystemic disorder.
As previously referred, α-synuclein pathology has also been found in the gut and in the vagus nerve. Chronic constipation, which is linked to peripheral inflammation [65], is one of the most frequent non-motor symptoms of PD. Enteric inflammation occurs in PD, and it has been demonstrated by increased levels of inflammation markers in the gastrointestinal tract, such as glial fibrillary acidic protein (GFAP) and pro-inflammatory cytokines, including IL-1β, IL-6, TNF-α, and IFN-γ, further reinforcing the role of peripheral inflammation in the initiation and/or progression of PD [120]. Remarkably, higher cytokine levels in the gut occur earlier during disease progression and decrease over time, possibly suggesting that gut inflammation could happen early during PD pathogenesis [121]. The occurrence of gastrointestinal dysfunction and gut inflammation in PD patients led to the investigation of variations in the gut microbiome of these individuals [121]. In fact, several studies have reported alterations in the gut microbiome of PD patients from multiple geographical populations [117,122,123,124,125,126,127]. For instance, Roseburia spp. and Faecalibacterium have been found to be decreased in PD stool. Roseburia spp. has been shown to have an anti-inflammatory role through (1) enhancement of intestinal barrier health [128], (2) downregulation of pro-inflammatory cytokines (e.g., IL-17) and differentiation of Treg cells [129], and (3) upregulation of anti-inflammatory cytokines (e.g., IL-10, TGFβ) [130]. Similarly, Faecalibacterium has been shown to reduce inflammation [131,132,133,134]. Together, these alterations indicate that the PD gut is more susceptible to inflammation. In general, the composition of gut microbiota is evidently altered in PD, and these findings are consistent with the hypothesis that gut dysbiosis is linked to an inflammatory environment that may contribute to the initiation and/or progression of PD pathology (Figure 2). The data presented suggest the crucial role of the immune system in both the pathogenesis and progression of PD, as several observations of aberrant innate and adaptive immune responses in the CNS, blood, and gut of PD patients link the immune system to PD risk.

4. The Interplay between Mitochondria and Innate Immunity

Upon microbial infection, the innate immune system is triggered through the recognition of PAMPs by PRRs expressed on the cells’ surface. Invading microorganisms and infected cells are then removed following an orchestrated pro-inflammatory immune response [65]. Nevertheless, PRRs have the ability to initiate innate immune responses independently of infection—a process known as sterile inflammation—through recognition of DAMPs, which are endogenous signals released upon injury or stress [135]. Mitochondria have been recognised as an important source of DAMPs; the reason behind this is the fact that mitochondria share a common origin with bacteria, and both display some similarities: circular DNA with CpG motifs, double-membrane structure with an abundance of cardiolipin in the inner membrane, secretion of N-formylated proteins, and reproduction by binary fission [135]. Upon injury or stress, the mitochondrial release of DAMPs activates the innate immune system, similarly to bacterial PAMPs, triggering sterile inflammation that mimics the response to infection [65,135].
Numerous studies have identified a critical role for mitochondria in regulating and activating the NLRP3 inflammasome [136], an intracellular immune sentinel activated upon changes in cellular homeostasis, which activates pro-inflammatory cytokines (IL-1β and IL-18) to trigger pyroptotic cell death [137,138]. Several studies demonstrate that mitochondrial dysfunction through ROS production is required for NLRP3 inflammasome activation [139,140]. Additional data have also revealed that mtDNA translocated to the cytosol can directly activate the NLRP3 inflammasome by binding to it [140,141]. Moreover, cardiolipin, a phospholipid found solely in inner mitochondrial and bacterial membranes [142,143], can also bind to and activate the NLRP3 inflammasome [144]. Mitochondria, along with mitochondrial DAMPs (mtDNA, ROS, and cardiolipin), are crucial for the activation and regulation of the NLRP3 inflammasome (Figure 3), which in turn incorporates mitochondrial dysfunction in a pro-inflammatory signalling response, thus elucidating the association of mitochondrial damage with inflammation [135]. Remarkably, exposure of neuronal and microglial cell lines to mitochondrial lysates led to an increase in inflammation markers, with mtDNA being proposed as the candidate DAMP, causing the inflammatory alterations detected [145]. Moreover, injecting mice with isolated mitochondria into the brain also resulted in an increase in inflammation markers, namely increased TNF-α, NF-κB phosphorylation, GFAP protein, and decreased Trem2 mRNA [146].
The cause of the neuronal loss observed in PD brains is still poorly understood. Accumulating evidence leads to the hypothesis that the activation of innate immunity in dopaminergic neurons, through the exposure to DAMPs originating from mitochondrial dysfunction, bacteria, or even their metabolites targeting the mitochondria, could promote low-grade inflammation [135]. The mitochondrial network was found to be highly fragmented in cellular and animal models of PD [147], which is a pre-requisite for the selective degradation by mitophagy [147,148]. It was demonstrated that cardiolipin exposure, as a result of mitochondrial fission, is an important intervenient in the disposal of dysfunctional mitochondria [149]. Because cardiolipin is only present in mitochondrial or bacterial membranes, it has been considered a mitochondrial-derived DAMP that is detected by the NLRP3 [150]. Hence, cardiolipin exposure increases due to mitochondrial dysfunction, leading to the activation of neuronal innate immunity.

5. miRNA Involvement in Parkinson’s Disease

5.1. Human and Animal Studies

Cells, either within the same tissue or in different tissues/organs, can communicate across a long distance by sending information from one cell to another to coordinate their behaviours in order to grow, develop and survive [151]. Recent studies proposed that miRNAs contribute to cell-to-cell communication, by being secreted and transported to other cells via circulation to affect recipient cells [151]. This hypothesis has been validated by the detection of extracellular/circulating miRNAs in a multitude of biological fluids, such as blood, CSF, saliva, breast milk, urine, and others [152]. miRNAs can be found circulating in vesicles (exosomes, microvesicles, apoptotic bodies) or associated with proteins (AGO2), and, contrary to cellular RNA, extracellular miRNAs present high stability, providing a desirable characteristic for long-distance cellular communication [153]. It is now accepted that extracellular/circulating miRNAs can be used as biomarkers and as a therapeutic approach for a wide range of diseases but are also important in cell-to-cell communication.
In the last few years, miRNA dysregulation has been implicated in several neurodegenerative diseases, including PD, where it contributes to neurodegeneration and disease progression [153].
The importance of miRNAs for CNS integrity has been demonstrated by inducing a selective depletion of Dicer in midbrain dopaminergic neurons in mice, which impairs miRNA biogenesis and results in neurodegeneration and locomotor symptoms mimicking PD [154]. Furthermore, a multitude of screening studies have reported differentially expressed miRNAs in the brain [155,156,157,158,159,160], CSF [161,162,163,164], and blood [165,166,167,168,169,170,171,172,173,174] of PD individuals. In PD, some miRNAs have been associated with mitochondrial dysfunction, neuroinflammation, and dopaminergic neuron demise, thereby worsening disease pathogenesis. Moreover, several studies have demonstrated that specific miRNAs regulate PD-related genes, such as SNCA, PRKN, DJ-1, and LRRK2, modulating their functions in different cellular and animal PD models [175]. It has been observed that overexpression of miR-494 significantly decreased the levels of DJ-1 both in vitro, in 3T3-L1 and Neuro-2a cell lines, rendering cells more susceptible to oxidative stress, and in vivo, in a MPTP mouse model, exacerbating MPTP-induced neurodegeneration [176]. On the other hand, miR-7 was shown to exert a protective role by repressing expression of α-synuclein and accelerating the clearance of α-synuclein and its aggregates through autophagy [177]. The reduced levels of miR-7 in the SNpc of a MPTP PD mouse model correlated to nigrostriatal neurodegeneration and α-synuclein upregulation [178]. A protective role for miR-153 in PD was also hypothesised, since its overexpression decreased MPP+-induced neurotoxicity in murine cortical neurons [179]. Kabaria and co-workers observed that miR-34b and miR-34c bind to SNCA mRNA and reduce α-synuclein expression. In contrast, downregulation of these miRNAs led to increased α-synuclein levels and formation of α-synuclein aggregates. The same group also detected a polymorphism in the binding site of miR-34b, interfering with its binding and consequently leading to α-synuclein overexpression [180]. Both miR-34b and miR-34c have been found to be downregulated in PD brains [160]. In vitro studies in differentiated SH-SY5Y cells concluded that depletion of miR-34 b/c resulted in cell death associated with mitochondrial dysfunction and oxidative stress [160]. LRRK2 gain-of-function mutations have been implicated in both genetic and sporadic PD [181,182]. Accordingly, LRRK2 levels were found to be increased in PD patients’ brains compared with healthy controls; however, no significant differences in LRRK2 transcripts were detected between both groups, suggesting a post-transcriptional modification of LRRK2 protein expression. In order to understand the reason behind this, Cho and colleagues proceeded to analyse the 3’UTR of LRRK2 and found a binding site for miR-205, a miRNA found to be downregulated in the brain of PD patients. Further studies confirmed that miR-205 upregulation in cell lines and primary neuron cultures resulted in the downregulation of LRRK2, possibly having a protective effect in the brains of PD patients [156]. Additionally, mutated LRRK2 seems to be able to negatively regulate miRNA-mediated translational repression in Drosophila melanogaster brains. Briefly, mutated LRRK2 inhibits the expression of two miRNAs (let-7 and miR-184) known to target E2F transcription factor 1 and dimerization partner transcription factor, leading to defective cell division and neuronal death. These results indicate that mutated LRRK2 might also play a key role in PD pathogenesis by modulating the miRNA pathway [183]. On the other hand, miRNAs can directly or indirectly modulate the expression of PD-related genes. In fact, a recent study found that miR-421 targets Pink1. Mice treated with MPTP and SH-SY5Y cells treated with MPP+ were shown to overexpress miR-421. Downregulation of miR-421 attenuated neurodegeneration in MPTP-treated mice and promoted mitophagy in MPP+-treated SH-SY5Y cells, highlighting the role of miR-421 in regulating mitophagy via the Pink1/Parkin pathway [184]. Moreover, Zeng and colleagues highlighted the protective role of miR-135b, since it inhibits pyroptosis by targeting FoxO1 in MPP+-treated SH-SY5Y and PC-12 cells [185]. A report from 2018 showed that miR-494-3p negatively regulates sirtuin 3 (SIRT3) expression in both MPP+-treated SH-SY5Y cells and in a MPTP-induced PD mouse model, worsening motor impairment of these mice [186]. Another study revealed that miR-486-3p targets sirtuin 2 (SIRT2) and reduces its expression levels. However, the authors reported a PD risk-conferring polymorphism in the SIRT2 gene and showed that a single nucleotide polymorphism (SNP) in this gene alters the binding efficiency of miR-486-3p to SIRT2, thereby increasing the expression level of SIRT2, which could increase α-synuclein aggregation and toxicity [187]. Furthermore, miRNAs have been shown to target several genes involved in neuroinflammation, a major hallmark of PD. Specifically, miR-155 was demonstrated to have an important role in the regulation of inflammation. The mentioned miRNA was found to be upregulated in a PD mouse model overexpressing α-synuclein. Deficiency of miR-155 prevented the increase in MHCII and the death of dopaminergic neurons triggered by α-synuclein overexpression. Oppositely, upregulation of miR-155 restored the inflammatory response to α-synuclein fibrils [188]. miR-7, in addition to having a role in the regulation of α-synuclein as previously described, emerged in the context of neuroinflammation as directly targeting the NLRP3 inflammasome gene. In fact, a study from 2016 determined that transfection of BV2 cells with miR-7 inhibited microglial NLRP3 inflammasome activation, while anti-miR-7 had the opposite effect, aggravating inflammasome activation in vitro. The same group reported that injecting miR-7 mimics directly into the striatum of a MPTP mouse model of PD suppressed NLRP3 inflammasome activation and ameliorated dopaminergic neuronal death [189]. Studies demonstrated the regulation of TNF-α levels by miR-7116-5p in a microglial cell culture model, where MPP+ potentiated TNF-α production by downregulating miR-7116-5p. Consistently, overexpressing miR-7116-5p in the microglia of an MPTP mouse model prevented the overproduction of TNF-α and the activation of glia, further reducing the loss of dopaminergic neurons [190]. Although research on this topic is still in its preliminary stages, and future studies are needed to better understand the role of miRNAs in neuroinflammation, these findings might help identify new therapeutic targets to downregulate microglial activation and potentially diminish dopaminergic neuron death in PD. Importantly, other non-coding RNAs may be involved in PD development progression. As an example, circular RNAs can function as miRNA sponges and act as competitive endogenous RNA to deregulate mRNA by miRNA. For instance, CircSLC8A1, which is increased in the SN of PD patients, regulates miR-128 function and/or activity affecting oxidative stress [191]. In addition, a recent study showed that transfer RNA fragment profiles, which were hypothesised to function as microRNAs, revealed disease-specific patterns in the CSF and blood of PD patients [192].

5.2. Gut Microbiota and microRNAs

Given the important involvement of gut dysbiosis and inflammation in PD and the potential of miRNAs to serve as diagnostic biomarkers, Kurz and colleagues [193] investigated the expression of miRNAs in routine colonic biopsies from PD patients and detected several differentially expressed miRNAs; amongst all, miR-486–5p upregulation showed the highest specificity for PD and correlated with age and disease severity in PD. In a follow-up analysis, 301 target genes of miR-486–5p were identified, as well as the biological processes affected by the mentioned miRNA, with brain development and post-synapse organization processes having the strongest functional association with the miR-486-5p target gene network [193].
The gut microbiota includes several microorganisms such as bacteria, viruses, protozoans, and fungi. Gut microbiota has a crucial role in the maintenance of gut homeostasis and integrity, since it interacts with the intestinal epithelial barrier (IEB) and intestinal epithelial cells [194]. Moreover, gut microbiota regulates intestinal epithelium growth, differentiation, and permeability [194]. Therefore, it comes as no surprise that alterations in gut microbiota composition can disrupt the IEB and lead to gut dysbiosis, with consequent immune and inflammatory response activation. Specific bacterial products (such as short-chain fatty acids (SCFAs), vitamins, or neurotransmitters) can interfere with the regulation of CNS immune and inflammatory processes, including microglial activity, by infiltrating into the bloodstream and traveling to the brain [195]. Additionally, they have the ability to directly activate circulating immune cells, which then travel to the CNS and regulate brain physiology [195]. Importantly, the gut–brain axis depends on a tightly regulated interplay between immunity and gut microbiota (Figure 2). As previously stated, it has been shown that gut microbiota is significantly altered in PD patients, compared with healthy controls; it is becoming evident that it might play an important role in the pathogenesis of PD. In addition, recent evidence indicates that miRNAs are involved in PD pathophysiology, suggesting a novel disease-associated mechanism that is now beginning to be explored and positing miRNAs as potential biomarkers for PD screening. Interestingly, miRNAs were found in human faecal samples, mainly derived from intestinal epithelial cells. Furthermore, it was recently demonstrated that secreted miRNAs are able to enter bacteria and regulate bacterial gene transcripts and affect their growth [196], giving rise to the possibility of the host’s miRNAs’ affecting and shaping their own gut microbiome in PD (Figure 4). This hypothesis was proposed after Fusobacterium nucleatum cultured with human miR-515-5p (a miRNA present in human faeces) showed an increased ratio of 16S rRNA/23S rRNA transcripts and altered growth [196]. Moreover, selective deletion of Dicer in mice promoted gut microbiota imbalance and exacerbation of dextran sulphate sodium (DSS)-induced colitis. These effects were reversed by faecal miRNA transplantation from wild-type littermates [196]. These studies further corroborate the hypothesis that miRNAs regulate gut microbiota and enhance the role of faecal miRNAs in influencing gut microbiota and preserving intestinal homeostasis. In order to better understand the potential interactions between miRNAs and the gut metagenome, Hewel and co-workers [197] performed an in silico target screen for binding sites of PD-associated miRNAs on human gut metagenome sequences, from which resulted a massive number of interactions. They found numerous miRNAs that may be key regulators in bacterial pathways relevant to PD, including the bacterial secretion system and lipopolysaccharide (LPS) biosynthesis [197]. On the other hand, gut microbiota, primarily through gut microbiota metabolites, may modulate human gene expression by affecting the host’s miRNA expression (Figure 4). In fact, Peck et al. demonstrated that the microbiota modulates miRNA expression in intestinal epithelium cells, which may alter intestinal homeostasis [198]. Different miRNA expression profiles were observed in the colon and ileum of germ-free mice colonised with gut microbiota from pathogen-free mice, when compared with germ-free littermates [199]. The analysis of faecal miRNA expression patterns also revealed significant differences between conventional mice and germ-free mice [200]. Additionally, it has also been demonstrated that depletion of gut microbiota using antibiotics induces alterations in faecal miRNA expression profiles in vivo [201]. A report from 2014 revealed that adherent-invasive E. coli, a pathogen highly prevalent in Crohn’s disease, upregulates miRNAs (miR-30c and miR-130a) that target genes involved in the autophagy response (ATG5 and ATG16L1) in mouse enterocytes, which may facilitate adherent-invasive E. coli replication and aggravation of intestinal inflammation [202]. Downregulation of miR-144 induced by Lactobacillus casei was reported to enhance intestinal barrier integrity through upregulation of occludin and zonula occludens 1 levels in intestinal epithelial cells, boosting intestinal barrier function and homeostasis [203]. Similarly, several probiotics have shown their ability to modulate miRNAs in intestinal epithelial cells and immune cells, altering intestinal barrier function and intestinal immune regulation [204,205,206]. A link between gut microbiota and brain miRNA expression has been established, since abnormal brain miRNA expression profiles have been described in the amygdala and prefrontal cortex of germ-free mice and mice treated with antibiotics to induce microbiota depletion [207]. Moreover, Bacteroides fragilis LPS, a microbial endotoxin, was shown to induce several miRNAs responsible for targeting genes that regulate synaptic architecture, amyloidogenesis, and brain inflammatory signalling [208]. These studies shed light on bidirectional communication between gut microbiota and miRNAs. Since both gut dysbiosis and differently expressed miRNAs have been reported in PD, the bidirectional interaction of gut microbiota–miRNAs might be involved in the pathophysiology of PD. We can hypothesize that microbial dysbiosis can potentiate the dysregulation of certain miRNAs, which can then target mitochondria and inflammatory pathways in the gut and later on in the brain, triggering PD pathological hallmarks (Figure 5).

6. Conclusions and Future Perspectives

The complexity and heterogeneity of PD have contributed to the challenging nature of PD diagnosis and treatment. The currently used diagnostic methods for PD are based on clinical symptoms that only emerge when the disease has already progressed to a stage of elevated neuronal loss. Therefore, there is a growing need to identify molecular biomarkers that allow an early and precise diagnosis. The possibility of using miRNAs as potential biomarkers for the diagnosis of PD during the prodromal phase gained relevance with the emergence of the hypothesis that miRNAs are involved in the pathogenesis and pathophysiology of PD. Several promising candidate miRNAs have surfaced from the numerous screenings performed so far on CSF, blood, and brain tissue of PD individuals or models. However, obtaining these samples is either invasive (CSF) or only possible post-mortem (brain tissue), making blood samples the ideal source of biomarkers, because they are easily extracted and analysed and facilitate patient monitoring over time. Moreover, considering the involvement of the gut in the aetiology and pathophysiology of PD, studying dysregulated miRNAs in the gut or even in the faecal material of PD patients might lead to the surfacing of an important and useful strategy for the early detection of gut-first PD and for the development of therapies able to slow or halt disease progression before it reaches the brain.
However, although microRNAs show potential to be reliable biomarkers, most of the candidate miRNAs are not organ-specific, and their profile is highly dependent on uncontrollable factors such as individual genetics and ethnicity, as reported in a study from 2020, where differences were observed between different ethnic cohorts of PD patients (American, Asian, and European) [209].
The potential of using miRNA-based therapies for the treatment of PD has also gained importance over the years. The scientific community has been developing strategies for miRNA modulation for many years, by using miRNA-mimics and antago-miRs to upregulate or downregulate miRNA levels, respectively. Nonetheless, the path to develop miRNA-based therapies has been paved with numerous challenges, including the lack of target specificity of miRNAs and their delivery to specific sites [210]. Unravelling relevant targets of miRNAs in PD models, followed by human validation of the results, may accelerate finding new biomarkers for an early diagnosis and novel therapeutic strategies for PD. To find a promising gut miRNA that can be a PD biomarker candidate would allow for therapeutic intervention in the prodromal phase of these patients, stopping PD progression and development. Hence, further studies of the microbiome–miRNA axis are a must in PD research.

Author Contributions

B.F.S.G. wrote the manuscript and performed the literature search and created the figures. A.R.E. drafted the manuscript. A.R.E. and S.M.C. critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the European Regional Development Fund (ERDF), through the COMPETE 2020–Operational Programme for Competitiveness and Internationalization, and by Portuguese national funds via FCT—Fundação para a Ciência e a Tecnologia under projects EXPL/MED-NEU/1515/2021, PTDC/MED-NEU/3644/2020, UIDB/04539/2020, UIDP/04539/2020 and LA/P/0058/2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  2. Jankovic, J.; Tan, E.K. Parkinson’s disease: Etiopathogenesis and treatment. J. Neurol. Neurosurg. Psychiatry 2020, 91, 795–808. [Google Scholar] [CrossRef]
  3. Mahlknecht, P.; Marini, K.; Werkmann, M.; Poewe, W.; Seppi, K. Prodromal Parkinson’s disease: Hype or hope for disease-modification trials? Transl. Neurodegener. 2022, 11, 11. [Google Scholar] [CrossRef]
  4. Cuenca, L.; Gil-Martinez, A.L.; Cano-Fernandez, L.; Sanchez-Rodrigo, C.; Estrada, C.; Fernandez-Villalba, E.; Herrero Ezquerro, M.T. Parkinson’s disease: A short story of 200 years. Histol. Histopathol. 2019, 34, 573–591. [Google Scholar] [CrossRef]
  5. Braak, H.; Del Tredici, K.; Rüb, U.; de Vos, R.A.; Steur, E.N.J.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
  6. Rietdijk, C.D.; Perez-Pardo, P.; Garssen, J.; Van Wezel, R.J.A.; Kraneveld, A.D. Exploring Braak’s Hypothesis of Parkinson’s Disease. Front. Neurol. 2017, 8, 37. [Google Scholar] [CrossRef]
  7. Chen, H.; Wang, K.; Scheperjans, F.; Killinger, B. Environmental triggers of Parkinson’s disease—Implications of the Braak and dual-hit hypotheses. Neurobiol. Dis. 2022, 163, 105601. [Google Scholar] [CrossRef]
  8. Simon, D.K.; Tanner, C.M.; Brundin, P. Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. Clin. Geriatr. Med. 2019, 36, 1–12. [Google Scholar] [CrossRef]
  9. Borghammer, P.; Horsager, J.; Andersen, K.; Berge, N.V.D.; Raunio, A.; Murayama, S.; Parkkinen, L.; Myllykangas, L. Neuropathological evidence of body-first vs. brain-first Lewy body disease. Neurobiol. Dis. 2021, 161, 105557. [Google Scholar] [CrossRef]
  10. Nuzum, N.D.; Loughman, A.; Szymlek-Gay, E.A.; Teo, W.-P.; Hendy, A.M.; Macpherson, H. To the Gut Microbiome and Beyond: The Brain-First or Body-First Hypothesis in Parkinson’s Disease. Front. Microbiol. 2022, 13, 791213. [Google Scholar] [CrossRef]
  11. Pajares, M.; Rojo, A.I.; Manda, G.; Boscá, L.; Cuadrado, A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic Implications. Cells 2020, 9, 1687. [Google Scholar] [CrossRef] [PubMed]
  12. Belvisi, D.; Pellicciari, R.; Fabbrini, A.; Costanzo, M.; Ressa, G.; Pietracupa, S.; De Lucia, M.; Modugno, N.; Magrinelli, F.; Dallocchio, C.; et al. Relationship between risk and protective factors and clinical features of Parkinson’s disease. Park. Relat. Disord. 2022, 98, 80–85. [Google Scholar] [CrossRef] [PubMed]
  13. Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; et al. Mutation in the α-Synuclein Gene Identified in Families with Parkinson’s Disease. Science 1997, 276, 2045–2047. [Google Scholar] [CrossRef] [PubMed]
  14. Jia, F.; Fellner, A.; Kumar, K.R. Monogenic Parkinson’s Disease: Genotype, Phenotype, Pathophysiology, and Genetic Testing. Genes 2022, 13, 471. [Google Scholar] [CrossRef]
  15. Guedes, L.C.; Mestre, T.; Outeiro, T.F.; Ferreira, J.J. Are genetic and idiopathic forms of Parkinson’s disease the same disease? J. Neurochem. 2020, 152, 515–522. [Google Scholar] [CrossRef] [PubMed]
  16. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef]
  17. Rodriguez, M.; Rodriguez-Sabate, C.; Morales, I.; Sanchez, A.; Sabate, M. Parkinson’s disease as a result of aging. Aging Cell 2015, 14, 293–308. [Google Scholar] [CrossRef]
  18. Parkinson, J. An Essay on the Shaking Palsy. J. Neuropsychiatry Clin. Neurosci. 1817, 14, 223–236, discussion 222. [Google Scholar] [CrossRef]
  19. Rani, L.; Mondal, A.C. Emerging concepts of mitochondrial dysfunction in Parkinson’s disease progression: Pathogenic and therapeutic implications. Mitochondrion 2020, 50, 25–34. [Google Scholar] [CrossRef]
  20. Langston, J.W.; Ballard, P.; Tetrud, J.W.; Irwin, I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science 1983, 219, 979–980. [Google Scholar] [CrossRef]
  21. Park, J.-S.; Davis, R.L.; Sue, C.M. Mitochondrial Dysfunction in Parkinson’s Disease: New Mechanistic Insights and Therapeutic Perspectives. Curr. Neurol. Neurosci. Rep. 2018, 18, 21. [Google Scholar] [CrossRef] [PubMed]
  22. Cardoso, S.M. The Mitochondrial Cascade Hypothesis for Parkinsons Disease. Curr. Pharm. Des. 2011, 17, 3390–3397. [Google Scholar] [CrossRef] [PubMed]
  23. Mizuno, Y.; Ohta, S.; Tanaka, M.; Takamiya, S.; Suzuki, K.; Sato, T.; Oya, H.; Ozawa, T.; Kagawa, Y. Deficiencies in Complex I subunits of the respiratory chain in Parkinson’s disease. Biochem. Biophys. Res. Commun. 1989, 163, 1450–1455. [Google Scholar] [CrossRef] [PubMed]
  24. Schapira, A.H.V.; Cooper, J.M.; Dexter, D.; Clark, J.B.; Jenner, P.; Marsden, C.D. Mitochondrial Complex I Deficiency in Parkinson’s Disease. J. Neurochem. 1990, 54, 823–827. [Google Scholar] [CrossRef]
  25. Keeney, P.M.; Xie, J.; Capaldi, R.A.; Bennett, J.P., Jr. Parkinson’s Disease Brain Mitochondrial Complex I Has Oxidatively Damaged Subunits and Is Functionally Impaired and Misassembled. J. Neurosci. 2006, 26, 5256–5264. [Google Scholar] [CrossRef]
  26. Parker, W.D., Jr.; Parks, J.K.; Swerdlow, R.H. Complex I Deficiency in Parkinson’s Disease Frontal Cortex. Brain Res. 2008, 1189, 215–218. [Google Scholar] [CrossRef]
  27. Benecke, R.; Strümper, P.; Weiss, H. Electron transfer complexes I and IV of platelets are abnormal in Parkinson’s disease but normal in Parkinson-plus syndromes. Brain 1993, 116 Pt 6, 1451–1463. [Google Scholar] [CrossRef]
  28. Parker, W.D., Jr.; Boyson, S.J.; Ba, J.K.P. Abnormalities of the electron transport chain in idiopathic parkinson’s disease. Ann. Neurol. 1989, 26, 719–723. [Google Scholar] [CrossRef]
  29. Krige, D.; Carroll, M.T.; Cooper, J.M.; Marsden, C.D.; Schapira, A.H.V.; The Royal Kings Queens Parkinson Disease Research Group. Platelet mitochondria function in Parkinson’s disease. Ann. Neurol. 1992, 32, 782–788. [Google Scholar] [CrossRef]
  30. Yoshino, H.; Nakagawa-Hattori, Y.; Kondo, T.; Mizuno, Y. Mitochondrial complex I and II activities of lymphocytes and platelets in Parkinson’s disease. J. Neural Transm. Parkinson’s Dis. Dement. Sect. 1992, 4, 27–34. [Google Scholar] [CrossRef]
  31. Mytilineou, C.; Werner, P.; Molinari, S.; Di Rocco, A.; Cohen, G.; Yahr, M.D. Impaired oxidative decarboxylation of pyruvate in fibroblasts from patients with Parkinson’s disease. J. Neural. Transm. Park Dis. Dement. Sect. 1994, 8, 223–228. [Google Scholar] [CrossRef]
  32. Winkler-Stuck, K.; Wiedemann, F.R.; Wallesch, C.-W.; Kunz, W.S. Effect of coenzyme Q10 on the mitochondrial function of skin fibroblasts from Parkinson patients. J. Neurol. Sci. 2004, 220, 41–48. [Google Scholar] [CrossRef] [PubMed]
  33. Bindoff, L.; Birch-Machin, M.; Cartlidge, N.; Parker, W.; Turnbull, D. Respiratory chain abnormalities in skeletal muscle from patients with Parkinson’s disease. J. Neurol. Sci. 1991, 104, 203–208. [Google Scholar] [CrossRef]
  34. Cardellach, F.; Martí, M.J.; Fernández-Solá, J.; Marín, C.; Hoek, J.B.; Tolosa, E.; Urbano-Márquez, A. Mitochondria1 respiratory chain activity in skeletal muscle from patients with Parkinson’s disease. Neurology 1993, 43, 2258. [Google Scholar] [CrossRef] [PubMed]
  35. Blin, O.; Desnuelle, C.; Rascol, O.; Borg, M.; Peyro Saint Paul, H.; Azulay, J.-P.; Billè, F.; Figarella, D.; Coulom, F.; Pellissier, J.F.; et al. Mitochondrial respiratory failure in skeletal muscle from patients with Parkinson’s disease and multiple system atrophy. J. Neurol. Sci. 1994, 125, 95–101. [Google Scholar] [CrossRef] [PubMed]
  36. Esteves, A.R.; Lu, J.; Rodova, M.; Onyango, I.; Lezi, E.; Dubinsky, R.; Lyons, K.E.; Pahwa, R.; Burns, J.M.; Cardoso, S.M.; et al. Mitochondrial respiration and respiration-associated proteins in cell lines created through Parkinson’s subject mitochondrial transfer. J. Neurochem. 2010, 113, 674–682. [Google Scholar] [CrossRef]
  37. Esteves, A.R.F.; Domingues, A.F.; Ferreira, I.; Januário, C.; Swerdlow, R.H.; Oliveira, C.; Cardoso, S.M. Mitochondrial function in Parkinson’s disease cybrids containing an nt2 neuron-like nuclear background. Mitochondrion 2008, 8, 219–228. [Google Scholar] [CrossRef] [PubMed]
  38. Tanner, C.M.; Kamel, F.; Ross, G.W.; Hoppin, J.A.; Goldman, S.M.; Korell, M.; Marras, C.; Bhudhikanok, G.S.; Kasten, M.; Chade, A.R.; et al. Rotenone, Paraquat, and Parkinson’s Disease. Environ. Health Perspect. 2011, 119, 866–872. [Google Scholar] [CrossRef]
  39. Eschbach, J.; von Einem, B.; Müller, K.; Bayer, H.; Scheffold, A.; Morrison, B.E.; Rudolph, K.L.; Thal, D.R.; Witting, A.; Weydt, P.; et al. Mutual exacerbation of peroxisome proliferator-activated receptor γ coactivator 1α deregulation and α-synuclein oligomerization. Ann. Neurol. 2015, 77, 15–32. [Google Scholar] [CrossRef]
  40. Shin, J.-H.; Ko, H.S.; Kang, H.; Lee, Y.; Lee, Y.-I.; Pletinkova, O.; Troconso, J.C.; Dawson, V.L.; Dawson, T.M. PARIS (ZNF746) Repression of PGC-1α Contributes to Neurodegeneration in Parkinson’s Disease. Cell 2011, 144, 689–702. [Google Scholar] [CrossRef]
  41. Zheng, B.; Liao, Z.; Locascio, J.J.; Lesniak, K.A.; Roderick, S.S.; Watt, M.L.; Eklund, A.C.; Zhang-James, Y.; Kim, P.D.; Hauser, M.A.; et al. PGC-1α, A Potential Therapeutic Target for Early Intervention in Parkinson’s Disease. Sci. Transl. Med. 2010, 2, 52ra73. [Google Scholar] [CrossRef]
  42. Jiang, H.; Kang, S.-U.; Zhang, S.; Karuppagounder, S.; Xu, J.; Lee, Y.-K.; Kang, B.-G.; Lee, Y.; Zhang, J.; Pletnikova, O.; et al. Adult Conditional Knockout of PGC-1α Leads to Loss of Dopamine Neurons. Eneuro 2016, 3, e0183-16. [Google Scholar] [CrossRef] [PubMed]
  43. Ciron, C.; Zheng, L.; Bobela, W.; Knott, G.W.; Leone, T.C.; Kelly, D.P.; Schneider, B.L. PGC-1α activity in nigral dopamine neurons determines vulnerability to α-synuclein. Acta Neuropathol. Commun. 2015, 3, 16. [Google Scholar] [CrossRef] [PubMed]
  44. Borsche, M.; Pereira, S.L.; Klein, C.; Grünewald, A. Mitochondria and Parkinson’s Disease: Clinical, Molecular, and Translational Aspects. J. Park. Dis. 2021, 11, 45–60. [Google Scholar] [CrossRef]
  45. Jenner, P.; Dexter, D.T.; Sian, J.; Schapira, A.H.; Marsden, C.D.; The Royal Kings and Queens Parkinson’s Disease Research Group. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental lewy body disease. Ann. Neurol. 1992, 32, S82–S87. [Google Scholar] [CrossRef]
  46. Grünewald, A.; Rygiel, K.A.; Hepplewhite, P.D.; Morris, C.M.; Picard, M.; Turnbull, D.M. MitochondrialDNADepletion in Respiratory Chain–DeficientParkinson Disease Neurons. Ann. Neurol. 2016, 79, 366–378. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, C.; Vincent, A.E.; Blain, A.P.; Smith, A.L.; Turnbull, D.M.; Reeve, A.K. Investigation of mitochondrial biogenesis defects in single substantia nigra neurons using post-mortem human tissues. Neurobiol. Dis. 2020, 134, 104631. [Google Scholar] [CrossRef]
  48. Bender, A.; Krishnan, K.J.; Morris, C.M.; Taylor, G.A.; Reeve, A.K.; Perry, R.H.; Jaros, E.; Hersheson, J.S.; Betts, J.; Klopstock, T.; et al. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat. Genet. 2006, 38, 515–517. [Google Scholar] [CrossRef]
  49. Reeve, A.K.; Krishnan, K.J.; Elson, J.L.; Morris, C.M.; Bender, A.; Lightowlers, R.N.; Turnbull, D.M. Nature of Mitochondrial DNA Deletions in Substantia Nigra Neurons. Am. J. Hum. Genet. 2008, 82, 228–235. [Google Scholar] [CrossRef]
  50. Dölle, C.; Flønes, I.; Nido, G.S.; Miletic, H.; Osuagwu, N.; Kristoffersen, S.; Lilleng, P.K.; Larsen, J.P.; Tysnes, O.-B.; Haugarvoll, K.; et al. Defective mitochondrial DNA homeostasis in the substantia nigra in Parkinson disease. Nat. Commun. 2016, 7, 13548. [Google Scholar] [CrossRef]
  51. Pyle, A.; Anugrha, H.; Kurzawa-Akanbi, M.; Yarnall, A.; Burn, D.; Hudson, G. Reduced mitochondrial DNA copy number is a biomarker of Parkinson’s disease. Neurobiol. Aging 2016, 38, 216.e7–216.e10. [Google Scholar] [CrossRef]
  52. Grünewald, A.; Kumar, K.R.; Sue, C.M. New insights into the complex role of mitochondria in Parkinson’s disease. Prog. Neurobiol. 2019, 177, 73–93. [Google Scholar] [CrossRef]
  53. Anglade, P.; Vyas, S.; Javoy-Agid, F.; Herrero, M.T.; Michel, P.P.; Marquez, J.; Mouatt-Prigent, A.; Ruberg, M.; Hirsch, E.C.; Agid, Y. Apoptosis and autophagy in nigral neurons of patients with Parkinson’s disease. Histol. Histopathol. 1997, 12, 25–31. [Google Scholar]
  54. Zhu, J.-H.; Guo, F.; Shelburne, J.; Watkins, S.; Chu, C.T. Localization of Phosphorylated ERK/MAP Kinases to Mitochondria and Autophagosomes in Lewy Body Diseases. Brain Pathol. 2003, 13, 473–481. [Google Scholar] [CrossRef]
  55. Zhu, J.-H.; Horbinski, C.; Guo, F.; Watkins, S.; Uchiyama, Y.; Chu, C.T. Regulation of Autophagy by Extracellular Signal-Regulated Protein Kinases During 1-Methyl-4-Phenylpyridinium-Induced Cell Death. Am. J. Pathol. 2007, 170, 75–86. [Google Scholar] [CrossRef]
  56. Cherra, S.J., III.; Steer, E.; Gusdon, A.M.; Kiselyov, K.; Chu, C.T. Mutant LRRK2 Elicits Calcium Imbalance and Depletion of Dendritic Mitochondria in Neurons. Am. J. Pathol. 2013, 182, 474–484. [Google Scholar] [CrossRef]
  57. Jin, S.M.; Youle, R.J. PINK1- and Parkin-mediated mitophagy at a glance. J. Cell Sci. 2012, 125 Pt 4, 795–799. [Google Scholar] [CrossRef]
  58. Martinez-Vicente, M.; Cuervo, A.M. Autophagy and neurodegeneration: When the cleaning crew goes on strike. Lancet Neurol. 2007, 6, 352–361. [Google Scholar] [CrossRef]
  59. Hattingen, E.; Magerkurth, J.; Pilatus, U.; Mozer, A.; Seifried, C.; Steinmetz, H.; Zanella, F.; Hilker, R. Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Brain 2009, 132 Pt 12, 3285–3297. [Google Scholar] [CrossRef]
  60. Bender, A.; Desplats, P.; Spencer, B.; Rockenstein, E.; Adame, A.; Elstner, M.; Laub, C.; Mueller, S.; Koob, A.O.; Mante, M.; et al. TOM40 Mediates Mitochondrial Dysfunction Induced by α-Synuclein Accumulation in Parkinson’s Disease. PLoS ONE 2013, 8, e62277. [Google Scholar] [CrossRef]
  61. Di Maio, R.; Barrett, P.J.; Hoffman, E.K.; Barrett, C.W.; Zharikov, A.; Borah, A.; Hu, X.; McCoy, J.; Chu, C.T.; Burton, E.A.; et al. α-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease. Sci. Transl. Med. 2016, 8, 342ra78. [Google Scholar] [CrossRef]
  62. Devi, L.; Raghavendran, V.; Prabhu, B.M.; Avadhani, N.G.; Anandatheerthavarada, H.K. Mitochondrial Import and Accumulation of alpha-Synuclein Impair Complex I in Human Dopaminergic Neuronal Cultures and Parkinson Disease Brain. J. Biol. Chem. 2008, 283, 9089–9100. [Google Scholar] [CrossRef]
  63. Liu, G.; Zhang, C.; Yin, J.; Li, X.; Cheng, F.; Li, Y.; Yang, H.; Uéda, K.; Chan, P.; Yu, S.; et al. α-Synuclein is differentially expressed in mitochondria from different rat brain regions and dose-dependently down-regulates complex I activity. Neurosci. Lett. 2009, 454, 187–192. [Google Scholar] [CrossRef]
  64. Luth, E.S.; Stavrovskaya, I.G.; Bartels, T.; Kristal, B.S.; Selkoe, D.J. Soluble, Prefibrillar α-Synuclein Oligomers Promote Complex I-dependent, Ca2+-induced Mitochondrial Dysfunction. J. Biol. Chem. 2014, 289, 21490–21507. [Google Scholar] [CrossRef]
  65. Esteves, A.R.; Silva, D.F.; Santos, D.; Candeias, E.; Filipe, F.; Cardoso, S.M. Mitochondria at the Base of Neuronal Innate Immunity in Alzheimer’s and Parkinson’s Diseases. In Mitochondrial Diseases; IntechOpen: Rijeka, Croatia, 2018. [Google Scholar]
  66. Cerri, S.; Mus, L.; Blandini, F. Parkinson’s Disease in Women and Men: What’s the Difference? J. Park. Dis. 2019, 9, 501–515. [Google Scholar] [CrossRef]
  67. López-Cerdán, A.; Andreu, Z.; Hidalgo, M.R.; Grillo-Risco, R.; Català-Senent, J.F.; Soler-Sáez, I.; Neva-Alejo, A.; Gordillo, F.; de la Iglesia-Vayá, M.; García-García, F. Unveiling sex-based differences in Parkinson’s disease: A comprehensive meta-analysis of transcriptomic studies. Biol. Sex Differ. 2022, 13, 68. [Google Scholar] [CrossRef]
  68. Dainichi, T.; Kabashima, K.; Ivanov, I.I.; Goto, Y. Editorial: Regulation of Immunity by Non-Immune Cells. Front. Immunol. 2021, 12, 770847. [Google Scholar] [CrossRef]
  69. Curciarello, R.; Canziani, K.E.; Docena, G.H.; Muglia, C.I. Contribution of Non-immune Cells to Activation and Modulation of the Intestinal Inflammation. Front. Immunol. 2019, 10, 647. [Google Scholar] [CrossRef]
  70. Cario, E. Toll-like receptors in inflammatory bowel diseases: A decade later. Inflamm. Bowel Dis. 2010, 16, 1583–1597. [Google Scholar] [CrossRef]
  71. Esteves, A.R.; Munoz-Pinto, M.F.; Nunes-Costa, D.; Candeias, E.; Silva, D.F.; Magalhães, J.D.; Pereira-Santos, A.R.; Ferreira, I.L.; Alarico, S.; Tiago, I.; et al. Footprints of a microbial toxin from the gut microbiome to mesencephalic mitochondria. Gut 2021, 72, 73–89. [Google Scholar] [CrossRef]
  72. Silva, D.F.; Candeias, E.; Esteves, A.R.; Magalhães, J.D.; Ferreira, I.L.; Nunes-Costa, D.; Rego, A.C.; Empadinhas, N.; Cardoso, S.M. Microbial BMAA elicits mitochondrial dysfunction, innate immunity activation, and Alzheimer’s disease features in cortical neurons. J. Neuroinflammation 2020, 17, 332. [Google Scholar] [CrossRef]
  73. Vijay, K. Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int. Immunopharmacol. 2018, 59, 391–412. [Google Scholar] [CrossRef] [PubMed]
  74. Fann, D.Y.-W.; Lim, Y.-A.; Cheng, Y.-L.; Lok, K.-Z.; Chunduri, P.; Baik, S.-H.; Drummond, G.R.; Dheen, S.T.; Sobey, C.G.; Jo, D.-G.; et al. Evidence that NF-κB and MAPK Signaling Promotes NLRP Inflammasome Activation in Neurons Following Ischemic Stroke. Mol. Neurobiol. 2018, 55, 1082–1096. [Google Scholar] [CrossRef] [PubMed]
  75. Sun, L.; Wang, X.; Saredy, J.; Yuan, Z.; Yang, X.; Wang, H. Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biol. 2020, 37, 101759. [Google Scholar] [CrossRef] [PubMed]
  76. McGeer, P.L.; Itagaki, S.; Boyes, B.E.; McGeer, E.G. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1988, 38, 1285–1291. [Google Scholar] [CrossRef]
  77. Langston, J.W.; Forno, L.S.; Tetrud, J.; Reeves, A.G.; Kaplan, J.A.; 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. Ann. Neurol. 1999, 46, 598–605. [Google Scholar] [CrossRef]
  78. Miklossy, J.; Doudet, D.; Schwab, C.; Yu, S.; McGeer, E.; McGeer, P. Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys. Exp. Neurol. 2006, 197, 275–283. [Google Scholar] [CrossRef]
  79. Croisier, E.; Moran, L.B.; Dexter, D.T.; Pearce, R.K.B.; Graeber, M.B. Microglial inflammation in the parkinsonian substantia nigra: Relationship to alpha-synuclein deposition. J. Neuroinflammation 2005, 2, 14. [Google Scholar] [CrossRef]
  80. Dzamko, N.; Gysbers, A.; Perera, G.; Bahar, A.; Shankar, A.; Gao, J.; Fu, Y.; Halliday, G.M. Toll-like receptor 2 is increased in neurons in Parkinson’s disease brain and may contribute to alpha-synuclein pathology. Acta Neuropathol. 2017, 133, 303–319. [Google Scholar] [CrossRef]
  81. 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 Neuropathol. 2003, 106, 518–526. [Google Scholar] [CrossRef]
  82. Orr, C.F.; Rowe, D.B.; Mizuno, Y.; Mori, H.; Halliday, G.M. A possible role for humoral immunity in the pathogenesis of Parkinson’s disease. Brain 2005, 128 Pt 11, 2665–2674. [Google Scholar] [CrossRef]
  83. Rozemuller, A.J.M.; Eikelenboom, P.; Theeuwes, J.W.; Steur, E.N.H.J.; de Vos, R.A.I. Activated microglial cells and complement factors are unrelated to cortical Lewy bodies. Acta Neuropathol. 2000, 100, 701–708. [Google Scholar] [CrossRef] [PubMed]
  84. 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. Ann. Neurol. 2005, 57, 168–175. [Google Scholar] [CrossRef] [PubMed]
  85. Gerhard, A.; Pavese, N.; Hotton, G.; Turkheimer, F.; Es, M.; Hammers, A.; Eggert, K.; Oertel, W.; Banati, R.B.; Brooks, D.J. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol. Dis. 2006, 21, 404–412. [Google Scholar] [CrossRef]
  86. Bartels, A.; Willemsen, A.; Doorduin, J.; de Vries, E.; Dierckx, R.; Leenders, K. [11C]-PK11195 PET: Quantification of neuroinflammation and a monitor of anti-inflammatory treatment in Parkinson’s disease? Park. Relat. Disord. 2010, 16, 57–59. [Google Scholar] [CrossRef] [PubMed]
  87. Iannaccone, S.; Cerami, C.; Alessio, M.; Garibotto, V.; Panzacchi, A.; Olivieri, S.; Gelsomino, G.; Moresco, R.; Perani, D. In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson’s disease. Park. Relat. Disord. 2013, 19, 47–52. [Google Scholar] [CrossRef]
  88. Terada, T.; Yokokura, M.; Yoshikawa, E.; Futatsubashi, M.; Kono, S.; Konishi, T.; Miyajima, H.; Hashizume, T.; Ouchi, Y. Extrastriatal spreading of microglial activation in Parkinson’s disease: A positron emission tomography study. Ann. Nucl. Med. 2016, 30, 579–587. [Google Scholar] [CrossRef]
  89. Su, X.; Maguire-Zeiss, K.A.; Giuliano, R.; Prifti, L.; Venkatesh, K.; Federoff, H.J. Synuclein activates microglia in a model of Parkinson’s disease. Neurobiol. Aging 2008, 29, 1690–1701. [Google Scholar] [CrossRef]
  90. Su, X.; Federoff, H.J.; Maguire-Zeiss, K.A. Mutant α-Synuclein Overexpression Mediates Early Proinflammatory Activity. Neurotox. Res. 2009, 16, 238–254. [Google Scholar] [CrossRef]
  91. Harms, A.; Cao, S.; Rowse, A.L.; Thome, A.; Li, X.; Mangieri, L.; Cron, R.; Shacka, J.J.; Raman, C.; Standaert, D.G. MHCII Is Required for -Synuclein-Induced Activation of Microglia, CD4 T Cell Proliferation, and Dopaminergic Neurodegeneration. J. Neurosci. 2013, 33, 9592–9600. [Google Scholar] [CrossRef]
  92. Béraud, D.; Hathaway, H.; Trecki, J.; Chasovskikh, S.; Johnson, D.A.; Johnson, J.A.; Federoff, H.J.; Shimoji, M.; Mhyre, T.R.; Maguire-Zeiss, K.A. Microglial Activation and Antioxidant Responses Induced by the Parkinson’s Disease Protein α-Synuclein. J. Neuroimmune Pharmacol. 2013, 8, 94–117. [Google Scholar] [CrossRef]
  93. Reynolds, A.D.; Glanzer, J.G.; Kadiu, I.; Ricardo-Dukelow, M.; Chaudhuri, A.; Ciborowski, P.; Cerny, R.; Gelman, B.; Thomas, M.P.; Mosley, R.L.; et al. Nitrated alpha-synuclein-activated microglial profiling for Parkinson’s disease. J. Neurochem. 2008, 104, 1504–1525. [Google Scholar] [CrossRef]
  94. Couch, Y.; Alvarez-Erviti, L.; Sibson, N.R.; Wood, M.J.; Anthony, D.C. The acute inflammatory response to intranigral α-synuclein differs significantly from intranigral lipopolysaccharide and is exacerbated by peripheral inflammation. J. Neuroinflammation 2011, 8, 166. [Google Scholar] [CrossRef] [PubMed]
  95. He, Y.; Appel, S.; Le, W. Minocycline inhibits microglial activation and protects nigral cells after 6-hydroxydopamine injection into mouse striatum. Brain Res. 2001, 909, 187–193. [Google Scholar] [CrossRef] [PubMed]
  96. Depino, A.M.; Earl, C.; Kaczmarczyk, E.; Ferrari, C.; Besedovsky, H.; Del Rey, A.; Pitossi, F.J.; Oertel, W.H. Microglial activation with atypical proinflammatory cytokine expression in a rat model of Parkinson’s disease. Eur. J. Neurosci. 2003, 18, 2731–2742. [Google Scholar] [CrossRef]
  97. McGeer, P.L.; 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. Ann. Neurol. 2003, 54, 599–604. [Google Scholar] [CrossRef]
  98. Barcia, C.; Bahillo, A.S.; Fernández-Villalba, E.; Bautista, V.; Poza, M.P.Y.; Fernández-Barreiro, A.; Hirsch, E.C.; Herrero, M.-T. Evidence of active microglia in substantia nigra pars compacta of parkinsonian monkeys 1 year after MPTP exposure. Glia 2004, 46, 402–409. [Google Scholar] [CrossRef]
  99. Sherer, T.B.; Betarbet, R.; Kim, J.-H.; Greenamyre, J. Selective microglial activation in the rat rotenone model of Parkinson’s disease. Neurosci. Lett. 2003, 341, 87–90. [Google Scholar] [CrossRef] [PubMed]
  100. Shaikh, S.B.; Nicholson, L.F. Effects of chronic low dose rotenone treatment on human microglial cells. Mol. Neurodegener. 2009, 4, 55. [Google Scholar] [CrossRef]
  101. Armentero, M.-T.; 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. Neurobiol. Dis. 2006, 24, 492–505. [Google Scholar] [CrossRef]
  102. Rodrigues, R.W.P.; Gomide, V.C.; Chadi, G. Astroglial and Microglial Reaction After A Partial Nigrostriatal Degeneration Induced by The Striatal Injection of Different Doses of 6-Hydroxydopamine. Int. J. Neurosci. 2001, 109, 91–126. [Google Scholar] [CrossRef]
  103. Rodrigues, R.W.P.; Gomide, V.C.; Chadi, G. Astroglial and microglial activation in the wistar rat ventral tegmental area after a single striatal injection of 6-hydroxydopamine. Int. J. Neurosci. 2004, 114, 197–216. [Google Scholar] [CrossRef]
  104. Mogi, M.; Harada, M.; Riederer, P.; Narabayashi, H.; Fujita, K.; Nagatsu, T. Tumor necrosis factor-α (TNF-α) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci. Lett. 1994, 165, 208–210. [Google Scholar] [CrossRef]
  105. Blum-Degena, D.; Müller, T.; Kuhn, W.; Gerlach, M.; Przuntek, H.; Riederer, P. Interleukin-1β and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci. Lett. 1995, 202, 17–20. [Google Scholar] [CrossRef]
  106. Mogi, M.; Harada, M.; Narabayashi, H.; Inagaki, H.; Minami, M.; Nagatsu, T. Interleukin (IL)-1β, IL-2, IL-4, IL-6 and transforming growth factor-α levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson’s disease. Neurosci. Lett. 1996, 211, 13–16. [Google Scholar] [CrossRef]
  107. Chen, X.; Hu, Y.; Cao, Z.; Liu, Q.; Cheng, Y. Cerebrospinal Fluid Inflammatory Cytokine Aberrations in Alzheimer’s Disease, Parkinson’s Disease and Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. Front. Immunol. 2018, 9, 2122. [Google Scholar] [CrossRef]
  108. Iwaoka, K.; Otsuka, C.; Maeda, T.; Yamahara, K.; Kato, K.; Takahashi, K.; Takahashi, K.; Terayama, Y. Impaired metabolism of kynurenine and its metabolites in CSF of parkinson’s disease. Neurosci. Lett. 2020, 714, 134576. [Google Scholar] [CrossRef]
  109. Schröder, J.B.; Pawlowski, M.; zu Horste, G.M.; Gross, C.; Wiendl, H.; Meuth, S.G.; Ruck, T.; Warnecke, T. Immune Cell Activation in the Cerebrospinal Fluid of Patients with Parkinson’s Disease. Front. Neurol. 2018, 9, 1081. [Google Scholar] [CrossRef]
  110. Kim, R.; Kim, H.-J.; Kim, A.; Jang, M.; Kim, A.; Kim, Y.; Yoo, D.; Im, J.H.; Choi, J.-H.; Jeon, B. Peripheral blood inflammatory markers in early Parkinson’s disease. J. Clin. Neurosci. 2018, 58, 30–33. [Google Scholar] [CrossRef]
  111. Alrafiah, A.; Al-Ofi, E.; Obaid, M.T.; Alsomali, N.; Muthuraju, S. Assessment of the Levels of Level of Biomarkers of Bone Matrix Glycoproteins and Inflammatory Cytokines from Saudi Parkinson Patients. BioMed Res. Int. 2019, 2019, 2690205. [Google Scholar] [CrossRef]
  112. Qin, X.-Y.; Zhang, S.-P.; Cao, C.; Loh, Y.P.; Cheng, Y. Aberrations in Peripheral Inflammatory Cytokine Levels in Parkinson Disease: A Systematic Review and Meta-analysis. JAMA Neurol. 2016, 73, 1316–1324. [Google Scholar] [CrossRef]
  113. Fan, Z.; Pan, Y.-T.; Zhang, Z.-Y.; Yang, H.; Yu, S.-Y.; Zheng, Y.; Ma, J.-H.; Wang, X.-M. Systemic activation of NLRP3 inflammasome and plasma α-synuclein levels are correlated with motor severity and progression in Parkinson’s disease. J. Neuroinflammation 2020, 17, 11. [Google Scholar] [CrossRef]
  114. Chatterjee, K.; Roy, A.; Banerjee, R.; Choudhury, S.; Mondal, B.; Halder, S.; Basu, P.; Shubham, S.; Dey, S.; Kumar, H. Inflammasome and α-synuclein in Parkinson’s disease: A cross-sectional study. J. Neuroimmunol. 2020, 338, 577089. [Google Scholar] [CrossRef]
  115. Kwiatek-Majkusiak, J.; Geremek, M.; Koziorowski, D.; Tomasiuk, R.; Szlufik, S.; Friedman, A. Serum levels of hepcidin and interleukin 6 in Parkinson’s disease. Acta Neurobiol. Exp. 2020, 80, 297–304. [Google Scholar] [CrossRef]
  116. Martin-Ruiz, C.; Williams-Gray, C.H.; Yarnall, A.J.; Boucher, J.J.; Lawson, R.A.; Wijeyekoon, R.S.; Barker, R.A.; Kolenda, C.; Parker, C.; Burn, D.J.; et al. Senescence and Inflammatory Markers for Predicting Clinical Progression in Parkinson’s Disease: The ICICLE-PD Study. J. Park. Dis. 2020, 10, 193–206. [Google Scholar] [CrossRef] [PubMed]
  117. Lin, C.-H.; Chen, C.-C.; Chiang, H.-L.; Liou, J.-M.; Chang, C.-M.; Lu, T.-P.; Chuang, E.Y.; Tai, Y.-C.; Cheng, C.; Lin, H.-Y.; et al. Altered gut microbiota and inflammatory cytokine responses in patients with Parkinson’s disease. J. Neuroinflammation 2019, 16, 129. [Google Scholar] [CrossRef]
  118. Eidson, L.N.; Kannarkat, G.T.; Barnum, C.J.; Chang, J.; Chung, J.; Caspell-Garcia, C.; Taylor, P.; Mollenhauer, B.; Schlossmacher, M.G.; Ereshefsky, L.; et al. Candidate inflammatory biomarkers display unique relationships with alpha-synuclein and correlate with measures of disease severity in subjects with Parkinson’s disease. J. Neuroinflammation 2017, 14, 164. [Google Scholar] [CrossRef] [PubMed]
  119. Rathnayake, D.; Chang, T.; Udagama, P. Selected serum cytokines and nitric oxide as potential multi-marker biosignature panels for Parkinson disease of varying durations: A case-control study. BMC Neurol. 2019, 19, 56. [Google Scholar] [CrossRef] [PubMed]
  120. Devos, D.; Lebouvier, T.; Lardeux, B.; Biraud, M.; Rouaud, T.; Pouclet, H.; Coron, E.; Bruley des Varannes, S.; Naveilhan, P.; Nguyen, J.-M.; et al. Colonic inflammation in Parkinson’s disease. Neurobiol. Dis. 2013, 50, 42–48. [Google Scholar] [CrossRef]
  121. Schonhoff, A.M.; Williams, G.P.; Wallen, Z.D.; Standaert, D.G.; Harms, A.S. Innate and adaptive immune responses in Parkinson’s disease. Prog. Brain Res. 2020, 252, 169–216. [Google Scholar] [CrossRef]
  122. Aho, V.T.E.; Pereira, P.A.B.; Voutilainen, S.; Paulin, L.; Pekkonen, E.; Auvinen, P.; Scheperjans, F. Gut microbiota in Parkinson’s disease: Temporal stability and relations to disease progression. EBioMedicine 2019, 44, 691–707. [Google Scholar] [CrossRef]
  123. Barichella, M.; Severgnini, M.; Cilia, R.; Cassani, E.; Bolliri, C.; Caronni, S.; Ferri, V.; Cancello, R.; Ceccarani, C.; Faierman, S.; et al. Unraveling gut microbiota in Parkinson’s disease and atypical parkinsonism. Mov. Disord. 2019, 34, 396–405. [Google Scholar] [CrossRef]
  124. Hill-Burns, E.M.; Debelius, J.W.; Morton, J.T.; Wissemann, W.T.; Lewis, M.R.; Wallen, Z.D.; Peddada, S.D.; Factor, S.A.; Molho, E.; Zabetian, C.P.; et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov. Disord. 2017, 32, 739–749. [Google Scholar] [CrossRef]
  125. Li, W.; Wu, X.; Hu, X.; Wang, T.; Liang, S.; Duan, Y.; Jin, F.; Qin, B. Structural changes of gut microbiota in Parkinson’s disease and its correlation with clinical features. Sci. China Life Sci. 2017, 60, 1223–1233. [Google Scholar] [CrossRef] [PubMed]
  126. Petrov, V.A.; Saltykova, I.V.; Zhukova, I.A.; Alifirova, V.M.; Zhukova, N.G.; Dorofeeva, Y.B.; Tyakht, A.V.; Kovarsky, B.A.; Alekseev, D.G.; Kostryukova, E.S.; et al. Analysis of Gut Microbiota in Patients with Parkinson’s Disease. Bull. Exp. Biol. Med. 2017, 162, 734–737. [Google Scholar] [CrossRef] [PubMed]
  127. Pietrucci, D.; Cerroni, R.; Unida, V.; Farcomeni, A.; Pierantozzi, M.; Mercuri, N.B.; Biocca, S.; Stefani, A.; Desideri, A. Dysbiosis of gut microbiota in a selected population of Parkinson’s patients. Park. Relat. Disord. 2019, 65, 124–130. [Google Scholar] [CrossRef] [PubMed]
  128. Tan, B.; Luo, W.; Shen, Z.; Xiao, M.; Wu, S.; Meng, X.; Wu, X.; Yang, Z.; Tian, L.; Wang, X. Roseburia intestinalis inhibits oncostatin M and maintains tight junction integrity in a murine model of acute experimental colitis. Scand. J. Gastroenterol. 2019, 54, 432–440. [Google Scholar] [CrossRef]
  129. Zhu, C.; Song, K.; Shen, Z.; Quan, Y.; Tan, B.; Luo, W.; Wu, S.; Tang, K.; Yang, Z.; Wang, X. Roseburia intestinalis inhibits interleukin-17 excretion and promotes regulatory T cells differentiation in colitis. Mol. Med. Rep. 2018, 17, 7567–7574. [Google Scholar] [CrossRef]
  130. Shen, Z.; Zhu, C.; Quan, Y.; Yang, J.; Yuan, W.; Yang, Z.; Wu, S.; Luo, W.; Tan, B.; Wang, X. Insights into Roseburia intestinalis which alleviates experimental colitis pathology by inducing anti-inflammatory responses. J. Gastroenterol. Hepatol. 2018, 33, 1751–1760. [Google Scholar] [CrossRef]
  131. Zhou, L.; Zhang, M.; Wang, Y.; Dorfman, R.G.; Liu, H.; Yu, T.; Chen, X.; Tang, D.; Xu, L.; Yin, Y.; et al. Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. Inflamm. Bowel Dis. 2018, 24, 1926–1940. [Google Scholar] [CrossRef]
  132. Martín, R.; Chain, F.; Miquel, S.; Lu, J.; Gratadoux, J.-J.; Sokol, H.; Verdu, E.F.; Bercik, P.; Bermúdez-Humarán, L.G.; Langella, P. The Commensal Bacterium Faecalibacterium prausnitzii Is Protective in DNBS-induced Chronic Moderate and Severe Colitis Models. Inflamm. Bowel Dis. 2014, 20, 417–430. [Google Scholar] [CrossRef] [PubMed]
  133. Breyner, N.M.; Michon, C.; de Sousa, C.S.; Boas, P.B.V.; Chain, F.; Azevedo, V.A.; Langella, P.; Chatel, J.M. Microbial Anti-Inflammatory Molecule (MAM) from Faecalibacterium prausnitzii Shows a Protective Effect on DNBS and DSS-Induced Colitis Model in Mice through Inhibition of NF-κB Pathway. Front. Microbiol. 2017, 8, 114. [Google Scholar] [CrossRef] [PubMed]
  134. Martín, R.; Miquel, S.; Chain, F.; Natividad, J.M.; Jury, J.; Lu, J.; Sokol, H.; Theodorou, V.; Bercik, P.; Verdu, E.F.; et al. Faecalibacterium prausnitzii prevents physiological damages in a chronic low-grade inflammation murine model. BMC Microbiol. 2015, 15, 67. [Google Scholar] [CrossRef] [PubMed]
  135. Cardoso, S.M.; Empadinhas, N. The Microbiome-Mitochondria Dance in Prodromal Parkinson’s Disease. Front. Physiol. 2018, 9, 471. [Google Scholar] [CrossRef] [PubMed]
  136. Gurung, P.; Lukens, J.R.; Kanneganti, T.-D. Mitochondria: Diversity in the regulation of the NLRP3 inflammasome. Trends Mol. Med. 2015, 21, 193–201. [Google Scholar] [CrossRef]
  137. Latz, E.; Xiao, T.S.; Stutz, A. Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 2013, 13, 397–411. [Google Scholar] [CrossRef]
  138. Schroder, K.; Tschopp, J. The Inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef] [PubMed]
  139. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef]
  140. Nakahira, K.; Haspel, J.A.; Rathinam, V.A.K.; Lee, S.-J.; Dolinay, T.; Lam, H.C.; Englert, J.A.; Rabinovitch, M.; Cernadas, M.; Kim, H.P.; et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 2011, 12, 222–230. [Google Scholar] [CrossRef]
  141. Shimada, K.; Crother, T.R.; Karlin, J.; Dagvadorj, J.; Chiba, N.; Chen, S.; Ramanujan, V.K.; Wolf, A.J.; Vergnes, L.; Ojcius, D.M.; et al. Oxidized Mitochondrial DNA Activates the NLRP3 Inflammasome during Apoptosis. Immunity 2012, 36, 401–414. [Google Scholar] [CrossRef] [PubMed]
  142. Rosa, I.D.A.; Einicker-Lamas, M.; Bernardo, R.R.; Previatto, L.M.; Borges, R.M.; Morgado-Díaz, J.A.; Benchimol, M. Cardiolipin in Hydrogenosomes: Evidence of Symbiotic Origin. Eukaryot. Cell 2006, 5, 784–787. [Google Scholar] [CrossRef] [PubMed]
  143. Kutschera, U.; Niklas, K.J. Endosymbiosis, cell evolution, and speciation. Theory Biosci. 2005, 124, 1–24. [Google Scholar] [CrossRef] [PubMed]
  144. Iyer, S.S.; He, Q.; Janczy, J.R.; Elliott, E.I.; Zhong, Z.; Olivier, A.K.; Sadler, J.J.; Knepper-Adrian, V.; Han, R.; Qiao, L.; et al. Mitochondrial Cardiolipin Is Required for Nlrp3 Inflammasome Activation. Immunity 2013, 39, 311–323. [Google Scholar] [CrossRef] [PubMed]
  145. Wilkins, H.M.; Carl, S.M.; Weber, S.G.; Ramanujan, S.A.; Festoff, B.W.; Linseman, D.A.; Swerdlow, R.H. Mitochondrial Lysates Induce Inflammation and Alzheimer’s Disease-Relevant Changes in Microglial and Neuronal Cells. J. Alzheimer’s Dis. 2015, 45, 305–318. [Google Scholar] [CrossRef]
  146. Wilkins, H.M.; Koppel, S.J.; Weidling, I.W.; Roy, N.; Ryan, L.N.; Stanford, J.A.; Swerdlow, R.H. Extracellular Mitochondria and Mitochondrial Components Act as Damage-Associated Molecular Pattern Molecules in the Mouse Brain. J. Neuroimmune Pharmacol. 2016, 11, 622–628. [Google Scholar] [CrossRef]
  147. Santos, M.C.J.; Esteves, A.R.; Silva, D.F.; Januário, C.; Cardoso, S.M. The Impact of Mitochondrial Fusion and Fission Modulation in Sporadic Parkinson’s Disease. Mol. Neurobiol. 2015, 52, 573–586. [Google Scholar] [CrossRef]
  148. Esteves, A.R.; Arduíno, D.M.; Silva, D.F.; Viana, S.D.; Pereira, F.C.; Cardoso, S.M. Mitochondrial Metabolism Regulates Microtubule Acetylome and Autophagy Trough Sirtuin-2: Impact for Parkinson’s Disease. Mol. Neurobiol. 2017, 55, 1440–1462. [Google Scholar] [CrossRef]
  149. Chu, T.C.; Ji, J.; Dagda, R.K.; Jiang, J.F.; Tyurina, Y.Y.; Kapralov, A.A.; Tyurin, V.A.; Yanamala, N.; Shrivastava, I.H.; Mohammadyani, D.; et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat. Cell Biol. 2013, 15, 1197–1205. [Google Scholar] [CrossRef]
  150. He, Y.; Hara, H.; Núñez, G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem. Sci. 2016, 41, 1012–1021. [Google Scholar] [CrossRef]
  151. Yao, S. MicroRNA biogenesis and their functions in regulating stem cell potency and differentiation. Biol. Proced. Online 2016, 18, 8. [Google Scholar] [CrossRef]
  152. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
  153. Nies, Y.H.; Najib, N.H.M.; Lim, W.L.; Kamaruzzaman, M.A.; Yahaya, M.F.; Teoh, S.L. MicroRNA Dysregulation in Parkinson’s Disease: A Narrative Review. Front. Neurosci. 2021, 15, 660379. [Google Scholar] [CrossRef] [PubMed]
  154. Schulz, J.; Takousis, P.; Wohlers, I.; Itua, I.O.; Dobricic, V.; Rücker, G.; Binder, H.; Middleton, L.; Ioannidis, J.P.; Perneczky, R.; et al. Meta-analyses identify differentially expressed microRNAs in Parkinson’s disease. Ann. Neurol. 2019, 85, 835–851. [Google Scholar] [CrossRef]
  155. Hoss, A.G.; Labadorf, A.; Beach, T.G.; Latourelle, J.C.; Myers, R.H. microRNA Profiles in Parkinson’s Disease Prefrontal Cortex. Front. Aging Neurosci. 2016, 8, 36. [Google Scholar] [CrossRef] [PubMed]
  156. Cho, H.J.; Liu, G.; Jin, S.M.; Parisiadou, L.; Xie, C.; Yu, J.; Sun, L.; Ma, B.; Ding, J.; Vancraenenbroeck, R.; et al. MicroRNA-205 regulates the expression of Parkinson’s disease-related leucine-rich repeat kinase 2 protein. Hum. Mol. Genet. 2013, 22, 608–620. [Google Scholar] [CrossRef] [PubMed]
  157. Briggs, C.E.; Wang, Y.; Kong, B.; Woo, T.-U.W.; Iyer, L.K.; Sonntag, K.C. Midbrain dopamine neurons in Parkinson׳s disease exhibit a dysregulated miRNA and target-gene network. Brain Res. 2015, 1618, 111–121. [Google Scholar] [CrossRef]
  158. Tatura, R.; Kraus, T.; Giese, A.; Arzberger, T.; Buchholz, M.; Höglinger, G.; Müller, U. Parkinson’s disease: SNCA-, PARK2-, and LRRK2- targeting microRNAs elevated in cingulate gyrus. Park. Relat. Disord. 2016, 33, 115–121. [Google Scholar] [CrossRef]
  159. Cardo, L.F.; Coto, E.; Ribacoba, R.; Menéndez, M.; Moris, G.; Suárez, E.; Alvarez, V. MiRNA Profile in the Substantia Nigra of Parkinson’s Disease and Healthy Subjects. J. Mol. Neurosci. 2014, 54, 830–836. [Google Scholar] [CrossRef]
  160. Miñones-Moyano, E.; Porta, S.; Escaramís, G.; Rabionet, R.; Iraola, S.; Kagerbauer, B.; Espinosa-Parrilla, Y.; Ferrer, I.; Estivill, X.; Martí, E. MicroRNA profiling of Parkinson’s disease brains identifies early downregulation of miR-34b/c which modulate mitochondrial function. Hum. Mol. Genet. 2011, 20, 3067–3078. [Google Scholar] [CrossRef]
  161. Burgos, K.; Malenica, I.; Metpally, R.; Courtright, A.; Rakela, B.; Beach, T.; Shill, H.; Adler, C.; Sabbagh, M.; Villa, S.; et al. Profiles of Extracellular miRNA in Cerebrospinal Fluid and Serum from Patients with Alzheimer’s and Parkinson’s Diseases Correlate with Disease Status and Features of Pathology. PLoS ONE 2014, 9, e94839. [Google Scholar] [CrossRef]
  162. Gui, Y.; Liu, H.; Zhang, L.; Lv, W.; Hu, X. Altered microRNA profiles in cerebrospinal fluid exosome in Parkinson disease and Alzheimer disease. Oncotarget 2015, 6, 37043–37053. [Google Scholar] [CrossRef] [PubMed]
  163. Marques, T.M.; Kuiperij, H.B.; Bruinsma, I.B.; van Rumund, A.; Aerts, M.B.; Esselink, R.A.J.; Bloem, B.R.; Verbeek, M.M. MicroRNAs in Cerebrospinal Fluid as Potential Biomarkers for Parkinson’s Disease and Multiple System Atrophy. Mol. Neurobiol. 2017, 54, 7736–7745. [Google Scholar] [CrossRef] [PubMed]
  164. Mo, M.; Xiao, Y.; Huang, S.; Cen, L.; Chen, X.; Zhang, L.; Luo, Q.; Li, S.; Yang, X.; Lin, X.; et al. MicroRNA expressing profiles in A53T mutant alpha-synuclein transgenic mice and Parkinsonian. Oncotarget 2017, 8, 15–28. [Google Scholar] [CrossRef] [PubMed]
  165. Oliveira, S.R.; Dionísio, P.A.; Guedes, L.C.; Gonçalves, N.; Coelho, M.; Rosa, M.M.; Amaral, J.D.; Ferreira, J.J.; Rodrigues, C.M.P. Circulating Inflammatory miRNAs Associated with Parkinson’s Disease Pathophysiology. Biomolecules 2020, 10, 945. [Google Scholar] [CrossRef]
  166. Margis, R.; Margis, R.; Rieder, C.R. Identification of blood microRNAs associated to Parkinsońs disease. J. Biotechnol. 2011, 152, 96–101. [Google Scholar] [CrossRef]
  167. Khoo, S.K.; Petillo, D.; Kang, U.J.; Resau, J.H.; Berryhill, B.; Linder, J.; Forsgren, L.; Neuman, L.A.; Tan, A.C. Plasma-Based Circulating MicroRNA Biomarkers for Parkinson’s Disease. J. Park. Dis. 2012, 2, 321–331. [Google Scholar] [CrossRef]
  168. Botta-Orfila, T.; Morató, X.; Compta, Y.; Lozano, J.J.; Falgàs, N.; Valldeoriola, F.; Pont-Sunyer, C.; Vilas, D.; Mengual, L.; Fernández, M.; et al. Identification of blood serum micro-RNAs associated with idiopathic andLRRK2Parkinson’s disease. J. Neurosci. Res. 2014, 92, 1071–1077. [Google Scholar] [CrossRef]
  169. Vallelunga, A.; Ragusa, M.; Di Mauro, S.; Iannitti, T.; Pilleri, M.; Biundo, R.; Weis, L.; Di Pietro, C.S.; De Iuliis, A.; Nicoletti, A.; et al. Identification of circulating microRNAs for the differential diagnosis of Parkinson’s disease and Multiple System Atrophy. Front. Cell. Neurosci. 2014, 8, 156. [Google Scholar] [CrossRef]
  170. Serafin, A.; Foco, L.; Zanigni, S.; Blankenburg, H.; Picard, A.; Zanon, A.; Giannini, G.; Pichler, I.; Facheris, M.F.; Cortelli, P.; et al. Overexpression of blood microRNAs 103a, 30b, and 29a in L-dopa-treated patients with PD. Neurology 2015, 84, 645–653. [Google Scholar] [CrossRef]
  171. Ding, H.; Huang, Z.; Chen, M.; Wang, C.; Chen, X.; Chen, J.; Zhang, J. Identification of a panel of five serum miRNAs as a biomarker for Parkinson’s disease. Park. Relat. Disord. 2016, 22, 68–73. [Google Scholar] [CrossRef]
  172. Cao, X.-Y.; Lu, J.-M.; Zhao, Z.-Q.; Li, M.-C.; Lu, T.; An, X.-S.; Xue, L.-J. MicroRNA biomarkers of Parkinson’s disease in serum exosome-like microvesicles. Neurosci. Lett. 2017, 644, 94–99. [Google Scholar] [CrossRef]
  173. Caggiu, E.; Paulus, K.; Mameli, G.; Arru, G.; Sechi, G.P.; Sechi, L.A. Differential expression of miRNA 155 and miRNA 146a in Parkinson’s disease patients. Eneurologicalsci 2018, 13, 1–4. [Google Scholar] [CrossRef]
  174. Chen, L.; Yang, J.; Lü, J.; Cao, S.; Zhao, Q.; Yu, Z. Identification of aberrant circulating miRNAs in Parkinson’s disease plasma samples. Brain Behav. 2018, 8, e00941. [Google Scholar] [CrossRef] [PubMed]
  175. Leggio, L.; Vivarelli, S.; L’episcopo, F.; Tirolo, C.; Caniglia, S.; Testa, N.; Marchetti, B.; Iraci, N. microRNAs in Parkinson’s Disease: From Pathogenesis to Novel Diagnostic and Therapeutic Approaches. Int. J. Mol. Sci. 2017, 18, 2698. [Google Scholar] [CrossRef] [PubMed]
  176. Xiong, R.; Wang, Z.; Zhao, Z.; Li, H.; Chen, W.; Zhang, B.; Wang, L.; Wu, L.; Li, W.; Ding, J.; et al. MicroRNA-494 reduces DJ-1 expression and exacerbates neurodegeneration. Neurobiol. Aging 2014, 35, 705–714. [Google Scholar] [CrossRef] [PubMed]
  177. Choi, D.C.; Yoo, M.; Kabaria, S.; Junn, E. MicroRNA-7 facilitates the degradation of alpha-synuclein and its aggregates by promoting autophagy. Neurosci. Lett. 2018, 678, 118–123. [Google Scholar] [CrossRef]
  178. Junn, E.; Lee, K.-W.; Jeong, B.S.; Chan, T.W.; Im, J.-Y.; Mouradian, M.M. Repression of alpha-synuclein expression and toxicity by microRNA-7. Proc. Natl. Acad. Sci. USA 2009, 106, 13052–13057. [Google Scholar] [CrossRef]
  179. Fragkouli, A.; Doxakis, E. miR-7 and miR-153 protect neurons against MPP+-induced cell death via upregulation of mTOR pathway. Front. Cell. Neurosci. 2014, 8, 182. [Google Scholar] [CrossRef]
  180. Kabaria, S.; Choi, D.C.; Chaudhuri, A.D.; Mouradian, M.M.; Junn, E. Inhibition of miR-34b and miR-34c enhances α-synuclein expression in Parkinson’s disease. FEBS Lett. 2015, 589, 319–325. [Google Scholar] [CrossRef]
  181. Li, J.-Q.; Tan, L.; Yu, J.-T. The role of the LRRK2 gene in Parkinsonism. Mol. Neurodegener. 2014, 9, 47. [Google Scholar] [CrossRef]
  182. Gilks, W.P.; Abou-Sleiman, P.M.; Gandhi, S.; Jain, S.; Singleton, A.; Lees, A.J.; Shaw, K.; Bhatia, K.P.; Bonifati, V.; Quinn, N.P.; et al. A common LRRK2 mutation in idiopathic Parkinson’s disease. Lancet 2005, 365, 415–416. [Google Scholar] [CrossRef] [PubMed]
  183. Gehrke, S.; Imai, Y.; Sokol, N.; Lu, B. Pathogenic LRRK2 negatively regulates microRNA-mediated translational repression. Nature 2010, 466, 637–641. [Google Scholar] [CrossRef] [PubMed]
  184. Dong, X.; He, X.; Yang, L.; Li, Q.; Xu, Y. Inhibition of miR-421 Preserves Mitochondrial Function and Protects against Parkinson’s Disease Pathogenesis via Pink1/Parkin-Dependent Mitophagy. Dis. Markers 2022, 2022, 5186252. [Google Scholar] [CrossRef]
  185. Zeng, R.; Luo, D.-X.; Li, H.-P.; Zhang, Q.-S.; Lei, S.-S.; Chen, J.-H. MicroRNA-135b alleviates MPP+-mediated Parkinson’s disease in in vitro model through suppressing FoxO1-induced NLRP3 inflammasome and pyroptosis. J. Clin. Neurosci. 2019, 65, 125–133. [Google Scholar] [CrossRef]
  186. Geng, L.; Zhang, T.; Liu, W.; Chen, Y. miR-494-3p modulates the progression of in vitro and in vivo Parkinson’s disease models by targeting SIRT3. Neurosci. Lett. 2018, 675, 23–30. [Google Scholar] [CrossRef] [PubMed]
  187. Wang, Y.; Cai, Y.; Huang, H.; Chen, X.; Chen, X.; Chen, X.; Mai, H.; Li, X.; Zhao, J.; Yang, J.; et al. miR-486-3p Influences the Neurotoxicity of a-Synuclein by Targeting the SIRT2 Gene and the Polymorphisms at Target Sites Contributing to Parkinson’s Disease. Cell. Physiol. Biochem. 2018, 51, 2732–2745. [Google Scholar] [CrossRef]
  188. Thome, A.D.; Harms, A.S.; Volpicelli-Daley, L.A.; Standaert, D.G. microRNA-155 Regulates Alpha-Synuclein-Induced Inflammatory Responses in Models of Parkinson Disease. J. Neurosci. 2016, 36, 2383–2390. [Google Scholar] [CrossRef]
  189. Zhou, Y.; Lu, M.; Du, R.-H.; Qiao, C.; Jiang, C.-Y.; Zhang, K.-Z.; Ding, J.-H.; Hu, G. MicroRNA-7 targets Nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson’s disease. Mol. Neurodegener. 2016, 11, 28. [Google Scholar] [CrossRef]
  190. He, Q.; Wang, Q.; Yuan, C.; Wang, Y. Downregulation of miR-7116-5p in microglia by MPP+sensitizes TNF-α production to induce dopaminergic neuron damage. Glia 2017, 65, 1251–1263. [Google Scholar] [CrossRef]
  191. Hanan, M.; Simchovitz, A.; Yayon, N.; Vaknine, S.; Cohen-Fultheim, R.; Karmon, M.; Madrer, N.; Rohrlich, T.M.; Maman, M.; Bennett, E.R.; et al. A Parkinson’s disease Circ RNA s Resource reveals a link between circ SLC 8A1 and oxidative stress. EMBO Mol. Med. 2020, 12, e11942. [Google Scholar] [CrossRef]
  192. Paldor, I.; Madrer, N.; Treidel, S.V.; Shulman, D.; Greenberg, D.S.; Soreq, H. Cerebrospinal fluid and blood profiles of transfer RNA fragments show age, sex, and Parkinson’s disease-related changes. J. Neurochem. 2023, 164, 671–683. [Google Scholar] [CrossRef]
  193. Kurz, A.; Kumar, R.; Northoff, B.H.; Wenk, C.; Schirra, J.; Donakonda, S.; Höglinger, G.U.; Schwarz, J.; Rozanski, V.; Hübner, R.; et al. Differential expression of gut miRNAs in idiopathic Parkinson’s disease. Park. Relat. Disord. 2021, 88, 46–50. [Google Scholar] [CrossRef]
  194. Pellegrini, C.; Antonioli, L.; Calderone, V.; Colucci, R.; Fornai, M.; Blandizzi, C. Microbiota-gut-brain axis in health and disease: Is NLRP3 inflammasome at the crossroads of microbiota-gut-brain communications? Prog. Neurobiol. 2020, 191, 101806. [Google Scholar] [CrossRef] [PubMed]
  195. Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [Google Scholar] [CrossRef] [PubMed]
  196. Liu, S.; da Cunha, A.P.; Rezende, R.M.; Cialic, R.; Wei, Z.; Bry, L.; Comstock, L.E.; Gandhi, R.; Weiner, H.L. The Host Shapes the Gut Microbiota via Fecal MicroRNA. Cell Host Microbe 2016, 19, 32–43. [Google Scholar] [CrossRef] [PubMed]
  197. Hewel, C.; Kaiser, J.; Wierczeiko, A.; Linke, J.; Reinhardt, C.; Endres, K.; Gerber, S. Common miRNA Patterns of Alzheimer’s Disease and Parkinson’s Disease and Their Putative Impact on Commensal Gut Microbiota. Front. Neurosci. 2019, 13, 113. [Google Scholar] [CrossRef]
  198. Peck, B.C.; Mah, A.T.; Pitman, W.A.; Ding, S.; Lund, P.K.; Sethupathy, P. Functional Transcriptomics in Diverse Intestinal Epithelial Cell Types Reveals Robust MicroRNA Sensitivity in Intestinal Stem Cells to Microbial Status. J. Biol. Chem. 2017, 292, 2586–2600. [Google Scholar] [CrossRef]
  199. Dalmasso, G.; Nguyen, H.T.T.; Yan, Y.; Laroui, H.; Charania, M.A.; Ayyadurai, S.; Sitaraman, S.V.; Merlin, D. Microbiota Modulate Host Gene Expression via MicroRNAs. PLoS ONE 2011, 6, e19293. [Google Scholar] [CrossRef]
  200. Viennois, E.; Chassaing, B.; Tahsin, A.; Pujada, A.; Wang, L.; Gewirtz, A.T.; Merlin, D. Host-derived fecal microRNAs can indicate gut microbiota healthiness and ability to induce inflammation. Theranostics 2019, 9, 4542–4557. [Google Scholar] [CrossRef]
  201. Moloney, G.; Viola, M.; Hoban, A.; Dinan, T.; Cryan, J. Faecal microRNAs: Indicators of imbalance at the host-microbe interface? Benef. Microbes 2018, 9, 175–183. [Google Scholar] [CrossRef]
  202. Nguyen, H.T.T.; Dalmasso, G.; Müller, S.; Carrière, J.; Seibold, F.; Darfeuille–Michaud, A. Crohn’s Disease–Associated Adherent Invasive Escherichia coli Modulate Levels of microRNAs in Intestinal Epithelial Cells to Reduce Autophagy. Gastroenterology 2014, 146, 508–519. [Google Scholar] [CrossRef] [PubMed]
  203. Hou, Q.; Huang, Y.; Wang, Y.; Liao, L.; Zhu, Z.; Zhang, W.; Liu, Y.; Li, P.; Chen, X.; Liu, F. Lactobacillus casei LC01 Regulates Intestinal Epithelial Permeability through miR-144 Targeting of OCLN and ZO1. J. Microbiol. Biotechnol. 2020, 30, 1480–1487. [Google Scholar] [CrossRef] [PubMed]
  204. Rodríguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Vezza, T.; Utrilla, M.P.; Chueca, N.; Garcia, F.; Olivares, M.; Rodríguez-Cabezas, M.E.; Gálvez, J. Differential intestinal anti-inflammatory effects of Lactobacillus fermentum and Lactobacillus salivarius in DSS mouse colitis: Impact on microRNAs expression and microbiota composition. Mol. Nutr. Food Res. 2017, 61, 1700144. [Google Scholar] [CrossRef] [PubMed]
  205. Kreuzer-Redmer, S.; Bekurtz, J.C.; Arends, D.; Bortfeldt, R.; Kutz-Lohroff, B.; Sharbati, S.; Einspanier, R.; Brockmann, G.A. Feeding of Enterococcus faecium NCIMB 10415 Leads to Intestinal miRNA-423-5p-Induced Regulation of Immune-Relevant Genes. Appl. Environ. Microbiol. 2016, 82, 2263–2269. [Google Scholar] [CrossRef]
  206. Sabharwal, H.; Cichon, C.; Ölschläger, T.A.; Sonnenborn, U.; Schmidt, M.A. Interleukin-8, CXCL1, and MicroRNA miR-146a Responses to Probiotic Escherichia coli Nissle 1917 and Enteropathogenic E. coli in Human Intestinal Epithelial T84 and Monocytic THP-1 Cells after Apical or Basolateral Infection. Infect. Immun. 2016, 84, 2482–2492. [Google Scholar] [CrossRef]
  207. Hoban, A.E.; Stilling, R.M.; Moloney, G.M.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F.; Clarke, G. Microbial regulation of microRNA expression in the amygdala and prefrontal cortex. Microbiome 2017, 5, 102. [Google Scholar] [CrossRef]
  208. Zhao, Y.; Lukiw, W.J. Bacteroidetes Neurotoxins and Inflammatory Neurodegeneration. Mol. Neurobiol. 2018, 55, 9100–9107. [Google Scholar] [CrossRef]
  209. Yadav, R.; Ramaswamy, P.; Pal, P.K.; Christopher, R. Clinical application of circulating micrornas in parkinson’s disease: The challenges and opportunities as diagnostic biomarker. Ann. Indian Acad. Neurol. 2020, 23, 84–97. [Google Scholar] [CrossRef]
  210. Goh, S.Y.; Chao, Y.X.; Dheen, S.T.; Tan, E.-K.; Tay, S.S.-W. Role of MicroRNAs in Parkinson’s Disease. Int. J. Mol. Sci. 2019, 20, 5649. [Google Scholar] [CrossRef]
Figure 1. Motor and non-motor symptoms associated with Parkinson’s disease. Diagnosis of Parkinson’s disease occurs with the onset of motor symptoms (early-stage Parkinson’s disease) but can be preceded by a prodromal phase of several years, which is characterised by specific non-motor symptoms (prodromal Parkinson’s disease). Created with Biorender.
Figure 1. Motor and non-motor symptoms associated with Parkinson’s disease. Diagnosis of Parkinson’s disease occurs with the onset of motor symptoms (early-stage Parkinson’s disease) but can be preceded by a prodromal phase of several years, which is characterised by specific non-motor symptoms (prodromal Parkinson’s disease). Created with Biorender.
Biomedicines 11 01349 g001
Figure 2. Dysregulation of the microbiota–gut–brain axis in the pathogenesis of Parkinson’s disease. Gut dysbiosis increases intestinal barrier permeability and leakage of bacterial products from the lumen to the lamina propria, activating inflammatory responses and leading to gut inflammation. These inflammatory processes induce misfolding of α-synuclein, which results in its aberrant aggregation and accumulation in the gut and enteric neurons. α-synuclein aggregates are then transported from the gut to the brain through the vagus nerve, eventually triggering an inflammatory response of the microglia. Alternatively, pro-inflammatory cytokines released during gut inflammation might infiltrate the bloodstream, leading to a systemic inflammation that further increases the permeability of the blood–brain barrier and allows the infiltration of immune cells into the brain. These processes result in neuroinflammation and consequent neurodegeneration. Created with Biorender.
Figure 2. Dysregulation of the microbiota–gut–brain axis in the pathogenesis of Parkinson’s disease. Gut dysbiosis increases intestinal barrier permeability and leakage of bacterial products from the lumen to the lamina propria, activating inflammatory responses and leading to gut inflammation. These inflammatory processes induce misfolding of α-synuclein, which results in its aberrant aggregation and accumulation in the gut and enteric neurons. α-synuclein aggregates are then transported from the gut to the brain through the vagus nerve, eventually triggering an inflammatory response of the microglia. Alternatively, pro-inflammatory cytokines released during gut inflammation might infiltrate the bloodstream, leading to a systemic inflammation that further increases the permeability of the blood–brain barrier and allows the infiltration of immune cells into the brain. These processes result in neuroinflammation and consequent neurodegeneration. Created with Biorender.
Biomedicines 11 01349 g002
Figure 3. NLRP3 inflammasome activation. The activation of the NLRP3 inflammasome is triggered by a number of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). PAMPs bind to Toll-like receptors (TLR) present on the cell membrane and upregulate the transcription of NLRP3 inflammasome components. DAMPs, such as ROS, mtDNA, or the externalization of cardiolipin to the outer mitochondrial membrane, also activate NLRP3. Assembly of the inflammasome activates caspase 1, which in turn cleaves pro-IL-1β and pro-IL-18 into IL-1β and IL-18, respectively. Created with Biorender.
Figure 3. NLRP3 inflammasome activation. The activation of the NLRP3 inflammasome is triggered by a number of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). PAMPs bind to Toll-like receptors (TLR) present on the cell membrane and upregulate the transcription of NLRP3 inflammasome components. DAMPs, such as ROS, mtDNA, or the externalization of cardiolipin to the outer mitochondrial membrane, also activate NLRP3. Assembly of the inflammasome activates caspase 1, which in turn cleaves pro-IL-1β and pro-IL-18 into IL-1β and IL-18, respectively. Created with Biorender.
Biomedicines 11 01349 g003
Figure 4. Reciprocal regulation of miRNAs and gut microbiota. Host’s intestinal epithelial cells release miRNAs that have the ability to regulate bacterial gene transcripts, affecting bacteria growth and replication. On the other hand, microbiota regulates host’s miRNA expression. Created with Biorender.
Figure 4. Reciprocal regulation of miRNAs and gut microbiota. Host’s intestinal epithelial cells release miRNAs that have the ability to regulate bacterial gene transcripts, affecting bacteria growth and replication. On the other hand, microbiota regulates host’s miRNA expression. Created with Biorender.
Biomedicines 11 01349 g004
Figure 5. miRNA–microbiota axis in PD. Microbial dysbiosis can potentiate the dysregulation of certain miRNAs, which can then target mitochondria and inflammatory pathways in the gut, leading to a proinflammatory response and to the loss of intestinal barrier integrity. This will allow miRNAs to travel freely or within vesicles through the blood or through the vagus nerve and reach the brain. Within the brain, miRNAs can target mitochondria and activate neuronal innate immunity, ultimately leading to PD hallmarks. Created with Biorender.
Figure 5. miRNA–microbiota axis in PD. Microbial dysbiosis can potentiate the dysregulation of certain miRNAs, which can then target mitochondria and inflammatory pathways in the gut, leading to a proinflammatory response and to the loss of intestinal barrier integrity. This will allow miRNAs to travel freely or within vesicles through the blood or through the vagus nerve and reach the brain. Within the brain, miRNAs can target mitochondria and activate neuronal innate immunity, ultimately leading to PD hallmarks. Created with Biorender.
Biomedicines 11 01349 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guedes, B.F.S.; Cardoso, S.M.; Esteves, A.R. The Impact of microRNAs on Mitochondrial Function and Immunity: Relevance to Parkinson’s Disease. Biomedicines 2023, 11, 1349. https://doi.org/10.3390/biomedicines11051349

AMA Style

Guedes BFS, Cardoso SM, Esteves AR. The Impact of microRNAs on Mitochondrial Function and Immunity: Relevance to Parkinson’s Disease. Biomedicines. 2023; 11(5):1349. https://doi.org/10.3390/biomedicines11051349

Chicago/Turabian Style

Guedes, Beatriz F. S., Sandra Morais Cardoso, and Ana Raquel Esteves. 2023. "The Impact of microRNAs on Mitochondrial Function and Immunity: Relevance to Parkinson’s Disease" Biomedicines 11, no. 5: 1349. https://doi.org/10.3390/biomedicines11051349

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop