Next Article in Journal
Hypertension Related to Obesity: Pathogenesis, Characteristics and Factors for Control
Next Article in Special Issue
What Are the Key Gut Microbiota Involved in Neurological Diseases? A Systematic Review
Previous Article in Journal
The Brain Protein Acylation System Responds to Seizures in the Rat Model of PTZ-Induced Epilepsy
Previous Article in Special Issue
Impact of Nutrition, Microbiota Transplant and Weight Loss Surgery on Dopaminergic Alterations in Parkinson’s Disease and Obesity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Interplay between Gut Microbiota and Parkinson’s Disease: Implications on Diagnosis and Treatment

by
Angelica Varesi
1,2,
Lucrezia Irene Maria Campagnoli
3,
Foroogh Fahmideh
3,
Elisa Pierella
4,
Marcello Romeo
1,
Giovanni Ricevuti
5,
Marchesi Nicoletta
3,
Salvatore Chirumbolo
6 and
Alessia Pascale
3,*
1
Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
2
Almo Collegio Borromeo, 27100 Pavia, Italy
3
Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
4
School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
5
Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
6
Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12289; https://doi.org/10.3390/ijms232012289
Submission received: 30 September 2022 / Revised: 5 October 2022 / Accepted: 12 October 2022 / Published: 14 October 2022
(This article belongs to the Special Issue Gut-Brain Axis of Neurodegenerative Disease)

Abstract

:
The bidirectional interaction between the gut microbiota (GM) and the Central Nervous System, the so-called gut microbiota brain axis (GMBA), deeply affects brain function and has an important impact on the development of neurodegenerative diseases. In Parkinson’s disease (PD), gastrointestinal symptoms often precede the onset of motor and non-motor manifestations, and alterations in the GM composition accompany disease pathogenesis. Several studies have been conducted to unravel the role of dysbiosis and intestinal permeability in PD onset and progression, but the therapeutic and diagnostic applications of GM modifying approaches remain to be fully elucidated. After a brief introduction on the involvement of GMBA in the disease, we present evidence for GM alterations and leaky gut in PD patients. According to these data, we then review the potential of GM-based signatures to serve as disease biomarkers and we highlight the emerging role of probiotics, prebiotics, antibiotics, dietary interventions, and fecal microbiota transplantation as supportive therapeutic approaches in PD. Finally, we analyze the mutual influence between commonly prescribed PD medications and gut-microbiota, and we offer insights on the involvement also of nasal and oral microbiota in PD pathology, thus providing a comprehensive and up-to-date overview on the role of microbial features in disease diagnosis and treatment.

1. Introduction

The chronic neurodegenerative pathology known as Parkinson’s disease (PD) can be described as an accumulation of a misfolded type of α-synuclein (the so-called Lewy bodies), an event occurring in dopaminergic neurons of the substantia nigra (SN), alongside with other related neuronal circuitries, which finally contribute both to non-motor (cognitive disorders, dysfunction in the olfactive sense, complications in the urogenital apparatus and in the gastrointestinal (GI) function) and mainly to motor symptoms (tremors, bradykinesia, abnormal gait and stiffness) [1,2,3,4]. The recent research suggests a relationship between gut microbiota (GM) and PD, due to the close interplay between the GM and the brain, known as “gut microbiota brain axis” (GMBA) [5], as recently reviewed in [6,7]. GMBA is usually described as a bidirectional functional system linking the enteric nervous system (ENS) in the gastrointestinal tract with the brain [5], but within the complex activity of our nervous system it might have a role much more puzzling and complicated than expected so far. The interplay of the GMBA with the immune system is particularly crucial [8,9]. A first, perhaps naïve, consideration is that a healthy composition of the GM warrants the integrity not only of the intestinal immune barrier, but also of the blood-brain barrier (BBB), via the regulation of the expression of fundamental proteins in the BBB such as tight junctions (i.e., claudin-5 and occludin), using the bacteria-produced short chain fatty acids (SCFAs) as modulatory signals for the brain synaptogenesis and development [10,11]. SCFAs are anti-inflammatory molecules with a key role in the GMBA [12] and some evidence reported that SCFAs-producing bacteria are reduced in PD [13]. The relationship between PD and GMBA has been also supported by the observation that α-synuclein is produced by the enteric neurons in the ENS, probably with functions related to uptake and neurotransmission [14]. Further, recent evidence has shown that a gut bacterial amyloid promotes the aggregation of α-synuclein, causing motor impairments in experimental mice [15]. The review by Mulak et al. addresses the issue of PD and GMBA as a particular item in the widest topic on the interplay between GM and brain in neurodegenerative disorders and indicated at least four levels of action [16]. The first level is represented by ENS, with neurons of sub-mucosal (Meissner’s) and myenteric (Auerbach’s) plexi, alongside the enteric glial cells [17]. The second level is represented by paravertebral ganglia, which modulate several visceral reflex responses at a peripheral level [18]. The third level is represented by the autonomous nervous system, while the fourth one is represented by the Central Nervous System (CNS) [16].
However, it seems that the current, more widely spread hypothesis about the etiopathogenetic role of GM in PD, is led by the ENS-derived α-synuclein (i.e., gut bacteria may induce the aggregation of α-synuclein in ENS), which, propagating in a prion-like manner, passes to the brain through the vagus nerve [15,19,20]. Notably, Desulfovibrio bacteria have been strictly associated with PD [21], where specifically hydrogen sulfide, which is also a known neuromodulator [22] produced by gut bacteria (Desulfovibrionaceae and Enterobacteriaceae families), has been associated with the etiopathogenesis of the disease [23].
The role of GMBA in PD may be considered, for certain aspects, a novel intriguing chapter in the complexity characterizing PD pathogenesis and evolution [24,25,26], for which the role of immunity (particularly T-reg cells) might be fundamental [27]. Nevertheless, further insights are needed to better disclose this captivating relationship.

2. Parkinson’s Disease and Gut Microbiota: Links and Mechanisms

2.1. Gut Microbiota Dysregulation in PD

Gut dysbiosis is defined as an imbalance of the intestinal flora due to an overgrowth of harmful taxa at the expense of beneficial commensal bacteria [28]. To date, this condition is known to participate in the pathophysiology of several GI and extraintestinal disorders, such as intestinal bowel syndrome, diabetes, obesity, chronic fatigue syndrome, autoimmune diseases, and several neuropsychiatric and neurologic disorders, including neurodegeneration [28,29,30,31,32,33,34,35,36,37,38,39]. Concerning PD, dysbiosis followed by GI symptoms and gut discomfort far precedes the onset of motor dysfunctions and it is linked to neuroinflammation, as well as to alterations in dopamine, serotonin, and kynurenine metabolism through the gut-brain axis [40,41,42,43,44,45,46] (Figure 1). Similar to what has been reported in Alzheimer’s disease (AD) and aging [30], feces from PD patients are enriched in opportunistic pathogens and pro-inflammatory taxa at the expense of anti-inflammatory microbes and SCFAs-producing bacteria, especially butyrate [47,48,49,50,51]. Accordingly, meta-analysis and systematic reviews report that PD patients are characterized by an overgrowth of the genera Bifidobacterium, Lactobacillus, Akkermansia and of the opportunistic pathogens Porphyromonas and Corynebacterium, together with a decreased abundance of the SCFAs producers Prevotellaceae, Lachnospiraceae and Faecalibacterium [52,53,54,55,56]. There is evidence that Faecalibacterium preserves the gut-barrier function through the production of the SCFA butyrate and the secretion of anti-inflammatory mediators [57,58]. Of note, lower levels of butyrate are linked to postural instability, gait disorders, Movement Disorder Society-Sponsored Revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) part III motor scores and depression among PD patients showing a reduced count of Faecalibacterium [59,60,61,62,63]. Other butyrate-producing taxa, such as Blautia, Caprococcus, Rosburia and Prevotella, also exert immunomodulatory activities and are reduced in PD patients compared to controls [50,60,64,65,66]. In line with these data, a study conducted in central China on 39 PD patients and their healthy spouses reported an inverse association between Prevotella abundance and disease severity, thus confirming the important role of inflammation in disease progression [64].
Lachnospiraceae are another family of bacteria underrepresented in PD-related gut microbiomes that contribute to the maintenance of intestinal homeostasis and prevent gut inflammation through the secretion of butyrate [67,68,69]. Their reduced presence, especially that of Ruminococcus, has been associated with cognitive impairment as measured by the Mini Mental State Examination (MMSE) test and postural instability in PD patients [59,68,69,70]. A similar phenotype was also associated to an increase in Lactobacillaceae, Enterobacteriaceae and Christensenellaceae, as reported by different studies [69,71,72]. Among Enterobacteriaceae, while Escherichia/Shigella have been negatively linked with disease duration in a case-control study involving 45 PD patients and their healthy spouses [73], Klebsiella showed a positive correlation in an independent report [64]. Concerning motor symptoms, it has been observed that the pro-inflammatory taxa Escherichia and Serratia are prevalent in non-tremor dominant patients [74], which present a more severe and faster progression of the disease compared to tremor dominant ones [75,76,77].
Akkermansia muciniphila is a bacterial genus belonging to the family of Verrucomicrobiaceae that is frequently found increased in patients with PD compared to controls [65,69,78,79,80,81]. Although Akkermansia muciniphila possesses the beneficial ability to convert mucin into SCFAs [82], its mucin degrading activity might also damage the gut-barrier triggering inflammation and promoting gut permeability [83,84]. Therefore, a high abundance of Akkermansia muciniphila may accelerate disease progression and favor α-synuclein (the already mentioned crucial protein involved in PD pathology) aggregation in gut enteroendocrine cells [85]. Phenotypically, studies show that increased levels of Akkermansia muciniphila are linked to stool consistency and constipation [86,87], but this association has yet to be validated in the context of PD [80].
Lactobacillus and Bifidobacterium are generally considered beneficial and often included in probiotic mixtures [88]. Their counterintuitive enrichment in stools from PD patients, reported in most studies [41,53,54,68,71,72,79] with few exceptions [78,89,90], could represent a compensatory mechanism to restore intestinal homeostasis [41].
There is evidence that GM composition changes according to ethnicity and geographic location [91,92]. Concerning PD, studies conducted on a population-specific cohort of patients shows the prominence of some geographic signatures, albeit on a similar background of bacterial dysbiosis [64,65,66,68,71,72,73,78]. For example, feces from Northern German PD patients showed increased abundance in the Barnesiellaceae family [72], while the genera Butyricicoccus and Clostridium XIVb were linked to a cognitive deficit in a Chinese PD cohort [73]. Among Chinese people, Parasutterella and Bilophila wadsworthia were more abundant in 39 PD patients compared to their healthy spouses [64], while the Northeastern Han population with PD showed reduced Bacteroides, as well as increased Ruminococcaceae and Lachnospiraceae NK4A [65]. These latter results, although in constrast with the previously described studies reporting lower levels of Lachnospiraceae and Ruminococcus among patients [59,69,81], could be explained as a population specific trait or may be a result of the small sample size. Furthermore, it has been reported that the Australian signature consists of decreased Colidextribacter, Agathobaculum, Kineothrix, Roseburia and Intestinibacter in favor of enriched Synergistetes and Proteobacteria, which elicit inflammation [50,66]. Lastly, feces from PD patients coming from various countries are enriched in Enterococcaceae [63,71,72], similar to what has been observed in AD [93].
GM and host metabolism are strictly interconnected [94,95]. Indeed, because of dysbiosis, lipid, amino acid and energy metabolism are often dysregulated in PD patients [96,97,98,99,100]. Studies show that a decrease in branched chain and aromatic amino acid biosynthesis, carbohydrate fermentation and butyrate synthesis is accompanied by an upregulation in lipopolysaccharides (LPS) production, bacterial type III secretion system biosynthesis (a complex bacterial structure involved in virulence) and proteolytic fermentation, which trigger inflammation and promote the release of harmful metabolites (i.e., phenylacetylglutamine and p-cresol) [50,101,102]. Furthermore, the pro-inflammatory taxa enriched in the gut of PD patients correlate with higher levels of plasma indole-3-propionic acid, hippuric acid (a carboxylic acid found in urine), as well as of the deoxycholic and glycodeoxycholic bile acids, which alter cholesterol metabolism [98,103]. Other dangerous molecules produced by the GM as a result of dysbiosis and associated to neurodegeneration are sphingolipids, trimethylamine N-oxide (TMAO) and the branched-chain fatty acid succinate that is linked to PD severity [61,104]. TMAO is an amine oxide product of the bacterial metabolism that has been implicated in oxidative stress enhancement, BBB disruption, mitochondria dysfunction, brain aging and neurodegeneration [105,106,107]. Often, increased levels of plasma TMAO are accompanied by a decreased production of several SCFAs required for the proper functioning of the ENS [61,104,108]. It has been shown that low levels of acetate, butyrate and above all propionate in feces from PD patients are linked to worse MDS-UPDRS part III scores [62,109], and the same is true for serum propionic acid [110]. Serum butyric acid and capronic acid are also downregulated in PD patients, while heptanoic acid is upregulated [110]. In plasma, Chen et al. report higher levels of acetate, propionate and butyrate in PD patients compared to controls, with plasma butyrate and valerate being inversely correlated with MMSE scores [62]. These conflicting results between stool and plasma could be due to increased intestinal permeability, a characteristic feature of PD patients (see Section 2.2 Gut permeability in PD) and which would facilitate the entry of SCFAs into the circulation [103]. In this respect, more research is needed to better clarify the relationship between blood and fecal levels of these molecules.
Given their important role, approaches aimed at restoring the optimal SCFAs levels have been proposed, with promising results [111]. These beneficial effects should be ascribed to the immunomodulatory and gut epithelial barrier-strengthening action of SCFAs, which together counteract inflammation [67]. Concerning PD, propionate supplementation was able to promote neurite outgrowth, tyrosine hydroxylase (TH) expression and dopaminergic cell survival in vitro [112]. in vivo, MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; a prodrug of the neurotoxin MPP+)-treated mice receiving propionate show a better performance in the stepping test, cylinder test and whisker test, in line with previous data [109,110]. Other investigations conducted with sodium butyrate have instead yielded conflicting results. Indeed, while sodium butyrate administration attenuated microglial activation and reduced cognitive deficits in preclinical models [113,114], other studies report increased dopaminergic neuronal toxicity, brain and colon inflammation, oxidative stress, and astrocyte activation both in vitro and in vivo [115,116]. Thus, unlike propionate, the contribution of sodium butyrate on neuroinflammation and gut inflammation remains unclear.

2.2. Gut Permeability in PD

The intestinal barrier is composed of a monolayer of epithelial cells that separate the GM present in the lumen from the internal tissues [117]. Under homeostatic conditions, the strict intercellular connections ensured by desmosomes, tight junctions and adherens junctions regulate the selective passage of nutrients, water and electrolytes while preventing the translocation of bacterial toxins, microorganisms and other harmful molecules [118]. However, disease-associated conditions such as dysbiosis, chronic intestinal inflammation and stress can damage the intestinal barrier integrity, allowing gut microbes to enter the circulation [33,119] (Figure 1). This condition, known as intestinal permeability or leaky gut, has been implicated in the pathophysiology of various GI, as well as extraintestinal disorders [120,121,122], including PD [117,123,124,125,126,127,128]. There is evidence that decreased levels of the colonic tight junction proteins occludin and zonula occludens-1 (ZO-1) trigger intestinal inflammation by activating the caspase-1 inflammasome signaling and increasing the levels of the pro-inflammatory interleukin 1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) [129,130,131]. Accordingly, higher concentrations of the leaky gut markers zonulin and α1-antitrypsin, as well as enhanced abundance of the colonic inflammatory markers calprotectin and lactoferrin, were noticed in feces from PD patients compared to controls [132,133,134]. These changes are often accompanied by variations in the concentration of stool SCFAs, with a reduction in fecal acetic, butyric, and propionic acids observed in PD patients [135] (see Section 2.1 Gut microbiota dysregulation in PD). Of note, MPTP mice treatment with propionate enhances the expression of ZO-1 and occludin via the Akt signaling, thus preserving the gut epithelial barrier integrity [109]. Other approaches useful to measure the extent of intestinal permeability and the degree of gut absorption are the lactulose/mannitol urinary test, the sucrose urinary assessment, the FITC (fluorescein)-dextran permeability test and the transepithelial resistance analysis [136,137]. In this respect, abnormal lactulose/mannitol ratio and sucrose concentration values found in subjects with PD have been associated with tight junction alterations [138,139]. Similar results were then replicated in 6-OHDA (hydroxydopamine)-treated rats (an animal model of PD), in which an increase in FITC-dextran permeability was observed as opposed to a decrease in transepithelial resistance, indicative of an impaired barrier functionality [140]. Of note, damage to the gut epithelial barrier alone without gut inflammation is not sufficient to explain disease severity and progression, as demonstrated in mice that overexpress human α -synuclein overexpressing (ASO mice) treated with dextran sodium sulfate, which is known to damage the mucosal epithelium [141]. These data demonstrate that a coordination of a series of complex, interconnected and related events is required to connect GI dysfunctions with PD symptoms.
Upon gut barrier breakdown, gram-negative endobacteria and their derived endotoxins, especially LPS, can enter the bloodstream, a condition known as metabolic endotoxemia [123,142]. Increased levels of LPS in the circulation are known to trigger systemic inflammation and to mediate BBB destruction, which allows microbial-derived inflammatory endotoxins to also enter the brain [123,129,142]. Moreover, LPS is known to interact with α-synuclein and to induce its nucleation [143], which will initiate a cascade of molecular events, leading to the formation of protein aggregates. Systemic administration of LPS and TNF-α to wild type adult mice is sufficient to stimulate the microglial expression of monocyte chemoattractant protein-1 (MCP-1), IL-1β, TNF-α and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and to enhance TH-positive neuronal loss in the SN, thus contributing to the onset of neurodegeneration [144]. Similarly, early motor symptoms are noticed in a mouse model of PD overexpressing the human α synuclein (also called Thy-1-α synuclein mouse) upon dietary intake of LPS [131]. In line with these data, serum from PD patients contains low levels of LPS binding protein [145,146], which are indicative of a chronic systemic exposure to LPS and gram negative endobacteria [147,148,149]. Accordingly, these changes were accompanied by increased positivity to gram-negative staining in the intestinal mucosa and higher levels of plasma TNF-α and interferon-γ [146,150]. Therapeutically, attempts to reduce systemic inflammation have been made by treating PD mice with analogues of the anti-inflammatory molecules cholecystokinin and glucagon-like peptide1. Results are promising, since reductions in tight junction leakage, colonic inflammation, gut α-synuclein aggregates and dopaminergic neural loss have been observed [151].
Although the leaky gut is becoming important for understanding PD pathophysiology, some conflicting evidence remains. In this regard, independent studies report no differences in serum IL-6, C-reactive protein, TNF-α, fecal zonulin, colonic ZO-1 and in the gut mucosal integrity marker serum diamine oxidase in samples obtained from PD patients compared to controls [130,134,145], and no changes in intestinal permeability were observed in mice treated with the PD-inducing compound rotenone, a well-known pesticide [152]. However, therapeutic options aimed at restoring dysbiosis and leaky gut, such as probiotics, prebiotics, fecal microbiota transplantation and dietary interventions appear to be promising and deserve further investigation.

3. Parkinson’s Disease and Gut Microbiota: Biomarkers and Drug Interactions

3.1. Gut Microbiota-Based PD Biomarkers

One important aspect of PD research is to find reliable, accurate, predictive, non-invasive, sensitive, and specific biomarkers for early disease detection and for following its course [153,154,155]. Since, as mentioned, GI symptoms often precede the onset of brain disorders, GM-based biomarkers have recently been considered as promising early and prodromal diagnostic tools in various neurological diseases, such as multiple sclerosis, AD and PD itself [30,156,157]. Concerning PD, it has been proposed that the severity of GI manifestations may be an early prediction of worse cognitive performances [158], and that the intestinal α-synuclein may function as a prodromal disease biomarker [159]. Moreover, GM composition might be used to monitor disease onset and progression [160]. In this respect, metagenomic sequencing, followed by random forest machine learning approaches, turned out to be the best method for new biomarker discovery in terms of precision and accuracy [161]. Accordingly, Qian et al. reported that a combination of 25 genetic microbial markers could effectively distinguish not only cases from controls but also differentially diagnose PD, AD and multiple system atrophy patients [157]. Among the bacterial features, a reduction of Prevotellaceae together with a rise in Akkermansia have been proposed as a possible PD diagnostic signature [79]. More recently, Guo et al. reported that the simultaneous measurement of Blautia levels in the feces, brain and blood can be used to differentiate cases from controls, as PD patients show reduced abundance of this genus [162]. Of note, improvements in predictive ability can be achieved by combining microbiome composition data with dietary information [163].
Besides looking for biomarkers based on the gut microbiota composition, other attempts have been made by measuring the abundance of GM-related molecules and/or SCFAs in the circulation [104]. For example, dementia development may be predicted by low levels of plasma TMAO measured at an early stage of the disease, thus serving as prognostic marker [164]. Moreover, disease severity has been associated with lower urine levels of urolithin, an anti-inflammatory molecule produced by the gut microbiota upon polyphenols intake [100]. When low, urolithin reflects a condition of dysbiosis characterized by an overgrowth of the pro-inflammatory Enterobacteriaceae at the expense of the beneficial Lachnospiraceae and Gordonibacter [100]. Another possible marker, namely blood LPS binding protein, has been shown to differentiate PD from controls without discriminating the disease stage; however the high variability measured among patients currently prevents its clinical application [147]. Concerning SCFAs, a study conducted by He et al. on 25 multiple system atrophy, 46 PD patients and 46 controls showed that lower plasma levels of acetic acid and propionic acid could discriminate the phenotypically similar multiple system atrophy and PD with 80% sensitivity and 91% specificity, thus enabling differential diagnosis among synucleinopathies [165].
Predicting and monitoring disease progression is of utmost importance to carry out personalized treatments [155]. So far, few studies have investigated the relationship between the GM and the rate of PD worsening, although with promising results. In 2021, Cilia et al. showed that a shortage of Roseburia in de novo PD patients was predictive of a faster deterioration in motor, non-motor, and intellectual ability within 3 years [160]. Similar results were then obtained by Lubomski et al. who reported that low levels of Barnesiella at the baseline and at 1-year follow-up were associated with a worse clinical evolution of PD [51]. Furthermore, a machine learning approach accounting for the abundance of various gut microbial species revealed that a reduction in Fusicatenibacter, Blautia and Faecalibacterium, together with an increased presence of Akkermansia, correlates with a faster progression of the disease [166]. Of note, patients without worsening symptoms over a one-year period (according to the Haehn&Yahr staging-scale and MPS-UPDRS-PartIII) showed stability in alpha and beta diversity, as well as gut microbiota composition, thus confirming the key role of gut microbiota in disease progression [167].
A summary of the main proposed GM-based PD biomarkers is shown in Figure 2.
Although conflicting evidence still exists [168] and data remain limited, the results of new clinical trials, such as the Dutch Parkinson Cohort (DUPARC) prospective cohort study, will be crucial to better define the possible role of GM as alternative PD biomarker for early diagnosis, differential discrimination, and progression monitoring [169]. Possibly, the use of several multimodal biomarkers in combination may strengthen the predictive capability by improving specificity and sensitivity [153,154].

3.2. Gut Microbiota-Drug Interactions

The mutual influence between GM and drug intake is well reported in the literature, and a growing body of evidence is emerging on the relationship between commonly prescribed PD drugs and the GM profile [170,171,172,173,174,175,176,177,178].
Levodopa (L-dopa) is a dopamine precursor and the leading compound for the treatment of PD [179]. To prevent its early conversion in dopamine before it reaches the brain, L-dopa is usually taken in combination with a dopa decarboxylase inhibitor, such as carbidopa [176,180]. However, carbidopa is not effective against the bacterial dopa decarboxylases [180], and this enables the GM to metabolize L-dopa, decreasing the drug availability while increasing the side effects [180,181]. Indeed, in the intestine, L-dopa is first transformed into dopamine by a dopa decarboxylase from E. faecalis and then converted into m-tyramine through the action of a dehydroxylase from Eggerthella lenta [181,182,183]. At the same time, C. sporogenes can also deaminate L-dopa, originating the metabolite 3-(3,4-dihydroxyphenyl) propionic acid, which is reported to impair ileal mobility [184]. Of note, considerable levels of this metabolite were reported in feces of L-dopa treated PD patients [184]. Finally, another bacterial species that is found with higher prevalence in PD patients compared to healthy subjects is Helicobacter pylori. Interestingly, the binding of L-dopa to H. pylori contributes to the reduced absorption of the drug, resulting in motor impairment [29].
In addition to being dependent on intestinal bacteria, L-dopa influences the GM composition itself. Accordingly, an increased relative abundance of Peptoniphilus, Finegoldia and Enterococcus, as well as a reduced presence of Faecalibacterium, Blautia and Lachnospirae were reported upon L-dopa exposure [185,186]. Treatment formulation may also change bacterial abundance. In this respect, patients receiving an L-dopa + carbidopa intestinal gel display higher levels of Enterobacteriaceae, Escherichia and Serratia compared to those receiving only L-dopa, while both groups show metabolic markers of gut inflammation [186]. In contrast, results from another study conducted on 19 PD patients before and after a 90-day long treatment with L-dopa reported no major differences in either α or β diversity between the two time points, suggesting that more research is needed to better clarify which factors are implicated in the L-dopa-mediated GM reshaping [187].
Catechol-o-methyl transferase (COMT) inhibitors, anticholinergics, monoaminoxidase inhibitors and dopamine agonists are additional PD drugs that are administered with or without L-dopa and may condition the dopaminergic balance [185]. Indeed, there is evidence that these medications have an impact on the abundance of the gut microbial dopa decarboxylases, thus potentially influencing dopamine metabolism [188]. Moreover, their intake has recently also been correlated with gut microbiota reshaping, although conflicting evidence remains [189,190]. For instance, treatment with dopamine agonists was associated with reduced intestinal motility and small intestinal bacteria overgrowth (SIBO; a kind of dysbiosis that frequently occurs in PD patients) in rats [191]. These effects were mediated by a greater relative abundance of Lactobacillus and Bifidobacterium, coupled with a decrease in Lachnospiraceae and Prevotellaceae [191]. In addition, COMT inhibitors and anticholinergics are known to induce GI side effects [192,193,194], which may be due to an imbalance of the intestinal flora towards harmful bacteria [190]. Further studies showed that certain gut microbial signatures, such as increased Bifidobacterium or Lactobacillaceae, are present in PD patients treated with COMT inhibitors [52,195,196,197]. In addition, a substantial decrease in Faecalibacterium prausnitzii abundance coupled with a trend towards lower fecal butyrate associate with entacapone, a widely prescribed COMT inhibitor [198], but not with the analogues opicapone and tolcapone (the latter one has been withdrawn from the market for safety reasons) [108,199]. Other studies confirmed the alteration of taxa related to GI disorders and constipation upon entacapone intake [185,200]. In particular, Fu et al. showed reduced Sellimonas, Lactobacillus, Faecalibacterium, Dorea, Intestinobacter and Blautia as well as augmented Eubacterium, Bifodobacterium and Christensenellacea_R-7_group in PD patients receiving entacapone combined with L-dopa versus those treated with L-dopa alone [200].
Finally, GM can also modify the action of some drugs useful within the context of PD [201,202]. For instance, the propionate produced by the GM appears necessary to mediate the benefits of the osteoblast-secreted protein osteocalcin, which improves PD motor and non-motor symptoms [203]. Further, upon oral berberine intake, the enterococcal TH enzyme synthetizes L-dopa, which is then converted to dopamine in the brain, thus improving cognitive symptoms [204]. Overall, although the relationship between GM and medications is intriguing, new research is needed to better delve into this mutual influence and to exploit it for therapeutic purposes.

4. Parkinson’s Disease and Gut Microbiota: Therapeutic Approaches

4.1. Gut Microbiota-Based PD Interventions: Antibiotics

Antibiotics are chemical compounds able to kill or arrest the growth of certain microorganisms. Although they are mainly used to counteract or prevent bacterial infections, their additional anti-inflammatory, immunomodulator, neuroprotective, antiamyloidogenic and antioxidant properties are becoming of increasing interest in the context of neurological disorders, including neurodegeneration [205,206,207,208,209]. Indeed, beside counteracting dysbiosis and constipation [210], it has been demonstrated that certain antibiotics can inhibit the activity of matrix metalloproteinases and prevent mitochondria dysfunction, microglia activation, protein misfolding and α-synuclein aggregation [211,212,213,214,215]. For example, treating mice where PD has been induced by MPTP with a cocktail of broad-spectrum antibiotics (ampicillin, metronidazole, and neomycin sulfate) was found to preserve TH and dopamine transporter immunoreactivities, which are generally lost upon MPTP administration [216]. This beneficial effect is mediated by an increase in Proteobacteria, as well as by a decrease in Deferribacteres and Saccharibacteria (TM7) abundance, which reflect an altered GM composition characterized by diversity loss [216]. Similar results were obtained in 6-OHDA-induced PD rats upon chronic treatment with an antibiotic mixture containing neomycin, pimaricin, bacitracin and vancomycin, which prevented dopaminergic neuronal death, relieved inflammation, ameliorated neurotoxicity and reduced motor impairments as measured by cylinder, rotation and stepping tests [217]. Recently, Cui et al. reported that vancomycin pretreatment of MPTP-induced PD mice improved motor symptoms by reducing SN astrocytes and microglia activation [218]. Notably, the authors proposed that neuroinflammation is indirectly inhibited by Akkermansia and Blautia, which increase in abundance upon vancomycin treatment and interfere with the toll like receptor 4 (TLR-4)/NF-κB pathway in the gut and in the brain [218]. Although Akkermansia is generally reported as harmful in PD patients, its dual negative and positive role may lean towards the latter when mucin conversion into SCFAs prevails over gut-barrier degradation, thus explaining this apparent discrepancy (see also Section 2.1 Gut microbiota dysregulation in PD). In humans, an intestinal decontamination therapy consisting of sodium phosphate enema, oral rifaximin and polyethylene glycol resulted effective in reducing dyskinesia and motor fluctuations related to PD, but more studies are required [219]. Other approaches focused on the use of certain specific antibiotics instead of cocktails have also been proposed to maximize the therapeutic benefit without impacting beneficial bacteria.
Rifaximin is a broad spectrum antibiotic with poor systemic absorption indicated to treat SIBO [210,220,221]. In this respect, rifaximin-mediated SIBO eradication in PD patients resulted in reduced motor fluctuations without impacting on L-dopa treatment [222]. This benefit should be ascribed to rifaximin-mediated modulation of the brain thyrotropin releasing hormone (THR) and THR-like peptides, which have caloric-restriction-like, anti-aging, neuroprotective properties and are known to be involved in the gut-brain axis [223]. However, no improvement in GI symptoms in 8 PD patients treated with rifaximin poses controversy over the actual efficacy of this antibiotic as PD treatment, calling for new studies [224].
Ceftriaxone (CTX) is a β-lactam antibiotic with a strong and safe past record [225,226]. The treatment of several PD animal models with CTX is known to improve neuroinflammatory and oxidative stress markers, stimulate neurogenesis and promote astrocyte viability through the suppression of NF-κB/c-Jun-mediated signaling [225,226,227,228]. Mechanistically, CTX also reduces extracellular glutamate levels by increasing the expression of the glutamate transporter-1 in astrocytes, thus avoiding brain excitotoxicity [226,228,229]. Moreover, it has been observed that CTX binds to α-synuclein with considerable affinity and prevents its polymerization in vitro [226,230,231]. in vivo, there is evidence that CTX treatment modifies the GM composition of MPTP-induced PD mice by disadvantaging the growth of Proteus while increasing the relative abundance of Akkermansia species, which act as probiotics when their SCFAs-converting activity exceeds that of intestinal barrier degradation (see also Section 2.1 Gut microbiota dysregulation in PD) [232].
Further studies proved the ability of CTX to reduce the levels of the main pro-inflammatory mediators TLR-4, MyD88 (myeloid differentiation primary response 88), IL-1β, TNF-α and NF-κB in the brain, TLR-4, MyD88, and NF-κB in the colon and IL-1β, TNF-α and IL-6 in the serum [232,233,234]. Similarly, CTX-mediated increase in the main antioxidant modulators glutathione, superoxide dismutase (SOD) and catalase was found to prevent the oxidative damage observed in rats treated with MPTP [233,234]. In line with these data, CTX administration was associated with reduced glial fibrillary acid protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA-1) expression, two markers of astrogliosis and microglia activation, respectively [232,235,236,237]. At the neuronal level, pre- or post- treatment with CTX prevented the loss of TH-positive neurons, reduced glutamatergic hyperactivity, and promoted neurogenesis at the level of SN and hippocampal dentate gyrus in different rat models of the disease [233,237,238,239,240,241,242]. As a consequence, dyskinesia, motor impairment and memory loss were all reverted upon CTX administration [233,234,237,238,239,240,242,243,244], although conflicting evidence still remains about its ability to improve learning outcomes [245]. Of note, CTX has been shown to interact synergistically with other compounds currently used or under investigation for the treatment of PD, such as erythropoietin, ropinirole and memantine, but the safety as well as the efficacy of these combinations should be further assessed [233,234,246].
Minocycline is a second-generation semisynthetic tetracycline with anti-microbial, anti-apoptotic, anti-inflammatory and antioxidant properties [247,248,249,250]. Thanks to the ability to efficiently cross the BBB, minocycline is considered neuroprotective for a variety of neurological conditions, including PD [251,252,253,254,255,256,257]. This effect is mainly ascribable to the minocycline-dependent suppression of microglia activation, which has been reported by several in vivo studies [251,258,259,260,261,262,263]. In this respect, microglial inactivation by minocycline correlates with decreased IL-1β formation, as well as reduced NADPH-oxidase and inducible nitric oxide synthase (iNOS) activity, suggesting that both anti-inflammatory and antioxidant pathways are involved [262,264]. In vitro, minocycline addition to 6-OHDA treated PC12 cells suppresses the release of lactate dehydrogenase, reactive oxygen species (ROS) and caspase 3 while supporting the activity of the antioxidant enzymes SOD and catalase [251,265,266,267]. Of note, these molecular changes seem to explain the increased striatal dopamine levels as well as the cognitive and locomotor improvements observed in zebrafish, mouse, and rat models [259,260,264,268,269,270,271]. Another mechanism through which minocycline prevents apoptosis is by limiting mitochondria dysfunction, inhibiting caspase 1 and 3 expression, and preventing the degradation of the antiapoptotic protein ICAD (the inhibitor of the caspase-activated deoxyribonuclease) [251,272,273,274]. However, despite the promising results, controversy remains. Indeed, an enhanced toxicity has been reported upon minocycline administration to MPTP-treated rodents and primates, resulting in disease exacerbation [275,276]. Moreover, results from a phase II clinical trial show no benefit from the use of minocycline and evidence decreased tolerability, although more studies are needed before drawing premature conclusions [277,278].
Doxycycline (DOX) is another broad-spectrum antibiotic belonging to tetracyclines that has been considered as PD treatment [279]. In vitro, DOX has shown anti-inflammatory properties by interfering with p38 MAP kinase and NF-κB pathways, reducing the expression of the activated microglia marker IBA-1 and inhibiting the production of the pro-oxidant and pro-inflammatory factors ROS, nitric oxide, iNOS, cyclooxygenase-2 (COX-2), IL-1β and TNF-α [280,281,282]. Concerning neuroprotection, DOX exerts an anti-apoptotic activity by repressing the matrix metallopeptidase-3 (MMP-3) in dopaminergic neurons and microglia both in vitro and in vivo [281]. In addition, DOX stimulates neurite growth through the activation of PI3K/Akt and MAPK/ERK pathways, independently from nerve growth factor activity [283]. Of note, recent studies demonstrated that DOX reduces the size and load of α-synuclein oligomers by converting them into high-molecular weight species that are not able to form fibrils, thus increasing cell viability [282,284]. When tested in vivo, DOX confirmed its neuroprotective activity by limiting dopaminergic neuronal loss in SN while increasing striatal dopamine levels [285,286]. This beneficial function is achieved by contrasting glial reactivity and by reducing the major histocompatibility complex-II expression in microglial cells [285,286]. In 6-OHDA-treated rats, both DOX and its derivative COL-3 showed an anti-dyskinetic potential when administered in combination with L-dopa [287]. According to the authors, the reduced levels of MMP-2/-9, MMP-3, ROS and of the dyskinesia-linked immunoreactivity markers FOSB, COX-2, GFAP and OX-42 would explain these benefits [287]. Nevertheless, despite promising in vivo data, clinical evidence is still lacking.
Rifampicin is a macrocyclic antibiotic with cytoprotective functions that have been considered for PD treatment [288,289]. Indeed, there is evidence that rifampicin prevents α-synuclein fibrillation by promoting SUMOylation, which increases fibril solubility preventing neuronal death [290,291,292]. Other studies reported a reduction in IL-1β, TNF-α, IL-6 and ROS released by cells double treated with rotenone and rifampicin, thus indicating a promotion of neuroprotection [293,294,295]. Although not completely defined, rifampicin appears to sustain cell viability through different mechanisms: (i) by enhancing autophagy [293,295]; (ii) via PI3K/Akt/GSK-3β/CREB pathway modulation [296]; (iii) by upregulating the unfolded protein response marker GRP78 through the PERK/eIF2α/ATF4 pathway [297]. In vivo, MPTP-induced PD mice treated with rifampicin showed increased striatal and SN TH immunoreactivity, attenuated levels of oxidative stress and re-established dopaminergic signaling in the striatum [298]. More recently, rotenone-induced PD in zebrafish has shown benefit from rifampicin administration due to the decrease in neuroinflammation [299].
Generally, although promising, two main concerns remain about the use of antibiotics in PD treatment: (i) antibiotics can kill some specific microbial populations leading to intestinal dysbiosis and neurological dysfunction and (ii) their prolonged and widespread intake would favor antibiotic resistance [213,300,301]. There is evidence that ceftriaxone (a third-generation cephalosporin)-induced dysbiosis worsens motor symptoms in 6-OHDA treated mice and correlates with dopaminergic neuron toxicity as well as intestinal and systemic inflammation [302]. Moreover, quinolones and β-lactams are known to trigger neurotoxicity through their interference with gamma-aminobutyric acid and benzodiazepine receptors signaling [213]. Mechanistically, it has been hypothesized that antibiotic-induced dysbiosis may favor the growth of Enterobacteria producing the bacterial α-synuclein curli, which promotes neurodegeneration [303,304]. In addition, leaky gut-mediated systemic inflammation might result from dysbiosis and mediate the BBB damage, allowing circulating neurotoxins to enter the brain [305]. In humans, a Finnish study conducted on 13,976 PD and 40,697 healthy individuals showed that taking certain antibiotics years earlier, especially macrolides and lincosamides, correlates with an increased risk of developing PD [306]. However, results from another prospective study involving 59,637 women did not report any correlation between antibiotic intake and PD incidence [307]. Overall, contrasting results and scarce long-term safety data remain a concern. Innovative drug delivery systems based on nanoparticles are now being tested to improve the clinical benefit of these antibiotics [279]. At the same time, synthetic tailoring to potentiate the neuroprotective chemical functions over the antimicrobial ones is another promising approach for the risk-benefit optimization [301].

4.2. Gut Microbiota-Based PD Interventions: Probiotics

Probiotics are defined as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [308]. It is widely reported that the most common bacteria used as probiotics (Lactobacilli, Bifidobacteria, and Enterococci) [309] have potential benefits in restoring the GM, reducing intestinal permeability, inflammation, and oxidative stress, improving immune homeostasis and GI symptoms (constipation, diarrhoea, bloating, and abdominal pain), as well as preventing or counteracting several conditions, including GI, liver, and cardiovascular diseases, obesity, diabetes, cancer, and H. pylori and urogenital infections [215,310,311,312,313]. Moreover, it is now evident that GM dysbiosis is a factor that takes part in the development of several neurological diseases, including PD, AD, multiple sclerosis, autism spectrum disorders (ASD), anxiety, depression, schizophrenia, and other mental illnesses [314,315]. Concerning PD, as previously mentioned, altered GM could contribute to the onset of some PD-related complications, such as constipation, the most common non-motor symptom [316]. Therefore, modulation of the microbiota-gut-brain axis using probiotics could be a promising complementary approach to traditional methods to prevent or counteract these disorders, including PD, as widely reported in literature [315,317,318,319,320]. For instance, Bacteroides fragilis has been documented to improve ASD symptoms and gut barrier integrity, and reduce intestinal permeability [321]; further, the probiotic SLAB51, a formulation of nine live bacterial strains (Streptococcus thermophilus, B. longum, B. breve, B. infantis, L. acidophilus, L. plantarum, L. paracasei, L. delbrueckii subsp. bulgaricus, and L. brevis) improves cognition and reduces the accumulation of amyloid plaques, brain injury, and inflammatory cytokines plasma levels in AD mice [322], while the assumption of a probiotic fermented milk drink containing L. acidophilus, L. casei, B. bifidum, and L. fermentum improves cognitive function in AD patients [323]. Concerning PD, many studies showed that probiotic intake can reduce neuroinflammation, inhibit the loss of dopaminergic neurons, and modulate brain functions, as explained in the sections that follow [324,325,326,327].

4.2.1. Preclinical Studies on Probiotics Supplementation in PD

Limited in vitro experiments have been carried out to evaluate the possible beneficial and neuroprotective effects of probiotics in alleviating the typical features of PD (Table 1) [325,328,329,330,331,332,333,334,335,336,337,338,339,340,341,342].
An in vitro GM model created with stool samples from PD patients and a cell culture model (Caco-2/THP1 cells) has been used to study the benefits of Symprove™ (Lactobacillus acidophilus NCIMB 30175, L. plantarum NCIMB 30173, L. rhamnosus NCIMB 30174 and Enterococcus faecium NCIMB 30176). Ghyselinck et al. found that the treatment with this multi-strain probiotic can change the bacterial composition (increase in Firmicutes and decrease in Bacteroidetes), stimulate the production of SCFAs and lactate, modulate mucosal inflammation (by increasing anti-inflammatory cytokines such as IL-6, IL-10 and decreasing the pro-inflammatory chemokine IL-8), and improve intestinal permeability [329]. Another study, performed by using peripheral blood mononuclear cells isolated from PD patients, Caco-2 cells, and Escherichia coli and Klebsiella pneumoniae inoculation, showed the potential use of L. salivarius LS01 and L. acidophilus LA02 in modulating inflammation (reduction in TNF-α, IL-6, and IL-17A, and increase in IL-4 and IL-10), oxidative stress, and gut permeability, and inhibiting the proliferation of pathogenic bacteria (E. coli and K. pneumoniae) [328]. In addition, Cheon et al. reported the neuroprotective effects of the heat-killed L. plantarum 200655 on H2O2-treated SH-SY5Y human neuroblastoma cells. Indeed, this probiotic is able to increase the brain-derived neurotrophic factor (BDNF) and TH mRNA expression, and decrease apoptosis [330]. Further, Castelli et al. investigated the effects of the probiotic formulation SLAB51 on the SH-SY5Y cell model of PD finding a reduction in dopaminergic neuronal loss in the SN and striatum; this effect was associated with a rise in the activation of the neuroprotective and neuronal survival BDNF pathway, a reduction in the neuronal death pathway, and a significant decrease in 4-hydroxynonenal protein adducts level, suggesting its potential antioxidant property [331].
Interestingly, Surwase and Jadhav observed the ability of the probiotic Bacillus sp. JPJ to synthesize L-dopa from L-tyrosine in vitro [343]; moreover, some probiotic strains (especially Enterococcus and Lactobacillus) have dopa decarboxylase genes in their genome, hence they can convert L-dopa to dopamine through this bacterial enzyme [180]. This evidence suggests that the combination of probiotics and L-dopa could represent a more efficient therapy for PD, although it is unlikely that dopamine may reach, as such, the brain due to the BBB presence [29].
Overall, these in vitro studies highlight the helpful role of specific probiotics in PD models; however, in vivo studies, with the same probiotics, are mandatory to support these positive results and help to better understand their safety and efficacy. Nevertheless, within this context, numerous studies, performed also by using animal models of PD, report the potential beneficial effects of probiotics supplementation on clinical symptoms and biochemical markers (Table 1). For instance, in a murine model of PD, Wang et al. demonstrated the neuroprotective role of L. plantarum DP189. Indeed, they reported the ability of this probiotic to reduce the aggregates of α-synuclein in the SN through the modulation of oxidative stress, inflammation, and GM dysbiosis. In this regard, they observed a rise in the abundance of Lactobacillus and Prevotella and a reduction in the content of Proteobacteria and Actinobacteria [335]. Furthermore, in a Caenorhabditis elegans model, it has been shown that also the probiotic Bacillus subtilis PXN21 can inhibit α-synuclein aggregation and promote the clearance of preformed aggregates [344]. In addition, a mixture of probiotics (L. plantarum CRL 2130, S. thermophilus CRL 807, and S. thermophilus CRL 808) has been shown to improve motor behaviour and neuroinflammation in a murine model of PD [345], while L. salivarius AP-32 enhanced the activity of antioxidant enzymes [SOD, glutathione peroxidase (GPx), and catalase] in a rat model of PD [346]. Another study, carried out in the 6-OHDA PD rat model, reports that supplementation with a mixture of probiotics (L. acidophilus, B. bifidum, L. reuteri, and L. fermentum) improves rotational behaviour and memory dysfunction, reduces the number of injured neurons and lipid peroxidation by decreasing malondialdehyde levels [347]. Moreover, treatment with Lacticaseibacillus rhamnosus HA-114 can improve cognition deficits but has no effects on anxiety-like behaviour in the 6-OHDA PD rat model [348].
Interestingly, creating probiotics genetically manipulated could be a strategy to increase their beneficial effects. For instance, by using a murine model of PD, it has been shown that the oral administration of the engineered strain Lactococcus lactis cremori that continually expresses glucagon-like peptide-1 (GLP-1) (MG1363-pMG36e-GLP-1) can increase TH expression, reduce locomotor and memory impairments, as well as α-synuclein production, attenuate neuroinflammation via down-regulating the TLR4/NF-κB pathway and some pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), arrest microglia and astrocyte activation, and restore GM dysbiosis [349,350]. Of note, GLP-1, crossing the BBB, binds to GLP-1 receptors, thus activating in the brain the insulin-signaling pathway involved in neurogenesis, synaptic plasticity, and neuronal metabolism [351].
Nevertheless, despite preclinical evidence suggests the benefits of probiotic supplementation in PD, in a murine model of PD, Dwyer et al. found that VSL#3 (a mixture of eight live bacterial strains) had no significant effects in modifying GM composition, preventing the reduction of dopaminergic neurons, and modulating inflammation [352,353]. These conflicting results emphasize the need of more studies aimed to better ascertain which specific probiotic supplementation, most likely a multi-strain mixture, can provide a more successful therapeutic support to face PD.

4.2.2. Clinical Studies on Probiotics Supplementation in PD

Human clinical trials have demonstrated the possible use of probiotic supplements as a potential therapeutic adjuvant for the treatment of PD. They could represent an alternative and complementary method to the traditional treatment approaches, useful to manage the disease and alleviate some of the common symptoms (Table 1).
A pilot study, performed by administering L. plantarum PS128 together with L-dopa, reports the ability of this probiotic to improve UPDRS motor scores and quality of life of PD patients, and to reduce plasma myeloperoxidase and urine creatinine levels, despite no significant changes found in non-motor symptoms [354]. Conversely, some double-blind, randomized, placebo-controlled single centre trials reported the efficacy of multi-strain probiotics in improving non-motor symptoms, including GI motility, stool consistency, and quality of life in PD patients with constipation [340,341]. In addition, Cassani et al. showed that consumption of fermented milk with L. casei Shirota can reduce abdominal pain and bloating, and ameliorate stool consistency and spontaneous defecation [336], while Georgescu et al., administering L. acidophilus and B. infantis to older PD patients, observed an improvement in abdominal pain and bloating [337]. In another randomized, double-blind, placebo-controlled clinical trial PD patients were treated with L. acidophilus, L. reuteri, L. fermentum, and B. bifidum. The obtained results show a reduction in UPDRS motor scores [339]. Conversely, the study conducted by Borzabadi et al. reported unaltered UPDRS motor scores and Non-Motor Symptom Scale (NMSS) scores in patients treated with probiotics versus the placebo cohort. Nevertheless, they observed, compared to placebo, a significant reduction in cytokines involved in inflammation (IL-1, IL-8 and TNF-α) [338]. Finally, another randomized, double-blind, placebo-controlled clinical trial, performed by using the probiotic strain Probio-M8 (B. animalis subsp. lactis Probio M-8), together with conventional drugs (benserazide and dopamine agonists), showed amelioration in sleep quality, cognitive dysfunction, defecation, and attenuation of GI symptoms [342].
In addition to being an effective support to manage some pathological conditions, probiotics can also be used as prevention tools. As an example, some evidence suggests that, during adolescence, environmental factors, such as stress, infections, inflammation, and use of antibiotics, can lead to dysbiosis, resulting in the risk to develop neurological disorders related to brain aging in adulthood. Therefore, the consumption of probiotics already from adolescence could be a good approach to prevent GM alteration and protect everyone against the onset of neurodegenerative diseases, such as PD [355].
Interestingly, some innovative approaches are also emerging from the technological/formulative point of view that could be exploited to improve the efficacy of the treatment [356,357]. For instance, Enck et al. designed a novel system for the delivery of probiotics. They encapsulated bacterial cells in modified alginate hydrogel, in order to protect the bacteria from the degradation by the acidic gastric environment [356].
In conclusion, despite the emerging benefits, the studies are still limited. Future extensive and long-term experiments are necessary to: (i) confirm the positive results obtained until now, (ii) investigate the precise mechanisms of action underlying probiotics effects, and (iii) obtain more knowledge on GM composition in different populations of patients, to find the best bacterial strains to be used, and define dosage, duration of administration, and the possible combination of different approaches.

4.3. Gut Microbiota-Based PD Interventions: Prebiotics

According to the International Scientific Association for Probiotics and Prebiotics (ISAPP), prebiotics are defined as “substrates selectively used by host microorganisms that confer health benefits to the host, while retaining the microflora-mediated health benefits” [358]. Prebiotics are dietary fibres originated from soybeans, raw oats, unrefined wheat and barley, non-digestible carbohydrates and oligosaccharides, including galacto-oligosaccharides (GOS), fructo-oligosaccharides (FOS), inulin, and lactulose [359,360]. Polyphenols (catechin, epicatechin and quercetin) can also act as prebiotics [361]. They can alter GM composition, by favouring the growth and the activity of beneficial bacteria, and by decreasing pathogens in the GI tract; further, they have positive effects on lipid metabolism, decrease the recurrence of Clostridium difficile infections, and alleviate GI and allergic disorders [362,363,364].
In the gut, the beneficial microbes metabolize the prebiotics, resulting in the generation of SCFAs (namely, acetate, propionate, butyrate) that are involved in neuromodulation, in anti-inflammatory processes, in the regulation of both intestinal and blood-brain barriers [365,366].
Like probiotics, prebiotics also play a beneficial role in managing neurological and neurodegenerative diseases [367]. For instance, lactulose and melibiose improve short-term memory and cognitive ability in AD mice [368]; bimuno-GOS ameliorate anti-social behavior in children with ASD [369]; oral administration of Marinda officinalis-derived oligosaccharides ameliorates memory and learning ability, decreases plaque formation, oxidative stress, and inflammation in both rats and mice AD models [370,371].
Concerning PD, to date, few studies have been conducted to evaluate the effects of prebiotics on PD animal models and patients (Table 2) [316,346,372,373,374,375]. In a mouse model of PD, Perez–Pardo et al. found that prebiotic fibers (FOS, GOS and nutriose, a soluble corn fibre) can normalize motor symptoms, reduce α-synuclein levels, and restore GI dysfunction, inflammation and dopamine transporter expression [372]; further, it has been shown that the prebiotic polymannuronic acid can prevent dopaminergic neuronal loss via SCFAs-mediated anti-inflammatory and anti-apoptotic mechanisms [373]. In addition, another study, performed by using 6-OHDA PD rat model, reported that the supplementation with the medium obtained from the probiotic L. salivarius subsp. salicinium AP-32 culture can reduce dopaminergic neuronal loss, motor dysfunctions, muscle atrophy, oxidative stress (increased SOD and GPx) and inflammation [346,376]. Interestingly, another study highlighted a raise in BDNF levels in the hippocampus of rats after the administration of FOS and GOS [377]. Since BDNF is involved in neuronal protection, survival, growth, and in synaptic plasticity [378], this finding suggests that prebiotics supplementation might have a role on brain neuroprotection. Finally, some studies performed in PD animal models report the beneficial effects of sodium butyrate in improving PD symptoms [379,380]; therefore, butyrogenic prebiotics could be used to increase butyrate concentration in the colon and help to manage PD [365].
In patients, two studies reported the effects of insoluble fibers on constipation. Indeed, both Astarloa et al., by administering wheat, pectin, and dimethylpolyoxyhexane-900, and Ashraf et al., by using psyllium, found a significant improvement in constipation [374,375]. Finally, another study investigated the effects of an oral supplementation with resistant starch, whose fermentation by anaerobic bacteria leads to the production of SCFAs, finding an increased butyrate concentration, as well as an improvement in non-motor symptoms [316].
In conclusion, despite few studies on PD, the satisfactory clinical outcomes on patients, especially on constipation, suggest that prebiotics might be a possible adjuvant therapy for PD, although more human clinical trials are mandatory to support this conclusion.

4.4. Gut Microbiota-Based PD Interventions: Diet

Although multifactorial interactions are involved in the prevalence and incidence of neurodegenerative diseases, nutrition plays an essential role in the pathogenesis and development of neurodegenerative diseases such as AD and PD [381,382]. Recent findings have revealed that diet, as a non-pharmacological element, plays an important role not only as a risk factor but also as a potential therapeutic approach for treating PD (Table 3) [309,383,384,385,386,387,388,389,390,391,392,393,394,395,396,397,398]. The effects of diet intervention on PD development can be attributed to different mechanisms. First, by altering intestinal microbiota composition and consequently affecting the gut-brain axis or by directly interfering with immune cells. As a matter of fact, diet is probably the most influential factor in determining the structure and metabolic function of the intestinal microbiota. Moreover, dietary components might also modulate the chronic inflammatory response that is associated with aging. Intriguingly, diet components can reduce constipation and improve L-dopa uptake, which is the first-line therapy for PD [399,400]. Therefore, consuming a constant diet on a long-term basis can impact the development of PD; however, it is still to be elucidated as to how a particular diet reduces the risk of this development. Here, we discuss how changes in diet may prevent or modify PD progression, with a special focus on Mediterranean, ketogenic, and omega-3-rich diets.

4.4.1. Mediterranean Diet

There has been extensive research on the Mediterranean diet (MD) over the years, and one large systematic review indicated that it is associated with a reduced incidence of cancer, cardiovascular disease, and AD [401]. High consumption of plant foods, including vegetables, nuts, fruits, and whole grain, moderate to weekly consumption of fish, poultry, eggs, and red wine, high intake of unsaturated fatty acids (mainly in the form of olive oil), a low to moderate intake of dairy products, and a limited use of saturated fatty acid, as well as red meat, are the major cornerstones of MD [384,387,400]. Of interest, adherence to MD is associated with a decreased risk of PD development [389,390,391,402]. One major component of MD is the high intake of dietary fibers (around 30 g/day) [403], which in turn can be utilized by the GM (especially by SCFAs-producing bacteria) as a source of energy [404]. Dietary fibers alter the gut microbiota composition in favor of fiber-fermenting bacteria rather than Gram-negative (LPS-producing) bacteria [405,406], leading to decreased neuroinflammation in PD [166,407]. Consuming a high-fiber diet may therefore improve intestinal barrier function, insulin resistance and sensitivity, GLP-1 and BDNF levels, all of which may contribute to slowing the progression of PD [166,408,409,410].
In addition to fiber, MD also contains a high quantity of flavonoid antioxidants, especially polyphenols, which have been associated with a reduced risk of PD [384,388,411]. Flavonoids in MD are present in fruits, vegetables, grains, olives, and tea. Each flavonoid molecule may act simultaneously on different mechanisms, which promote the restoration of oxidative homeostasis, the reduction of the neuroinflammatory process, and the enhancement of α-synuclein aggregates clearance [412,413,414]. Moreover, the potential of polyphenols as antioxidants and anti-inflammatory agents, as well as their ability to improve endothelial function, may contribute to lowering the risk of developing PD [411,415,416,417,418].
On the other hand, one of the features of MD is the moderate intake of dairy products. In this regard, a comprehensive umbrella study shows that high consumption of total dairy foods compared to a low intake is associated with an increased risk of PD [419]. A possible explanation is that milk proteins (casein and lactalbumin) lower serum urate levels, where urate may play a protective role against PD [420,421]. It has also been suggested that the pesticide content in dairy foods may play a role: specifically, it has been suggested that genetic susceptibility to pesticide metabolism, elimination, and transport, as well as mitochondrial dysfunction, oxidative stress, and neuronal loss, could all promote PD [422].
Another type of diet, known as the Dietary Approaches to Stop Hypertension (DASH), aims to treat and prevent high blood pressure. DASH diet shares many of the same principles as MD. Recently, some experts have suggested combining Mediterranean and DASH diets to boost cognitive function. This approach, named Mediterranean-DASH Intervention for Neurodegenerative Delay or MIND diet, has the potential to postpone the decline of cognitive scores in patients with neurodegenerative diseases [423]. Interestingly, the findings of a cross-sectional study revealed that MIND diet adherence is associated with an older age of PD onset in a superior manner to that of the MD itself [383]. Furthermore, this diet may improve fatigue and depression in PD patients [393].
In conclusion, although still limited, these observations support the need for conducting randomized controlled trials in PD patients to determine whether MD or MIND can influence neuroinflammation or the course of PD and, if so, which components provide the most benefit.

4.4.2. Ketogenic Diet

The ketogenic diet (KD) is defined as a high-fat (70–80% fat from total daily calories), adequate-protein (10–20%), low-carbohydrate (5–10%) intake [424]. As a result of such changes in macronutrient proportions, a process called ketosis results, which allows glucose to be replaced by ketone bodies in the form of acetoacetic acid, β-hydroxybutyrate (BHB), and acetone [425,426]. There have been several studies on animal models of PD showing the benefits of ketone bodies [427,428,429,430], but only a limited number of human studies have been conducted [395,396]. In this regard, the UPDRS scores greatly improved for five patients following a KD for 4 weeks [395]. In addition, a 3-month KD was also shown to improve the voice handicap index in PD patients in comparison to a regular diet [394]. Furthermore, Phillips and colleagues [396] conducted an 8-week experiment comparing a low-fat, high-carbohydrate diet with a KD. Although motor and non-motor symptoms improved in both diet groups, non-motor symptoms, such as cognitive function, ameliorated more in the KD group.
Likewise, in a rodent model of PD, BHB protects dopaminergic neurons from damage triggered by MPTP [431]. Moreover, BHB injections into the brain of mice can ameliorate the symptoms of MPTP-induced dopaminergic neurodegeneration and motor deficits [430]. Ketone bodies can exert their beneficial effects through a variety of mechanisms. For instance, adenosine triphosphate (ATP) production by ketones increases mitochondrial respiration, thus providing a neuroprotective effect [432]. It should be also mentioned that ketone bodies such as BHB provide the brain with more energy per unit of oxygen than glucose. In any case, these processes give rise to enhanced potassium channels activity, which is sensitive to ATP and adenosine, increased neurotrophic factor expression, expanded energy reserves, and stabilization of neuronal action potentials that lead to an improvement in PD symptoms [425,433]. Another intriguing potential mechanism is the effect of ketone bodies on CNS inflammation. These compounds can cross the BBB, inhibit the inflammatory response by up-regulating anti-inflammatory genes, such as MAP3K8 and TLR5, and by down-regulating pro-inflammatory genes such as TNFSF6, TNF-α, and nuclear factor-κB (NF-κB), thus decreasing inflammatory factors such as interleukins (IL-1β, IL-6) and TNF-α in SN and reducing microglia activation in various animal and in vitro models [434,435,436,437,438].
On the flip side, any diet intervention may potentially also affect PD’s pharmacotherapy. To the best of our knowledge, just one study investigated the effects of KD on L-dopa (as mentioned, one of the gold standard treatments for PD) properties. However, the findings of this study indicate that KD does not significantly influence the pharmacokinetics and pharmacodynamics of L-dopa [439].
Once again, although it is a must to investigate the detailed mechanism of action of KD’s components, as well as the overall effectiveness of this diet in large randomized clinical trials and cohort studies, it must be highlighted that patient compliance with the KD (even when modified) is poor, due to the restrictive nature of the diet and GI discomfort symptoms.

4.4.3. Omega-3 Fatty Acids

Omega-3 (ω3) polyunsaturated fatty acids (PUFAs), including docosahexaenoic acid [DHA, 22:6 (n-3)], eicosapentaenoic acid [EPA, 20:5(n-3)], and α-linolenic acid [ALA, 18:3 (n-3)], play a crucial role in maintaining the structure and healthy function of different organs. They are involved in inflammatory and immunological processes, as well as in hormonal regulation [440,441]. Some limited human studies have shown that consuming PUFAs, being components of different diets such as MD or KD, and specifically the supplementation with EPA and DHA may reduce the risk of PD and relieve some of its symptoms like motor symptoms [442,443,444]. Besides the motor symptoms, depression affects over 40% of PD cases [445]. According to both epidemiological and clinical studies, a reduced dietary intake of fatty acids, especially ω3, is associated with mood disorders and depression [446,447,448,449]. Interestingly, in a clinical study where PD patients were taking fish oil (a great source of ω3) with or without antidepressants presented improvements in depressive symptoms [450]. As a result, the addition of ω3 to the diet of PD patients may be a valuable approach to reduce depression symptoms, which can also impact other clinical aspects of the disease.
Besides having a crucial role in membrane fluidity and a broad spectrum of activities, ω3 may be also beneficial at several levels of the neuronal degenerative process observed in PD [451]. As mentioned, PD, like other neurodegenerative diseases, is associated with oxidative stress and inflammation pathways, which are tightly linked and interdependent [452]. Elevated levels of cytokines and prostaglandins have been detected in the cerebrospinal fluid and brains of PD patients where they may in turn activate the microglia, induce migration of microglia to SN, and release pro-inflammatory cytokines, as well as neurotoxic molecules capable of further exacerbating the disease [453,454,455,456]. Within this context, ω3 are able to ameliorate oxidative stress and neuroinflammation, which may explain part of their beneficial effects on PD [457]. Indeed, a clinical study found that supplementation of PD patients with ω3 has a favorable effect on glutathione concentrations, as well as on the total antioxidant capacity, and it is associated with a lower amount of C-reactive protein [458]. These findings are supported by experimental evidence that ω3-PUFAs reduce NOS activity and increase BDNF levels in the CNS [459,460]. In addition to their neuroprotective effects, ω3 (especially DHA) may modulate the brain’s dopamine systems through different mechanisms [461]. In conclusion, these observations provide strong scientific support for conducting randomized controlled trials to assess whether ω3 supplements can slow down degeneration and thereby modify the course of the disease.

4.5. Gut Microbiota-Based PD Interventions: Fecal Microbiota Transplantation

Fecal microbiota transplantation (FMT) consists in the transfer of resuspended and filtered stool material from a healthy donor to a patient’s gut. The aim of this approach is to counteract dysbiosis while favoring the establishment of a beneficial and balanced microbiota [462,463]. Although colonoscopy is the preferred method of transplantation, delivery through nasogastric or nasojejunal tube, enema, or orally administered capsules have also been tested [462,464]. Following the successful use of FMT in the treatment of refractory or recurrent Clostridium difficile infection, several studies have been conducted to explore FMT as a therapeutic strategy for a wide range of neurological disorders, including multiple sclerosis, epilepsy, ASD, Tourette syndrome, diabetic neuropathy, AD and PD, with promising preclinical and clinical data [30,465,466,467,468]. Concerning PD, consistent preclinical studies and a handful of human case reports have shown that FMT might be exploited to reduce motor and non-motor symptoms, as well as constipation, at least in the short term [40,465,469,470,471,472,473,474,475,476,477] (Table 4). Early evidence came in 2016 from the work of Sampson et al., who first reported that the transfer of fecal matter from human PD patients to α-synuclein overexpressing mice substantially worsened their physical symptoms in comparison with mice receiving feces from healthy human donors [40]. These results were then confirmed in 2018, when Sun et al. showed that fecal microbiota transfer from PD mice to their healthy counterpart increases motor deficits while reducing the striatal levels of the neurotransmitters dopamine, serotonin and their metabolites, thus reproducing the typical features of the disease [469]. Conversely, fecal matter transplantation from healthy mice to PD recipient mice improved physical performance, ameliorated motor symptoms and reduced dysbiosis in several independent studies [469,470,471,472]. Looking at the GM composition, there is evidence that FMT re-establishes eubiosis by disadvantaging the growth of Desulfovibrio, Akkermansia and Proteobacteria (orders Enterobacteriales and Turicibacteriales), while simultaneously favoring the proliferation of beneficial bacteria such as Bacteroidetes and Actinobacteria phyla, with a particular effect on Blautia and Prevotella species [469,470,472]. Moreover, FMT appears to protect from gut inflammation by promoting intestinal barrier integrity and reducing the levels of LPS in the colon, serum, and SN, therefore preventing leaky gut and systemic inflammation [470]. At the brain level, FMT contrasts cognitive damage by decreasing α-synuclein expression and restoring the optimal levels of the striatal neurotransmitters dopamine and serotonin, thus supporting neuroprotection [469,471,473]. Notably, decreased neuroinflammation following FMT has been reported by numerous preclinical studies [469,470,471,472]. This beneficial effect should be ascribed to the ability of GM to modulate microglia and astrocyte activation in SN by regulating the TLR4/NF-κB pro-inflammatory pathway and reducing the expression of GSK3β, iNOS and IL-1β, which are implicated in PD pathogenesis and progression [469,470,471,472,478,479,480].
Nevertheless, FMT studies involving humans are still limited. One case study conducted in 2019 by Huang et al. investigated the potential therapeutic benefit of FMT in a 71-year-old male PD patient reporting constipation (>3 years) and motor symptoms (for 7 years). While FMT successfully promoted regular defecation, tremor disappeared only temporarily and then reappeared two months after transplant [474]. Three subsequent studies involving 6, 11 and 15 PD patients, respectively, confirmed the reduction in constipation, as well as in motor and non-motor symptoms following FMT, as indicated by the decreased scores registered in various PD assessment tests [475,476,477].
On the whole, from a clinical point of view, better and longer-term outcomes were obtained using colonoscopy compared to nasointestinal delivery [476]. In line with preclinical data, PD patients undergoing FMT showed an increased presence of Blautia and Prevotella and a diminished overall abundance of Bacteroidetes, thus confirming the efficacy of this approach in modifying the GM composition [475].
Despite the therapeutic potential of FMT for the treatment of PD, several limitations still exist and need to be addressed. Standard clinical protocols, delivery methods, periodicity, donor’s selection criteria, patient’s inclusion criteria, long-term benefits and potential risks remain an issue [463,467,481,482,483,484,485]. Within this context, 6 cases of adverse events occurred in human studies: flatulence (1), diarrhea (2), hospitalization under observation (1) and GI pain (2) [476,477]. Therefore, although not life-threatening, the nature of these complications should be better investigated.
In addition, randomized controlled trials involving a considerable number of patients are required to better assess feasibility, therapeutic efficacy, safety and long-term benefits of this promising GM-modifying approach [486].

5. Oral and Nasal Microbiota: Other Important Districts Involved in the Disease

Preliminary evidence is showing that specific oral microbiota compositions, also known as oral microbiota signatures, are associated with human healthy aging [487]. Accordingly, alterations in the abundance of oral bacteria have been directly or indirectly linked to the onset of different disorders, such as cancer, cardiovascular and pulmonary diseases, stroke, diabetes and even neurodegeneration [487,488,489,490,491]. In the latter case, it has been hypothesized that the increased prevalence of mouth anaerobic microbes observed during aging might generate a TNF-α-mediated pro-inflammatory environment that damages the BBB integrity and favors bacteria spreading [492]. In this respect, 16S rRNA sequencing of the oral microbes performed in mild cognitive impaired (MCI) or healthy individuals revealed that increased Pasteurellaceae and decreased Lautropia mirabilis were associated with altered cerebrospinal fluid levels of the inflammatory mediators oncostatin M, T-cell surface glycoprotein CD8 alpha chain, MMP10, thymic stromal lymphopoietin and chemokine ligand 3 [493]. In AD, there is evidence that oral microbiota dysbiosis induced by alcohol consumption takes part in AD pathogenesis through the modulation of eIF2, eIF4 and mTOR pathways [494]. Moreover, results from a study conducted in a cohort of Canadian individuals with neurodegeneration reported higher microbial diversity (in contrast to what is observed in the GM), diminished Streptococcaceae and Actinomycetaceae, as well as enhanced Weeksellaceae and Porphyromonas in these patients compared to controls, but these changes did not correlate with cognitive decline [495]. Concerning PD, oral features associated with the disease such as dysphagia, salivary pH and drooling may influence the oral microbiota by altering the β-diversity index and favoring the growth of opportunistic oral microbes and Lactobacillus species, which may exacerbate the clinical symptoms and correlate with worse Hamilton Anxiety Scale and Hamilton Depression Rating Scale scores [70,496,497]. Moreover, combined oral-gut microbiome metagenomic sequencing revealed a link between oral Lactobacillus and gut opportunistic pathogens, suggesting a connection between these two microbial communities [498]. Other oral microbial species whose abundance has been found altered in PD patients are: Actinomyces AFQC_s, Scardovia, Kingella oralis, Streptococcus mutans, Veillonella AFUJ_s, Prevotella, Firmicutes, Negativicutes (all increased) and Lachnospiraceae AM420052_s, Treponema KE332528_s, Proteobacteria, Tenercutes and Pastescibacteria (all decreased) [499,500]. Of note, these alterations can modulate the expression of salivary host mRNAs involved in multiple brain activities, thus supporting the relationship between oral microbiota and cognitive disorders [497].
Because of dysbiosis, the aminoacidic and energetic metabolisms associated with oral bacteria vary [497]. In this respect, 16SrRNA sequencing performed on 91 PD patients and 91 control individuals reported an increase in pathways related to oxidative phosphorylation and carbohydrate metabolism among oral microbes associated with the disease [70]. Another consequence of dysbiosis is the establishment of a pro-inflammatory environment, in a closely interconnected feedback loop [70,501]. For example, an increase in different local pro-inflammatory molecules, such as IL1-RA, IL-1β and TNF-α, has been associated with dysbiosis in a study comparing 20 PD patients and 20 matched controls, which may contribute to the onset of PD [499]. Moreover, mice receiving Porphyromonas gingivalis, a pathogen associated with chronic periodontitis, showed sustained colon TNF-α and IL-1β expression, as well as higher levels of serum IL-17A and its receptor (IL-17AR) in the brain, which were associated with loss of dopaminergic neurons, α -synuclein accumulation and microglia activation [502]. However, more studies are needed to better address the role of oral dysbiosis in triggering local, intestinal and systemic inflammation, and to evaluate the impact of this highly interconnected pathway on brain function.
Since olfactory dysfunction is an early marker of PD, which arises far before the appearance of the motor symptoms, few authors started to investigate the potential role of nasal microbiota in disease pathogenesis [503]. Given the interconnection between nose and brain, some studies hypothesized that changes in nasal microbiota community may promote neuroinflammation through the olfactory bulb, thus establishing a nose-to-brain axis [503,504]. However, until today, data remain scarce. So far, the only evidence comes from a study by Pal et al., who performed 16SrRNA sequencing of the deep nasal cavity in PD patients versus healthy controls and revealed a positive correlation between the abundance of the opportunistic bacterium Moraxella catarrhalis and motor symptoms in PD [503]. Nevertheless, no significant differences in nasal microbiota composition have been reported in two other studies comparing 91 PD patients to 91 controls and 76 PD patients to 78 healthy controls, respectively [70,80]. This discrepancy probably reflects the struggle to consider different variables, such as sampling location, collection method and analysis pipeline, which often differ among studies [503]. Moreover, the nasal microbiota composition, as well as the oral one, changes according to dietary habits, host factors, environmental conditions and even the season [503,505].
Overall, promising evidence is emerging for an association between oral/nasal microbiota and PD, but data remain limited and sometimes conflicting. Once better investigated, these two microbial communities might be exploited as early diagnostic and therapeutic tools for PD, as already proposed [504]. For example, the combination of 11 taxonomic features associated with the oral microbiota has allowed PD diagnosis reaching up to 84.5% accuracy, but better performances are expected to come [497]. Moreover, yeast and phage populations seem also to differ in PD patients compared to controls, and their further investigation might be of interest [497]. Until now, however, since the number of studies investigating the role of nasal and oral microbial communities on PD remains scarce, more data are needed before drawing any conclusions [506].

6. Discussion

Overall, our narrative review summarizes the main data on the link between dysbiosis and PD, with a particular focus on the possibility of taking advantage of this knowledge not only for diagnostic purposes, but also for a therapeutic application. However, several limitations still exist and need to be discussed. A major concern is the discrepancy and variability sometimes found in GM studies, which limit the reproducibility. In this respect, the adoption of common laboratory protocols, unique bioinformatics pipelines and shared analysis methods is fundamental to reduce external confounders [507]. In addition, it is known that differences in geography, diet and lifestyle can greatly influence the GM composition [91,508,509], and host genetics may play a major role [510]. Since many of these inherent covariates cannot be eliminated, the search for biomarkers and therapies focused on specific populations, as well as more “personalized” approaches, may be of interest and improve consistency [511,512]. However, the feasibility of these more targeted approaches highly depends on the availability of a large amount of data, which for the moment remains a limitation. When considering diagnosis, the multi-omics analysis of samples from PD patients at different stages of disease progression, including prodromal and early subjects is of utmost importance to better define stage-associated signatures [507]. Moreover, the lack of sufficient comparisons between microbial profiles obtained from patients under conventional pharmacological treatment versus treatment-naïve PD individuals limits their diagnostic applicability, since drugs intake may greatly affect GM composition [185,186,188]. Additionally, the dose and time of administration of GM-modifying drugs may also vary depending on the stage of the disease, thus possibly further influencing the study outcomes. Furthermore, the optimal probiotic-prebiotic cocktail to be employed has yet to be identified, as well as the best dietary intervention. Since a crucial aspect in the context of aging-related neurodegenerative diseases is prevention, forthcoming research should better assess if preventative benefits can be obtained by undergoing eubiosis-reestablishing therapies in a prodromal phase of the disease, rather than focusing entirely on disease treatment [351,513]. In the future, the administration of engineered microbes (also known as live biotherapeutic products), which carry the traits of interest, together with defined and balanced diets, may be an accessible, non-invasive and practicable intervention in support of pharmacological PD treatment, but more research is needed before a clinical application [215].
Overall, although much work remains to be done and large clinical trials are required, GM is emerging as a promising diagnostic and therapeutic tool for PD and deserves further investigation.

Author Contributions

Conceptualization, A.V. and A.P.; writing—original draft preparation, A.V., L.I.M.C., F.F., E.P. and S.C.; writing—review and editing, A.V., M.R., M.N. and A.P.; supervision, G.R. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

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. Sousa-Fraguas, M.C.; Rodríguez-Fuentes, G.; Conejo, N.M. Frailty and Cognitive Impairment in Parkinson’s Disease: A Systematic Review. Neurol. Sci. 2022, 1–14. [Google Scholar] [CrossRef] [PubMed]
  2. Leta, V.; Urso, D.; Batzu, L.; Lau, Y.H.; Mathew, D.; Boura, I.; Raeder, V.; Falup-Pecurariu, C.; van Wamelen, D.; Ray Chaudhuri, K. Viruses, Parkinsonism and Parkinson’s Disease: The Past, Present and Future. J. Neural Transm. 2022, 129, 1119–1132. [Google Scholar] [CrossRef] [PubMed]
  3. Zeng, J.; Wang, X.; Pan, F.; Mao, Z. The Relationship between Parkinson’s Disease and Gastrointestinal Diseases. Front. Aging Neurosci. 2022, 14, 955919. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, R.; Berardelli, A.; Bhattacharya, A.; Bologna, M.; Chen, K.-H.S.; Fasano, A.; Helmich, R.C.; Hutchison, W.D.; Kamble, N.; Kühn, A.A.; et al. Clinical Neurophysiology of Parkinson’s Disease and Parkinsonism. Clin. Neurophysiol. Pract 2022, 7, 201–227. [Google Scholar] [CrossRef] [PubMed]
  5. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and Enteric Nervous Systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  6. Klann, E.M.; Dissanayake, U.; Gurrala, A.; Farrer, M.; Shukla, A.W.; Ramirez-Zamora, A.; Mai, V.; Vedam-Mai, V. The Gut–Brain Axis and Its Relation to Parkinson’s Disease: A Review. Front. Aging Neurosci. 2022, 13, 782082. [Google Scholar] [CrossRef] [PubMed]
  7. Thangaleela, S.; Sivamaruthi, B.S.; Kesika, P.; Bharathi, M.; Chaiyasut, C. Role of the Gut–Brain Axis, Gut Microbial Composition, Diet, and Probiotic Intervention in Parkinson’s Disease. Microorganisms 2022, 10, 1544. [Google Scholar] [CrossRef] [PubMed]
  8. Sherman, M.P.; Zaghouani, H.; Niklas, V. Gut Microbiota, the Immune System, and Diet Influence the Neonatal Gut–Brain Axis. Pediatr. Res. 2015, 77, 127–135. [Google Scholar] [CrossRef] [PubMed]
  9. Smith, D.; Jheeta, S.; Fuentes, H.V.; Palacios-Pérez, M. Feeding Our Microbiota: Stimulation of the Immune/Semiochemical System and the Potential Amelioration of Non-Communicable Diseases. Life 2022, 12, 1197. [Google Scholar] [CrossRef] [PubMed]
  10. Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J.K.; Carding, S.R.; Glen, R.C.; McArthur, S. Microbiome–Host Systems Interactions: Protective Effects of Propionate upon the Blood–Brain Barrier. Microbiome 2018, 6, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Tran, S.M.-S.; Mohajeri, M.H. The Role of Gut Bacterial Metabolites in Brain Development, Aging and Disease. Nutrients 2021, 13, 732. [Google Scholar] [CrossRef] [PubMed]
  12. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hirayama, M.; Ohno, K. Parkinson’s Disease and Gut Microbiota. Ann. Nutr. Metab. 2021, 77, 28–35. [Google Scholar] [CrossRef]
  14. Grathwohl, S.A.; Steiner, J.A.; Britschgi, M.; Brundin, P. Mind the Gut: Secretion of α-Synuclein by Enteric Neurons. J. Neurochem. 2013, 125, 487–490. [Google Scholar] [CrossRef] [PubMed]
  15. Sampson, T.R.; Challis, C.; Jain, N.; Moiseyenko, A.; Ladinsky, M.S.; Shastri, G.G.; Thron, T.; Needham, B.D.; Horvath, I.; Debelius, J.W.; et al. A Gut Bacterial Amyloid Promotes α-Synuclein Aggregation and Motor Impairment in Mice. Elife 2020, 9, e53111. [Google Scholar] [CrossRef] [PubMed]
  16. Mulak, A. Brain-Gut-Microbiota Axis in Parkinson’s Disease. World J. Gastroenterol. 2015, 21, 10609. [Google Scholar] [CrossRef] [PubMed]
  17. Schemann, M.; Neunlist, M. The Human Enteric Nervous System. Neurogastroenterol. Motil. 2004, 16, 55–59. [Google Scholar] [CrossRef] [PubMed]
  18. Szurszewski, J.H. Physiology of Mammalian Prevertebral Ganglia. Annu. Rev. Physiol. 1981, 43, 53–68. [Google Scholar] [CrossRef] [PubMed]
  19. Dogra, N.; Mani, R.J.; Katare, D.P. The Gut-Brain Axis: Two Ways Signaling in Parkinson’s Disease. Cell Mol. Neurobiol. 2022, 42, 315–332. [Google Scholar] [CrossRef] [PubMed]
  20. Chen, S.G.; Stribinskis, V.; Rane, M.J.; Demuth, D.R.; Gozal, E.; Roberts, A.M.; Jagadapillai, R.; Liu, R.; Choe, K.; Shivakumar, B.; et al. Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha-Synuclein Aggregation in Aged Fischer 344 Rats and Caenorhabditis Elegans. Sci. Rep. 2016, 6, 34477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Murros, K.E.; Huynh, V.A.; Takala, T.M.; Saris, P.E.J. Desulfovibrio Bacteria Are Associated With Parkinson’s Disease. Front. Cell Infect Microbiol. 2021, 11, 652617. [Google Scholar] [CrossRef] [PubMed]
  22. Yuan, Y.-Q.; Wang, Y.-L.; Yuan, B.-S.; Yuan, X.; Hou, X.-O.; Bian, J.-S.; Liu, C.-F.; Hu, L.-F. Impaired CBS-H2S Signaling Axis Contributes to MPTP-Induced Neurodegeneration in a Mouse Model of Parkinson’s Disease. Brain Behav. Immun. 2018, 67, 77–90. [Google Scholar] [CrossRef] [PubMed]
  23. Murros, K.E. Hydrogen Sulfide Produced by Gut Bacteria May Induce Parkinson’s Disease. Cells 2022, 11, 978. [Google Scholar] [CrossRef] [PubMed]
  24. Dinan, K.; Dinan, T.G. Gut Microbes and Neuropathology: Is There a Causal Nexus? Pathogens 2022, 11, 796. [Google Scholar] [CrossRef]
  25. Ettinger, S. Diet, Gut Microbiome, and Cognitive Decline. Curr. Nutr. Rep. 2022. [Google Scholar] [CrossRef]
  26. Costa, H.N.; Esteves, A.R.; Empadinhas, N.; Cardoso, S.M. Parkinson’s Disease: A Multisystem Disorder. Neurosci. Bull 2022. [Google Scholar] [CrossRef]
  27. Choi, J.; Kim, B.-R.; Akuzum, B.; Chang, L.; Lee, J.-Y.; Kwon, H.-K. TREGking From Gut to Brain: The Control of Regulatory T Cells Along the Gut-Brain Axis. Front. Immunol. 2022, 13, 3193. [Google Scholar] [CrossRef]
  28. Pascale, A.; Marchesi, N.; Marelli, C.; Coppola, A.; Luzi, L.; Govoni, S.; Giustina, A.; Gazzaruso, C. Microbiota and Metabolic Diseases. Endocrine 2018, 61, 357–371. [Google Scholar] [CrossRef]
  29. Pascale, A.; Marchesi, N.; Govoni, S.; Barbieri, A. Targeting the Microbiota in Pharmacology of Psychiatric Disorders. Pharmacol. Res. 2020, 157, 104856. [Google Scholar] [CrossRef]
  30. Varesi, A.; Pierella, E.; Romeo, M.; Piccini, G.B.; Alfano, C.; Bjørklund, G.; Oppong, A.; Ricevuti, G.; Esposito, C.; Chirumbolo, S.; et al. The Potential Role of Gut Microbiota in Alzheimer’s Disease: From Diagnosis to Treatment. Nutrients 2022, 14, 668. [Google Scholar] [CrossRef]
  31. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of Brain Amyloidosis with Pro-Inflammatory Gut Bacterial Taxa and Peripheral Inflammation Markers in Cognitively Impaired Elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef] [PubMed]
  32. Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut Microbiome Alterations in Alzheimer’s Disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Varesi, A.; Deumer, U.-S.; Ananth, S.; Ricevuti, G. The Emerging Role of Gut Microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Current Evidence and Potential Therapeutic Applications. J. Clin. Med. 2021, 10, 5077. [Google Scholar] [CrossRef] [PubMed]
  34. Galicia-Garcia, U.; Benito-Vicente, A.; Jebari, S.; Larrea-Sebal, A.; Siddiqi, H.; Uribe, K.B.; Ostolaza, H.; Martín, C. Pathophysiology of Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2020, 21, 6275. [Google Scholar] [CrossRef]
  35. Abenavoli, L.; Scarpellini, E.; Colica, C.; Boccuto, L.; Salehi, B.; Sharifi-Rad, J.; Aiello, V.; Romano, B.; de Lorenzo, A.; Izzo, A.A.; et al. Gut Microbiota and Obesity: A Role for Probiotics. Nutrients 2019, 11, 2690. [Google Scholar] [CrossRef] [Green Version]
  36. Gerhardt, S.; Mohajeri, M. Changes of Colonic Bacterial Composition in Parkinson’s Disease and Other Neurodegenerative Diseases. Nutrients 2018, 10, 708. [Google Scholar] [CrossRef] [Green Version]
  37. Weiss, G.A.; Hennet, T. Mechanisms and Consequences of Intestinal Dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [Google Scholar] [CrossRef] [Green Version]
  38. Schwabe, R.F.; Jobin, C. The Microbiome and Cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [Green Version]
  39. Perez-Pardo, P.; Dodiya, H.B.; Engen, P.A.; Naqib, A.; Forsyth, C.B.; Green, S.J.; Garssen, J.; Keshavarzian, A.; Kraneveld, A.D. Gut Bacterial Composition in a Mouse Model of Parkinson’s Disease. Benef. Microbes 2018, 9, 799–814. [Google Scholar] [CrossRef] [Green Version]
  40. Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [Green Version]
  41. Sun, M.-F.; Shen, Y.-Q. Dysbiosis of Gut Microbiota and Microbial Metabolites in Parkinson’s Disease. Ageing Res. Rev. 2018, 45, 53–61. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, X.; Qian, Y.; Xu, S.; Song, Y.; Xiao, Q. Longitudinal Analysis of Fecal Microbiome and Pathologic Processes in a Rotenone Induced Mice Model of Parkinson’s Disease. Front. Aging Neurosci. 2018, 9, 441. [Google Scholar] [CrossRef] [Green Version]
  43. Lai, F.; Jiang, R.; Xie, W.; Liu, X.; Tang, Y.; Xiao, H.; Gao, J.; Jia, Y.; Bai, Q. Intestinal Pathology and Gut Microbiota Alterations in a Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP) Mouse Model of Parkinson’s Disease. Neurochem. Res. 2018, 43, 1986–1999. [Google Scholar] [CrossRef] [PubMed]
  44. Zhu, Y.; Huan, F.; Wang, J.; Xie, X.; Yu, G.; Wang, X.; Jiang, L.; Gao, R.; Xiao, H.; Ding, H.; et al. 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Induced Parkinson’s Disease in Mouse: Potential Association between Neurotransmitter Disturbance and Gut Microbiota Dysbiosis. ACS Chem. Neurosci. 2020, 11, 3366–3376. [Google Scholar] [CrossRef] [PubMed]
  45. Takahashi, K.; Nishiwaki, H.; Ito, M.; Iwaoka, K.; Takahashi, K.; Suzuki, Y.; Taguchi, K.; Yamahara, K.; Tsuboi, Y.; Kashihara, K.; et al. Altered Gut Microbiota in Parkinson’s Disease Patients with Motor Complications. Park. Relat. Disord. 2022, 95, 11–17. [Google Scholar] [CrossRef] [PubMed]
  46. Cassani, E.; Barichella, M.; Cancello, R.; Cavanna, F.; Iorio, L.; Cereda, E.; Bolliri, C.; Zampella Maria, P.; Bianchi, F.; Cestaro, B.; et al. Increased Urinary Indoxyl Sulfate (Indican): New Insights into Gut Dysbiosis in Parkinson’s Disease. Park. Relat. Disord. 2015, 21, 389–393. [Google Scholar] [CrossRef]
  47. Zhang, F.; Yue, L.; Fang, X.; Wang, G.; Li, C.; Sun, X.; Jia, X.; Yang, J.; Song, J.; Zhang, Y.; et al. Altered Gut Microbiota in Parkinson’s Disease Patients/Healthy Spouses and Its Association with Clinical Features. Park. Relat. Disord. 2020, 81, 84–88. [Google Scholar] [CrossRef]
  48. Wallen, Z.D.; Appah, M.; Dean, M.N.; Sesler, C.L.; Factor, S.A.; Molho, E.; Zabetian, C.P.; Standaert, D.G.; Payami, H. Characterizing Dysbiosis of Gut Microbiome in PD: Evidence for Overabundance of Opportunistic Pathogens. NPJ Park. Dis. 2020, 6, 11. [Google Scholar] [CrossRef]
  49. Nishiwaki, H.; Hamaguchi, T.; Ito, M.; Ishida, T.; Maeda, T.; Kashihara, K.; Tsuboi, Y.; Ueyama, J.; Shimamura, T.; Mori, H.; et al. Short-Chain Fatty Acid-Producing Gut Microbiota Is Decreased in Parkinson’s Disease but Not in Rapid-Eye-Movement Sleep Behavior Disorder. mSystems 2020, 5, e00797-20. [Google Scholar] [CrossRef]
  50. Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic Bacterial Composition in Parkinson’s Disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef]
  51. Lubomski, M.; Xu, X.; Holmes, A.J.; Muller, S.; Yang, J.Y.H.; Davis, R.L.; Sue, C.M. The Gut Microbiome in Parkinson’s Disease: A Longitudinal Study of the Impacts on Disease Progression and the Use of Device-Assisted Therapies. Front. Aging Neurosci. 2022, 14. [Google Scholar] [CrossRef] [PubMed]
  52. Nishiwaki, H.; Ito, M.; Ishida, T.; Hamaguchi, T.; Maeda, T.; Kashihara, K.; Tsuboi, Y.; Ueyama, J.; Shimamura, T.; Mori, H.; et al. Meta-Analysis of Gut Dysbiosis in Parkinson’s Disease. Mov. Disord. 2020, 35, 1626–1635. [Google Scholar] [CrossRef] [PubMed]
  53. Shen, T.; Yue, Y.; He, T.; Huang, C.; Qu, B.; Lv, W.; Lai, H.-Y. The Association Between the Gut Microbiota and Parkinson’s Disease, a Meta-Analysis. Front. Aging Neurosci. 2021, 13, 636545. [Google Scholar] [CrossRef] [PubMed]
  54. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-Analysis of the Parkinson’s Disease Gut Microbiome Suggests Alterations Linked to Intestinal Inflammation. NPJ Park. Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
  55. Wallen, Z.D.; Stone, W.J.; Factor, S.A.; Molho, E.; Zabetian, C.P.; Standaert, D.G.; Payami, H. Exploring Human-Genome Gut-Microbiome Interaction in Parkinson’s Disease. NPJ Park. Dis. 2021, 7, 74. [Google Scholar] [CrossRef]
  56. Ji, T.; Huang, H.; Liu, J.; Peng, T.; Zhou, X.; Tan, Q.; Yuan, J.; Hua, H.; Ding, S.; Liu, H. Leveraging Sequence-based Faecal Microbial Community Survey Data to Identify Alterations in Gut Microbiota among Patients with Parkinson’s Disease. Eur. J. Neurosci. 2021, 53, 687–696. [Google Scholar] [CrossRef]
  57. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.-J.; Blugeon, S.; Bridonneau, C.; Furet, J.-P.; Corthier, G.; et al. Faecalibacterium Prausnitzii Is an Anti-Inflammatory Commensal Bacterium Identified by Gut Microbiota Analysis of Crohn Disease Patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [Green Version]
  58. Lenoir, M.; Martín, R.; Torres-Maravilla, E.; Chadi, S.; González-Dávila, P.; Sokol, H.; Langella, P.; Chain, F.; Bermúdez-Humarán, L.G. Butyrate Mediates Anti-Inflammatory Effects of Faecalibacterium Prausnitzii in Intestinal Epithelial Cells through Dact3. Gut Microbes 2020, 12, 1826748. [Google Scholar] [CrossRef] [PubMed]
  59. Cosma-Grigorov, A.; Meixner, H.; Mrochen, A.; Wirtz, S.; Winkler, J.; Marxreiter, F. Changes in Gastrointestinal Microbiome Composition in PD: A Pivotal Role of Covariates. Front. Neurol. 2020, 11, 1041. [Google Scholar] [CrossRef]
  60. Xie, A.; Ensink, E.; Li, P.; Gordevičius, J.; Marshall, L.L.; George, S.; Pospisilik, J.A.; Aho, V.T.E.; Houser, M.C.; Pereira, P.A.B.; et al. Bacterial Butyrate in Parkinson’s Disease Is Linked to Epigenetic Changes and Depressive Symptoms. Mov. Disord. 2022, 37, 1644–1653. [Google Scholar] [CrossRef]
  61. Tan, A.H.; Chong, C.W.; Lim, S.; Yap, I.K.S.; Teh, C.S.J.; Loke, M.F.; Song, S.; Tan, J.Y.; Ang, B.H.; Tan, Y.Q.; et al. Gut Microbial Ecosystem in Parkinson Disease: New Clinicobiological Insights from Multi-Omics. Ann. Neurol. 2021, 89, 546–559. [Google Scholar] [CrossRef]
  62. Chen, S.-J.; Chen, C.-C.; Liao, H.-Y.; Lin, Y.-T.; Wu, Y.-W.; Liou, J.-M.; Wu, M.-S.; Kuo, C.-H.; Lin, C.-H. Association of Fecal and Plasma Levels of Short-Chain Fatty Acids With Gut Microbiota and Clinical Severity in Patients With Parkinson Disease. Neurology 2022, 98, e848–e858. [Google Scholar] [CrossRef] [PubMed]
  63. 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]
  64. Mao, L.; Zhang, Y.; Tian, J.; Sang, M.; Zhang, G.; Zhou, Y.; Wang, P. Cross-Sectional Study on the Gut Microbiome of Parkinson’s Disease Patients in Central China. Front. Microbiol. 2021, 12, 728479. [Google Scholar] [CrossRef] [PubMed]
  65. Li, F.; Wang, P.; Chen, Z.; Sui, X.; Xie, X.; Zhang, J. Alteration of the Fecal Microbiota in North-Eastern Han Chinese Population with Sporadic Parkinson’s Disease. Neurosci. Lett. 2019, 707, 134297. [Google Scholar] [CrossRef] [PubMed]
  66. Kenna, J.E.; Chua, E.G.; Bakeberg, M.; Tay, A.; McGregor, S.; Gorecki, A.; Horne, M.; Marshall, B.; Mastaglia, F.L.; Anderton, R.S. Changes in the Gut Microbiome and Predicted Functional Metabolic Effects in an Australian Parkinson’s Disease Cohort. Front. Neurosci. 2021, 15, 756951. [Google Scholar] [CrossRef]
  67. Parada Venegas, D.; de la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  68. Lin, A.; Zheng, W.; He, Y.; Tang, W.; Wei, X.; He, R.; Huang, W.; Su, Y.; Huang, Y.; Zhou, H.; et al. Gut Microbiota in Patients with Parkinson’s Disease in Southern China. Park. Relat. Disord. 2018, 53, 82–88. [Google Scholar] [CrossRef]
  69. 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]
  70. Li, Z.; Lu, G.; Luo, E.; Wu, B.; Li, Z.; Guo, J.; Xia, Z.; Zheng, C.; Su, Q.; Zeng, Y.; et al. Oral, Nasal, and Gut Microbiota in Parkinson’s Disease. Neuroscience 2022, 480, 65–78. [Google Scholar] [CrossRef]
  71. 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]
  72. Hopfner, F.; Künstner, A.; Müller, S.H.; Künzel, S.; Zeuner, K.E.; Margraf, N.G.; Deuschl, G.; Baines, J.F.; Kuhlenbäumer, G. Gut Microbiota in Parkinson Disease in a Northern German Cohort. Brain Res. 2017, 1667, 41–45. [Google Scholar] [CrossRef] [PubMed]
  73. Qian, Y.; Yang, X.; Xu, S.; Wu, C.; Song, Y.; Qin, N.; Chen, S.-D.; Xiao, Q. Alteration of the Fecal Microbiota in Chinese Patients with Parkinson’s Disease. Brain Behav. Immun. 2018, 70, 194–202. [Google Scholar] [CrossRef] [PubMed]
  74. Vascellari, S.; Melis, M.; Palmas, V.; Pisanu, S.; Serra, A.; Perra, D.; Santoru, M.L.; Oppo, V.; Cusano, R.; Uva, P.; et al. Clinical Phenotypes of Parkinson’s Disease Associate with Distinct Gut Microbiota and Metabolome Enterotypes. Biomolecules 2021, 11, 144. [Google Scholar] [CrossRef]
  75. Ransmayr, G.; Poewe, W.; Plörer, S.; Gerstenbrand, F.; Leidlmair, K.; Mayr, U. Prognostic Implications of the Motor Symptoms of Parkinson’s Disease with Respect to Clinical, Computertomographic and Psychometric Parameters. J. Neural. Transm. 1986, 67, 1–14. [Google Scholar] [CrossRef]
  76. Rajput, A.H.; Pahwa, R.; Pahwa, P.; Rajput, A. Prognostic Significance of the Onset Mode in Parkinsonism. Neurology 1993, 43, 829–830. [Google Scholar] [CrossRef] [PubMed]
  77. Zetusky, W.J.; Jankovic, J.; Pirozzolo, F.J. The Heterogeneity of Parkinson’s Disease: Clinical and Prognostic Implications. Neurology 1985, 35, 522–526. [Google Scholar] [CrossRef]
  78. Li, C.; Cui, L.; Yang, Y.; Miao, J.; Zhao, X.; Zhang, J.; Cui, G.; Zhang, Y. Gut Microbiota Differs Between Parkinson’s Disease Patients and Healthy Controls in Northeast China. Front. Mol. Neurosci. 2019, 12, 171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Vidal-Martinez, G.; Chin, B.; Camarillo, C.; Herrera, G.V.; Yang, B.; Sarosiek, I.; Perez, R.G. A Pilot Microbiota Study in Parkinson’s Disease Patients versus Control Subjects, and Effects of FTY720 and FTY720-Mitoxy Therapies in Parkinsonian and Multiple System Atrophy Mouse Models. J. Park. Dis. 2020, 10, 185–192. [Google Scholar] [CrossRef] [Green Version]
  80. Heintz-Buschart, A.; Pandey, U.; Wicke, T.; Sixel-Döring, F.; Janzen, A.; Sittig-Wiegand, E.; Trenkwalder, C.; Oertel, W.H.; Mollenhauer, B.; Wilmes, P. The Nasal and Gut Microbiome in Parkinson’s Disease and Idiopathic Rapid Eye Movement Sleep Behavior Disorder. Mov. Disord. 2018, 33, 88–98. [Google Scholar] [CrossRef] [Green Version]
  81. Zapała, B.; Stefura, T.; Wójcik-Pędziwiatr, M.; Kabut, R.; Bałajewicz-Nowak, M.; Milewicz, T.; Dudek, A.; Stój, A.; Rudzińska-Bar, M. Differences in the Composition of Gut Microbiota between Patients with Parkinson’s Disease and Healthy Controls: A Cohort Study. J. Clin. Med. 2021, 10, 5698. [Google Scholar] [CrossRef]
  82. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia Muciniphila Gen. Nov., Sp. Nov., a Human Intestinal Mucin-Degrading Bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the Human Gut Microbiome in Multiple Sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ganesh, B.P.; Klopfleisch, R.; Loh, G.; Blaut, M. Commensal Akkermansia Muciniphila Exacerbates Gut Inflammation in Salmonella Typhimurium-Infected Gnotobiotic Mice. PLoS ONE 2013, 8, e74963. [Google Scholar] [CrossRef]
  85. Amorim Neto, D.P.; Bosque, B.P.; Pereira de Godoy, J.V.; Rodrigues, P.V.; Meneses, D.D.; Tostes, K.; Costa Tonoli, C.C.; Faustino de Carvalho, H.; González-Billault, C.; de Castro Fonseca, M. Akkermansia Muciniphila Induces Mitochondrial Calcium Overload and α -Synuclein Aggregation in an Enteroendocrine Cell Line. iScience 2022, 25, 103908. [Google Scholar] [CrossRef]
  86. Vandeputte, D.; Falony, G.; Vieira-Silva, S.; Tito, R.Y.; Joossens, M.; Raes, J. Stool Consistency Is Strongly Associated with Gut Microbiota Richness and Composition, Enterotypes and Bacterial Growth Rates. Gut 2016, 65, 57–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Gobert, A.P.; Sagrestani, G.; Delmas, E.; Wilson, K.T.; Verriere, T.G.; Dapoigny, M.; Del’homme, C.; Bernalier-Donadille, A. The Human Intestinal Microbiota of Constipated-Predominant Irritable Bowel Syndrome Patients Exhibits Anti-Inflammatory Properties. Sci. Rep. 2016, 6, 39399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Skrzydło-Radomańska, B.; Prozorow-Król, B.; Cichoż-Lach, H.; Majsiak, E.; Bierła, J.B.; Kosikowski, W.; Szczerbiński, M.; Gantzel, J.; Cukrowska, B. The Effectiveness of Synbiotic Preparation Containing Lactobacillus and Bifidobacterium Probiotic Strains and Short Chain Fructooligosaccharides in Patients with Diarrhea Predominant Irritable Bowel Syndrome—A Randomized Double-Blind, Placebo-Controlled Study. Nutrients 2020, 12, 1999. [Google Scholar] [CrossRef]
  89. Minato, T.; Maeda, T.; Fujisawa, Y.; Tsuji, H.; Nomoto, K.; Ohno, K.; Hirayama, M. Progression of Parkinson’s Disease Is Associated with Gut Dysbiosis: Two-Year Follow-up Study. PLoS ONE 2017, 12, e0187307. [Google Scholar] [CrossRef] [Green Version]
  90. Bolliri, C.; Fontana, A.; Cereda, E.; Barichella, M.; Cilia, R.; Ferri, V.; Caronni, S.; Calandrella, D.; Morelli, L.; Pezzoli, G. Gut Microbiota in Monozygotic Twins Discordant for Parkinson’s Disease. Ann. Neurol. 2022, 92, 631–636. [Google Scholar] [CrossRef]
  91. Porras, A.M.; Shi, Q.; Zhou, H.; Callahan, R.; Montenegro-Bethancourt, G.; Solomons, N.; Brito, I.L. Geographic Differences in Gut Microbiota Composition Impact Susceptibility to Enteric Infection. Cell Rep. 2021, 36, 109457. [Google Scholar] [CrossRef]
  92. Gupta, V.K.; Paul, S.; Dutta, C. Geography, Ethnicity or Subsistence-Specific Variations in Human Microbiome Composition and Diversity. Front. Microbiol. 2017, 8, 1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhuang, Z.-Q.; Shen, L.-L.; Li, W.-W.; Fu, X.; Zeng, F.; Gui, L.; Lü, Y.; Cai, M.; Zhu, C.; Tan, Y.-L.; et al. Gut Microbiota Is Altered in Patients with Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 63, 1337–1346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Fan, Y.; Pedersen, O. Gut Microbiota in Human Metabolic Health and Disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef] [PubMed]
  95. Martínez-Oca, P.; Robles-Vera, I.; Sánchez-Roncero, A.; Escrivá, F.; Pérez-Vizcaíno, F.; Duarte, J.; Álvarez, C.; Fernández-Millán, E. Gut DYSBIOSIS and Altered Barrier Function Precedes the Appearance of Metabolic Syndrome in a Rat Model of Nutrient-Induced Catch-up Growth. J. Nutr. Biochem. 2020, 81, 108383. [Google Scholar] [CrossRef] [PubMed]
  96. Hertel, J.; Harms, A.C.; Heinken, A.; Baldini, F.; Thinnes, C.C.; Glaab, E.; Vasco, D.A.; Pietzner, M.; Stewart, I.D.; Wareham, N.J.; et al. Integrated Analyses of Microbiome and Longitudinal Metabolome Data Reveal Microbial-Host Interactions on Sulfur Metabolism in Parkinson’s Disease. Cell Rep. 2019, 29, 1767–1777.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Pereira, P.A.B.; Trivedi, D.K.; Silverman, J.; Duru, I.C.; Paulin, L.; Auvinen, P.; Scheperjans, F. Multiomics Implicate Gut Microbiota in Altered Lipid and Energy Metabolism in Parkinson’s Disease. NPJ Park. Dis. 2022, 8, 39. [Google Scholar] [CrossRef]
  98. Li, P.; Killinger, B.A.; Ensink, E.; Beddows, I.; Yilmaz, A.; Lubben, N.; Lamp, J.; Schilthuis, M.; Vega, I.E.; Woltjer, R.; et al. Gut Microbiota Dysbiosis Is Associated with Elevated Bile Acids in Parkinson’s Disease. Metabolites 2021, 11, 29. [Google Scholar] [CrossRef]
  99. Rosario, D.; Bidkhori, G.; Lee, S.; Bedarf, J.; Hildebrand, F.; le Chatelier, E.; Uhlen, M.; Ehrlich, S.D.; Proctor, G.; Wüllner, U.; et al. Systematic Analysis of Gut Microbiome Reveals the Role of Bacterial Folate and Homocysteine Metabolism in Parkinson’s Disease. Cell Rep. 2021, 34, 108807. [Google Scholar] [CrossRef]
  100. Romo-Vaquero, M.; Fernández-Villalba, E.; Gil-Martinez, A.-L.; Cuenca-Bermejo, L.; Espín, J.C.; Herrero, M.T.; Selma, M.V. Urolithins: Potential Biomarkers of Gut Dysbiosis and Disease Stage in Parkinson’s Patients. Food Funct. 2022, 13, 6306–6316. [Google Scholar] [CrossRef]
  101. Cirstea, M.S.; Yu, A.C.; Golz, E.; Sundvick, K.; Kliger, D.; Radisavljevic, N.; Foulger, L.H.; Mackenzie, M.; Huan, T.; Finlay, B.B.; et al. Microbiota Composition and Metabolism Are Associated With Gut Function in Parkinson’s Disease. Mov. Disord. 2020, 35, 1208–1217. [Google Scholar] [CrossRef]
  102. Yan, Z.; Yang, F.; Cao, J.; Ding, W.; Yan, S.; Shi, W.; Wen, S.; Yao, L. Alterations of Gut Microbiota and Metabolome with Parkinson’s Disease. Microb Pathog. 2021, 160, 105187. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, S.-J.; Chen, C.-C.; Liao, H.-Y.; Wu, Y.-W.; Liou, J.-M.; Wu, M.-S.; Kuo, C.-H.; Lin, C.-H. Alteration of Gut Microbial Metabolites in the Systemic Circulation of Patients with Parkinson’s Disease. J. Park. Dis. 2022, 12, 1219–1230. [Google Scholar] [CrossRef] [PubMed]
  104. Voigt, R.M.; Wang, Z.; Brown, J.M.; Engen, P.A.; Naqib, A.; Goetz, C.G.; Hall, D.A.; Metman, L.V.; Shaikh, M.; Forsyth, C.B.; et al. Gut Microbial Metabolites in Parkinson’s Disease: Association with Lifestyle, Disease Characteristics, and Treatment Status. Neurobiol. Dis. 2022, 170, 105780. [Google Scholar] [CrossRef]
  105. Arrona Cardoza, P.; Spillane, M.B.; Morales Marroquin, E. Alzheimer’s Disease and Gut Microbiota: Does Trimethylamine N-Oxide (TMAO) Play a Role? Nutr. Rev. 2022, 80, 271–281. [Google Scholar] [CrossRef] [PubMed]
  106. Kritsilis, M.; Rizou, S.V.; Koutsoudaki, P.; Evangelou, K.; Gorgoulis, V.; Papadopoulos, D. Ageing, Cellular Senescence and Neurodegenerative Disease. Int. J. Mol. Sci. 2018, 19, 2937. [Google Scholar] [CrossRef] [Green Version]
  107. Li, D.; Ke, Y.; Zhan, R.; Liu, C.; Zhao, M.; Zeng, A.; Shi, X.; Ji, L.; Cheng, S.; Pan, B.; et al. Trimethylamine-N-Oxide Promotes Brain Aging and Cognitive Impairment in Mice. Aging Cell 2018, 17, e12768. [Google Scholar] [CrossRef] [PubMed]
  108. Unger, M.M.; Spiegel, J.; Dillmann, K.-U.; Grundmann, D.; Philippeit, H.; Bürmann, J.; Faßbender, K.; Schwiertz, A.; Schäfer, K.-H. Short Chain Fatty Acids and Gut Microbiota Differ between Patients with Parkinson’s Disease and Age-Matched Controls. Park. Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
  109. Huang, T.; Shi, H.; Xu, Y.; Ji, L. The Gut Microbiota Metabolite Propionate Ameliorates Intestinal Epithelial Barrier Dysfunction-Mediated Parkinson’s Disease via the AKT Signaling Pathway. Neuroreport 2021, 32, 244–251. [Google Scholar] [CrossRef] [PubMed]
  110. Wu, G.; Jiang, Z.; Pu, Y.; Chen, S.; Wang, T.; Wang, Y.; Xu, X.; Wang, S.; Jin, M.; Yao, Y.; et al. Serum Short-Chain Fatty Acids and Its Correlation with Motor and Non-Motor Symptoms in Parkinson’s Disease Patients. BMC Neurol. 2022, 22, 13. [Google Scholar] [CrossRef]
  111. Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host Microbiota Constantly Control Maturation and Function of Microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
  112. Ostendorf, F.; Metzdorf, J.; Gold, R.; Haghikia, A.; Tönges, L. Propionic Acid and Fasudil as Treatment against Rotenone Toxicity in an In Vitro Model of Parkinson’s Disease. Molecules 2020, 25, 2502. [Google Scholar] [CrossRef] [PubMed]
  113. Patnala, R.; Arumugam, T.V.; Gupta, N.; Dheen, S.T. HDAC Inhibitor Sodium Butyrate-Mediated Epigenetic Regulation Enhances Neuroprotective Function of Microglia During Ischemic Stroke. Mol. Neurobiol. 2017, 54, 6391–6411. [Google Scholar] [CrossRef] [PubMed]
  114. Rane, P.; Shields, J.; Heffernan, M.; Guo, Y.; Akbarian, S.; King, J.A. The Histone Deacetylase Inhibitor, Sodium Butyrate, Alleviates Cognitive Deficits in Pre-Motor Stage PD. Neuropharmacology 2012, 62, 2409–2412. [Google Scholar] [CrossRef] [PubMed]
  115. Jin, H.; Kanthasamy, A.; Harischandra, D.S.; Kondru, N.; Ghosh, A.; Panicker, N.; Anantharam, V.; Rana, A.; Kanthasamy, A.G. Histone Hyperacetylation Up-Regulates Protein Kinase Cδ in Dopaminergic Neurons to Induce Cell Death. J. Biol. Chem. 2014, 289, 34743–34767. [Google Scholar] [CrossRef] [Green Version]
  116. Qiao, C.-M.; Sun, M.-F.; Jia, X.-B.; Li, Y.; Zhang, B.-P.; Zhao, L.-P.; Shi, Y.; Zhou, Z.-L.; Zhu, Y.-L.; Cui, C.; et al. Sodium Butyrate Exacerbates Parkinson’s Disease by Aggravating Neuroinflammation and Colonic Inflammation in MPTP-Induced Mice Model. Neurochem. Res. 2020, 45, 2128–2142. [Google Scholar] [CrossRef] [PubMed]
  117. van IJzendoorn, S.C.D.; Derkinderen, P. The Intestinal Barrier in Parkinson’s Disease: Current State of Knowledge. J. Park. Dis. 2019, 9, S323–S329. [Google Scholar] [CrossRef] [Green Version]
  118. Groschwitz, K.R.; Hogan, S.P. Intestinal Barrier Function: Molecular Regulation and Disease Pathogenesis. J. Allergy Clin. Immunol. 2009, 124, 3–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the Immune System. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef]
  120. Stadlbauer, V.; Engertsberger, L.; Komarova, I.; Feldbacher, N.; Leber, B.; Pichler, G.; Fink, N.; Scarpatetti, M.; Schippinger, W.; Schmidt, R.; et al. Dysbiosis, Gut Barrier Dysfunction and Inflammation in Dementia: A Pilot Study. BMC Geriatr. 2020, 20, 248. [Google Scholar] [CrossRef] [PubMed]
  121. Michielan, A.; D’Incà, R. Intestinal Permeability in Inflammatory Bowel Disease: Pathogenesis, Clinical Evaluation, and Therapy of Leaky Gut. Mediat. Inflamm 2015, 2015, 628157. [Google Scholar] [CrossRef] [PubMed]
  122. Hanning, N.; Edwinson, A.L.; Ceuleers, H.; Peters, S.A.; de Man, J.G.; Hassett, L.C.; de Winter, B.Y.; Grover, M. Intestinal Barrier Dysfunction in Irritable Bowel Syndrome: A Systematic Review. Therap. Adv. Gastroenterol. 2021, 14, 175628482199358. [Google Scholar] [CrossRef] [PubMed]
  123. Baizabal-Carvallo, J.F.; Alonso-Juarez, M. The Link between Gut Dysbiosis and Neuroinflammation in Parkinson’s Disease. Neuroscience 2020, 432, 160–173. [Google Scholar] [CrossRef] [PubMed]
  124. Anderson, G.; Seo, M.; Berk, M.; Carvalho, A.; Maes, M. Gut Permeability and Microbiota in Parkinson’s Disease: Role of Depression, Tryptophan Catabolites, Oxidative and Nitrosative Stress and Melatonergic Pathways. Curr. Pharm. Des. 2016, 22, 6142–6151. [Google Scholar] [CrossRef]
  125. Seguella, L.; Sarnelli, G.; Esposito, G. Leaky Gut, Dysbiosis, and Enteric Glia Activation: The Trilogy behind the Intestinal Origin of Parkinson’s Disease. Neural Regen. Res. 2020, 15, 1037. [Google Scholar] [CrossRef]
  126. Tan, A.H.; Lim, S.Y.; Lang, A.E. The Microbiome–Gut–Brain Axis in Parkinson Disease—from Basic Research to the Clinic. Nat. Rev. Neurol. 2022, 18, 476–495. [Google Scholar] [CrossRef]
  127. Loffredo, L.; Ettorre, E.; Zicari, A.M.; Inghilleri, M.; Nocella, C.; Perri, L.; Spalice, A.; Fossati, C.; de Lucia, M.C.; Pigozzi, F.; et al. Oxidative Stress and Gut-Derived Lipopolysaccharides in Neurodegenerative Disease: Role of NOX2. Oxid Med. Cell Longev. 2020, 2020, 8630275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Drobny, A.; Ngo, P.A.; Neurath, M.F.; Zunke, F.; López-Posadas, R. Molecular Communication Between Neuronal Networks and Intestinal Epithelial Cells in Gut Inflammation and Parkinson’s Disease. Front. Med. 2021, 8, 8630275. [Google Scholar] [CrossRef]
  129. Pellegrini, C.; D’Antongiovanni, V.; Miraglia, F.; Rota, L.; Benvenuti, L.; di Salvo, C.; Testa, G.; Capsoni, S.; Carta, G.; Antonioli, L.; et al. Enteric α-Synuclein Impairs Intestinal Epithelial Barrier through Caspase-1-Inflammasome Signaling in Parkinson’s Disease before Brain Pathology. NPJ Park. Dis. 2022, 8, 9. [Google Scholar] [CrossRef]
  130. Clairembault, T.; Leclair-Visonneau, L.; Coron, E.; Bourreille, A.; le Dily, S.; Vavasseur, F.; Heymann, M.-F.; Neunlist, M.; Derkinderen, P. Structural Alterations of the Intestinal Epithelial Barrier in Parkinson’s Disease. Acta Neuropathol. Commun. 2015, 3, 12. [Google Scholar] [CrossRef] [Green Version]
  131. Gorecki, A.M.; Preskey, L.; Bakeberg, M.C.; Kenna, J.E.; Gildenhuys, C.; MacDougall, G.; Dunlop, S.A.; Mastaglia, F.L.; Akkari, P.A.; Koengten, F.; et al. Altered Gut Microbiome in Parkinson’s Disease and the Influence of Lipopolysaccharide in a Human α-Synuclein Over-Expressing Mouse Model. Front. Neurosci. 2019, 13, 839. [Google Scholar] [CrossRef]
  132. Schwiertz, A.; Spiegel, J.; Dillmann, U.; Grundmann, D.; Bürmann, J.; Faßbender, K.; Schäfer, K.-H.; Unger, M.M. Fecal Markers of Intestinal Inflammation and Intestinal Permeability Are Elevated in Parkinson’s Disease. Park. Relat. Disord. 2018, 50, 104–107. [Google Scholar] [CrossRef]
  133. Dumitrescu, L.; Marta, D.; Dănău, A.; Lefter, A.; Tulbă, D.; Cozma, L.; Manole, E.; Gherghiceanu, M.; Ceafalan, L.C.; Popescu, B.O. Serum and Fecal Markers of Intestinal Inflammation and Intestinal Barrier Permeability Are Elevated in Parkinson’s Disease. Front. Neurosci. 2021, 15, 689723. [Google Scholar] [CrossRef]
  134. Mulak, A.; Koszewicz, M.; Panek-Jeziorna, M.; Koziorowska-Gawron, E.; Budrewicz, S. Fecal Calprotectin as a Marker of the Gut Immune System Activation Is Elevated in Parkinson’s Disease. Front. Neurosci. 2019, 13, 992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Yang, X.; Ai, P.; He, X.; Mo, C.; Zhang, Y.; Xu, S.; Lai, Y.; Qian, Y.; Xiao, Q. Parkinson’s Disease Is Associated with Impaired Gut–Blood Barrier for Short-Chain Fatty Acids. Mov. Disord. 2022, 37, 1634–1643. [Google Scholar] [CrossRef]
  136. Schoultz, I.; Keita, Å.V. The Intestinal Barrier and Current Techniques for the Assessment of Gut Permeability. Cells 2020, 9, 1909. [Google Scholar] [CrossRef]
  137. Mishra, A.; Makharia, G.K. Techniques of Functional and Motility Test: How to Perform and Interpret Intestinal Permeability. J. Neurogastroenterol. Motil 2012, 18, 443–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Salat-Foix, D.; Tran, K.; Ranawaya, R.; Meddings, J.; Suchowersky, O. Increased Intestinal Permeability and Parkinson Disease Patients: Chicken or Egg? Can. J. Neurol. Sci. / J. Can. Des Sci. Neurol. 2012, 39, 185–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Davies, K.N.; King, D.; Billington, D.; Barrett, J.A. Intestinal Permeability and Orocaecal Transit Time in Elderly Patients with Parkinson’s Disease. Postgrad. Med. J. 1996, 72, 164–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Feng, X.-Y.; Yan, J.-T.; Zhang, X.-L.; Zhu, J.-X. Gastrointestinal Non-Motor Dysfunction in Parkinson’s Disease Model Rats with 6-Hydroxydopamine. Physiol. Res. 2019, 68, 295–303. [Google Scholar] [CrossRef]
  141. Jackson, A.; Engen, P.A.; Forsyth, C.B.; Shaikh, M.; Naqib, A.; Wilber, S.; Frausto, D.M.; Raeisi, S.; Green, S.J.; Bradaric, B.D.; et al. Intestinal Barrier Dysfunction in the Absence of Systemic Inflammation Fails to Exacerbate Motor Dysfunction and Brain Pathology in a Mouse Model of Parkinson’s Disease. Front. Neurol. 2022, 13, 689723. [Google Scholar] [CrossRef]
  142. Mohammad, S.; Thiemermann, C. Role of Metabolic Endotoxemia in Systemic Inflammation and Potential Interventions. Front. Immunol. 2021, 11, 594150. [Google Scholar] [CrossRef]
  143. Bhattacharyya, D.; Mohite, G.M.; Krishnamoorthy, J.; Gayen, N.; Mehra, S.; Navalkar, A.; Kotler, S.A.; Ratha, B.N.; Ghosh, A.; Kumar, R.; et al. Lipopolysaccharide from Gut Microbiota Modulates α-Synuclein Aggregation and Alters Its Biological Function. ACS Chem. Neurosci. 2019, 10, 2229–2236. [Google Scholar] [CrossRef] [PubMed]
  144. Qin, L.; Wu, X.; Block, M.L.; Liu, Y.; Breese, G.R.; Hong, J.-S.; Knapp, D.J.; Crews, F.T. Systemic LPS Causes Chronic Neuroinflammation and Progressive Neurodegeneration. Glia 2007, 55, 453–462. [Google Scholar] [CrossRef] [Green Version]
  145. Hasegawa, S.; Goto, S.; Tsuji, H.; Okuno, T.; Asahara, T.; Nomoto, K.; Shibata, A.; Fujisawa, Y.; Minato, T.; Okamoto, A.; et al. Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson’s Disease. PLoS ONE 2015, 10, e0142164. [Google Scholar] [CrossRef] [Green Version]
  146. Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased Intestinal Permeability Correlates with Sigmoid Mucosa Alpha-Synuclein Staining and Endotoxin Exposure Markers in Early Parkinson’s Disease. PLoS ONE 2011, 6, e28032. [Google Scholar] [CrossRef] [Green Version]
  147. Pal, G.D.; Shaikh, M.; Forsyth, C.B.; Ouyang, B.; Keshavarzian, A.; Shannon, K.M. Abnormal Lipopolysaccharide Binding Protein as Marker of Gastrointestinal Inflammation in Parkinson Disease. Front. Neurosci. 2015, 9, 306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Gutsmann, T.; Müller, M.; Carroll, S.F.; MacKenzie, R.C.; Wiese, A.; Seydel, U. Dual Role of Lipopolysaccharide (LPS)-Binding Protein in Neutralization of LPS and Enhancement of LPS-Induced Activation of Mononuclear Cells. Infect Immun. 2001, 69, 6942–6950. [Google Scholar] [CrossRef] [Green Version]
  149. Minter, R.M.; Bi, X.; Ben-Josef, G.; Wang, T.; Hu, B.; Arbabi, S.; Hemmila, M.R.; Wang, S.C.; Remick, D.G.; Su, G.L. LPS-Binding Protein Mediates LPS-Induced Liver Injury and Mortality in the Setting of Biliary Obstruction. Am. J. Physiol.-Gastrointest. Liver Physiol. 2009, 296, G45–G54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. 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. Neuroinflamm. 2019, 16, 129. [Google Scholar] [CrossRef]
  151. Su, Y.; Liu, N.; Zhang, Z.; Li, H.; Ma, J.; Yuan, Y.; Shi, M.; Liu, J.; Zhao, Z.; Zhang, Z.; et al. Cholecystokinin and Glucagon-like Peptide-1 Analogues Regulate Intestinal Tight Junction, Inflammation, Dopaminergic Neurons and α-Synuclein Accumulation in the Colon of Two Parkinson’s Disease Mouse Models. Eur. J. Pharmacol. 2022, 926, 175029. [Google Scholar] [CrossRef]
  152. Tasselli, M.; Chaumette, T.; Paillusson, S.; Monnet, Y.; Lafoux, A.; Huchet-Cadiou, C.; Aubert, P.; Hunot, S.; Derkinderen, P.; Neunlist, M. Effects of Oral Administration of Rotenone on Gastrointestinal Functions in Mice. Neurogastroenterol. Motil. 2013, 25, e183–e193. [Google Scholar] [CrossRef] [PubMed]
  153. He, R.; Yan, X.; Guo, J.; Xu, Q.; Tang, B.; Sun, Q. Recent Advances in Biomarkers for Parkinson’s Disease. Front. Aging Neurosci. 2018, 10, 305. [Google Scholar] [CrossRef]
  154. Li, T.; Le, W. Biomarkers for Parkinson’s Disease: How Good Are They? Neurosci. Bull 2020, 36, 183–194. [Google Scholar] [CrossRef]
  155. Delenclos, M.; Jones, D.R.; McLean, P.J.; Uitti, R.J. Biomarkers in Parkinson’s Disease: Advances and Strategies. Park. Relat. Disord. 2016, 22, S106–S110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Farrokhi, V.; Nemati, R.; Nichols, F.C.; Yao, X.; Anstadt, E.; Fujiwara, M.; Grady, J.; Wakefield, D.; Castro, W.; Donaldson, J.; et al. Bacterial Lipodipeptide, Lipid 654, Is a Microbiome-Associated Biomarker for Multiple Sclerosis. Clin. Transl. Immunol. 2013, 2, e8. [Google Scholar] [CrossRef] [PubMed]
  157. Qian, Y.; Yang, X.; Xu, S.; Huang, P.; Li, B.; Du, J.; He, Y.; Su, B.; Xu, L.-M.; Wang, L.; et al. Gut Metagenomics-Derived Genes as Potential Biomarkers of Parkinson’s Disease. Brain 2020, 143, 2474–2489. [Google Scholar] [CrossRef] [PubMed]
  158. Jones, J.D.; Rahmani, E.; Garcia, E.; Jacobs, J.P. Gastrointestinal Symptoms Are Predictive of Trajectories of Cognitive Functioning in de Novo Parkinson’s Disease. Park. Relat. Disord. 2020, 72, 7–12. [Google Scholar] [CrossRef]
  159. Fricova, D.; Harsanyiova, J.; Kralova Trancikova, A. Alpha-Synuclein in the Gastrointestinal Tract as a Potential Biomarker for Early Detection of Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 8666. [Google Scholar] [CrossRef]
  160. Cilia, R.; Piatti, M.; Cereda, E.; Bolliri, C.; Caronni, S.; Ferri, V.; Cassani, E.; Bonvegna, S.; Ferrarese, C.; Zecchinelli, A.L.; et al. Does Gut Microbiota Influence the Course of Parkinson’s Disease? A 3-Year Prospective Exploratory Study in de Novo Patients. J. Park. Dis. 2021, 11, 159–170. [Google Scholar] [CrossRef]
  161. Pietrucci, D.; Teofani, A.; Unida, V.; Cerroni, R.; Biocca, S.; Stefani, A.; Desideri, A. Can Gut Microbiota Be a Good Predictor for Parkinson’s Disease? A Machine Learning Approach. Brain Sci. 2020, 10, 242. [Google Scholar] [CrossRef]
  162. Guo, X.; Tang, P.; Hou, C.; Chong, L.; Zhang, X.; Liu, P.; Chen, L.; Liu, Y.; Zhang, L.; Li, R. Integrated Microbiome and Host Transcriptome Profiles Link Parkinson’s Disease to Blautia Genus: Evidence From Feces, Blood, and Brain. Front. Microbiol. 2022, 13. [Google Scholar] [CrossRef]
  163. Lubomski, M.; Xu, X.; Holmes, A.J.; Muller, S.; Yang, J.Y.H.; Davis, R.L.; Sue, C.M. Nutritional Intake and Gut Microbiome Composition Predict Parkinson’s Disease. Front. Aging Neurosci. 2022, 14. [Google Scholar] [CrossRef]
  164. Chung, S.J.; Rim, J.H.; Ji, D.; Lee, S.; Yoo, H.S.; Jung, J.H.; Baik, K.; Choi, Y.; Ye, B.S.; Sohn, Y.H.; et al. Gut Microbiota-Derived Metabolite Trimethylamine N-Oxide as a Biomarker in Early Parkinson’s Disease. Nutrition 2021, 83, 111090. [Google Scholar] [CrossRef]
  165. He, X.; Qian, Y.; Xu, S.; Zhang, Y.; Mo, C.; Guo, W.; Yang, X.; Xiao, Q. Plasma Short-Chain Fatty Acids Differences in Multiple System Atrophy from Parkinson’s Disease. J. Park. Dis. 2021, 11, 1167–1176. [Google Scholar] [CrossRef]
  166. Nishiwaki, H.; Ito, M.; Hamaguchi, T.; Maeda, T.; Kashihara, K.; Tsuboi, Y.; Ueyama, J.; Yoshida, T.; Hanada, H.; Takeuchi, I.; et al. Short Chain Fatty Acids-Producing and Mucin-Degrading Intestinal Bacteria Predict the Progression of Early Parkinson’s Disease. NPJ Park. Dis. 2022, 8, 65. [Google Scholar] [CrossRef]
  167. Cerroni, R.; Pietrucci, D.; Teofani, A.; Chillemi, G.; Liguori, C.; Pierantozzi, M.; Unida, V.; Selmani, S.; Mercuri, N.B.; Stefani, A. Not Just a Snapshot: An Italian Longitudinal Evaluation of Stability of Gut Microbiota Findings in Parkinson’s Disease. Brain Sci. 2022, 12, 739. [Google Scholar] [CrossRef]
  168. Plassais, J.; Gbikpi-Benissan, G.; Figarol, M.; Scheperjans, F.; Gorochov, G.; Derkinderen, P.; Cervino, A.C.L. Gut Microbiome Alpha-Diversity Is Not a Marker of Parkinson’s Disease and Multiple Sclerosis. Brain Commun. 2021, 3, fcab113. [Google Scholar] [CrossRef]
  169. Boertien, J.M.; van der Zee, S.; Chrysou, A.; Gerritsen, M.J.J.; Jansonius, N.M.; Spikman, J.M.; van Laar, T.; Verwey, N.A.; van Harten, B.; Portman, A.T.; et al. Study Protocol of the DUtch PARkinson Cohort (DUPARC): A Prospective, Observational Study of de Novo Parkinson’s Disease Patients for the Identification and Validation of Biomarkers for Parkinson’s Disease Subtypes, Progression and Pathophysiology. BMC Neurol. 2020, 20, 245. [Google Scholar] [CrossRef]
  170. Falony, G.; Joossens, M.; Vieira-Silva, S.; Wang, J.; Darzi, Y.; Faust, K.; Kurilshikov, A.; Bonder, M.J.; Valles-Colomer, M.; Vandeputte, D.; et al. Population-Level Analysis of Gut Microbiome Variation. Science (1979) 2016, 352, 560–564. [Google Scholar] [CrossRef]
  171. Zhernakova, A.; Kurilshikov, A.; Bonder, M.J.; Tigchelaar, E.F.; Schirmer, M.; Vatanen, T.; Mujagic, Z.; Vila, A.V.; Falony, G.; Vieira-Silva, S.; et al. Population-Based Metagenomics Analysis Reveals Markers for Gut Microbiome Composition and Diversity. Science (1979) 2016, 352, 565–569. [Google Scholar] [CrossRef] [PubMed]
  172. Collins, S.L.; Patterson, A.D. The Gut Microbiome: An Orchestrator of Xenobiotic Metabolism. Acta Pharm. Sin. B 2020, 10, 19–32. [Google Scholar] [CrossRef] [PubMed]
  173. Li, H.; He, J.; Jia, W. The Influence of Gut Microbiota on Drug Metabolism and Toxicity. Expert Opin. Drug Metab. Toxicol. 2016, 12, 31–40. [Google Scholar] [CrossRef] [Green Version]
  174. Vich Vila, A.; Collij, V.; Sanna, S.; Sinha, T.; Imhann, F.; Bourgonje, A.R.; Mujagic, Z.; Jonkers, D.M.A.E.; Masclee, A.A.M.; Fu, J.; et al. Impact of Commonly Used Drugs on the Composition and Metabolic Function of the Gut Microbiota. Nat. Commun. 2020, 11, 362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Brüssow, H. Parkinson Disease, Levodopa and the Gut Microbiota—When Microbiology Meets Pharmacology. Environ. Microbiol. 2020, 22, 808–812. [Google Scholar] [CrossRef] [PubMed]
  176. Keshavarzian, A.; Engen, P.; Bonvegna, S.; Cilia, R. The Gut Microbiome in Parkinson’s Disease: A Culprit or a Bystander? Prog. Brain Res. 2020, 252, 357–450. [Google Scholar]
  177. Zhang, X.; Han, Y.; Huang, W.; Jin, M.; Gao, Z. The Influence of the Gut Microbiota on the Bioavailability of Oral Drugs. Acta Pharm. Sin. B 2021, 11, 1789–1812. [Google Scholar] [CrossRef]
  178. Lubomski, M.; Xu, X.; Holmes, A.J.; Yang, J.Y.H.; Sue, C.M.; Davis, R.L. The Impact of Device-Assisted Therapies on the Gut Microbiome in Parkinson’s Disease. J. Neurol. 2022, 269, 780–795. [Google Scholar] [CrossRef]
  179. Salat, D.; Tolosa, E. Levodopa in the Treatment of Parkinson’s Disease: Current Status and New Developments. J. Park. Dis. 2013, 3, 255–269. [Google Scholar] [CrossRef] [Green Version]
  180. van Kessel, S.P.; Frye, A.K.; El-Gendy, A.O.; Castejon, M.; Keshavarzian, A.; van Dijk, G.; el Aidy, S. Gut Bacterial Tyrosine Decarboxylases Restrict Levels of Levodopa in the Treatment of Parkinson’s Disease. Nat. Commun. 2019, 10, 310. [Google Scholar] [CrossRef] [Green Version]
  181. Maini Rekdal, V.; Bess, E.N.; Bisanz, J.E.; Turnbaugh, P.J.; Balskus, E.P. Discovery and Inhibition of an Interspecies Gut Bacterial Pathway for Levodopa Metabolism. Science (1979) 2019, 364, eaau6323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Jameson, K.G.; Hsiao, E.Y. A Novel Pathway for Microbial Metabolism of Levodopa. Nat. Med. 2019, 25, 1195–1197. [Google Scholar] [CrossRef]
  183. Hitchings, R.; Kelly, L. Drug Metabolism as a Community Effort. Cell Metab. 2019, 30, 235–237. [Google Scholar] [CrossRef]
  184. van Kessel, S.P.; de Jong, H.R.; Winkel, S.L.; van Leeuwen, S.S.; Nelemans, S.A.; Permentier, H.; Keshavarzian, A.; el Aidy, S. Gut Bacterial Deamination of Residual Levodopa Medication for Parkinson’s Disease. BMC Biol. 2020, 18, 137. [Google Scholar] [CrossRef] [PubMed]
  185. Weis, S.; Schwiertz, A.; Unger, M.M.; Becker, A.; Faßbender, K.; Ratering, S.; Kohl, M.; Schnell, S.; Schäfer, K.-H.; Egert, M. Effect of Parkinson’s Disease and Related Medications on the Composition of the Fecal Bacterial Microbiota. NPJ Park. Dis. 2019, 5, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Melis, M.; Vascellari, S.; Santoru, M.L.; Oppo, V.; Fabbri, M.; Sarchioto, M.; Murgia, D.; Zibetti, M.; Lopiano, L.; Serra, A.; et al. Gut Microbiota and Metabolome Distinctive Features in Parkinson Disease: Focus on Levodopa and Levodopa-carbidopa Intrajejunal Gel. Eur. J. Neurol. 2021, 28, 1198–1209. [Google Scholar] [CrossRef] [PubMed]
  187. Palacios, N.; Hannoun, A.; Flahive, J.; Ward, D.; Goostrey, K.; Deb, A.; Smith, K.M. Effect of Levodopa Initiation on the Gut Microbiota in Parkinson’s Disease. Front. Neurol. 2021, 12, 574529. [Google Scholar] [CrossRef]
  188. van Kessel, S.P.; Auvinen, P.; Scheperjans, F.; el Aidy, S. Gut Bacterial Tyrosine Decarboxylase Associates with Clinical Variables in a Longitudinal Cohort Study of Parkinsons Disease. NPJ Park. Dis. 2021, 7, 115. [Google Scholar] [CrossRef]
  189. Bedarf, J.R.; Hildebrand, F.; Coelho, L.P.; Sunagawa, S.; Bahram, M.; Goeser, F.; Bork, P.; Wüllner, U. Functional Implications of Microbial and Viral Gut Metagenome Changes in Early Stage L-DOPA-Naïve Parkinson’s Disease Patients. Genome Med. 2017, 9, 39. [Google Scholar] [CrossRef] [Green Version]
  190. Chen, Z.-J.; Liang, C.-Y.; Yang, L.-Q.; Ren, S.-M.; Xia, Y.-M.; Cui, L.; Li, X.-F.; Gao, B.-L. Association of Parkinson’s Disease With Microbes and Microbiological Therapy. Front. Cell Infect. Microbiol. 2021, 11, 619354. [Google Scholar] [CrossRef]
  191. van Kessel, S.P.; Bullock, A.; van Dijk, G.; el Aidy, S. Parkinson’s Disease Medication Alters Small Intestinal Motility and Microbiota Composition in Healthy Rats. mSystems 2022, 7. [Google Scholar] [CrossRef] [PubMed]
  192. Gray, R.; Patel, S.; Ives, N.; Rick, C.; Woolley, R.; Muzerengi, S.; Gray, A.; Jenkinson, C.; McIntosh, E.; Wheatley, K.; et al. Long-Term Effectiveness of Adjuvant Treatment With Catechol-O-Methyltransferase or Monoamine Oxidase B Inhibitors Compared With Dopamine Agonists Among Patients With Parkinson Disease Uncontrolled by Levodopa Therapy. JAMA Neurol. 2022, 79, 131. [Google Scholar] [CrossRef] [PubMed]
  193. Ness, J.; Hoth, A.; Barnett, M.J.; Shorr, R.I.; Kaboli, P.J. Anticholinergic Medications in Community-Dwelling Older Veterans: Prevalence of Anticholinergic Symptoms, Symptom Burden, and Adverse Drug Events. Am. J. Geriatr. Pharm. 2006, 4, 42–51. [Google Scholar] [CrossRef]
  194. Kaakkola, S. Clinical Pharmacology, Therapeutic Use and Potential of COMT Inhibitors in Parkinson’s Disease. Drugs 2000, 59, 1233–1250. [Google Scholar] [CrossRef] [PubMed]
  195. Scheperjans, F.; Aho, V.; Pereira, P.A.B.; Koskinen, K.; Paulin, L.; Pekkonen, E.; Haapaniemi, E.; Kaakkola, S.; Eerola-Rautio, J.; Pohja, M.; et al. Gut Microbiota Are Related to Parkinson’s Disease and Clinical Phenotype. Mov. Disord. 2015, 30, 350–358. [Google Scholar] [CrossRef]
  196. 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] [Green Version]
  197. 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] [Green Version]
  198. Gordin, A.; Kaakkola, S.; Teräväinen, H. Clinical Advantages of COMT Inhibition with Entacapone ? A Review. J. Neural. Transm. 2004, 111, 1343–1363. [Google Scholar] [CrossRef]
  199. Grün, D.; Zimmer, V.C.; Kauffmann, J.; Spiegel, J.; Dillmann, U.; Schwiertz, A.; Faßbender, K.; Fousse, M.; Unger, M.M. Impact of Oral COMT-Inhibitors on Gut Microbiota and Short Chain Fatty Acids in Parkinson’s Disease. Park. Relat. Disord. 2020, 70, 20–22. [Google Scholar] [CrossRef] [Green Version]
  200. Fu, S.-C.; Lee, C.-H.; Hsieh, Y.-C.; Wu, P.-H.; Lin, S.-H.; Wang, H. A Pilot Study Exploring the Association of Entacapone, Gut Microbiota, and the Subsequent Side Effects in Patients With Parkinson’s Disease. Front. Cell Infect. Microbiol. 2022, 12, 837019. [Google Scholar] [CrossRef]
  201. Enright, E.F.; Gahan, C.G.M.; Joyce, S.A.; Griffin, B.T. The Impact of the Gut Microbiota on Drug Metabolism and Clinical Outcome. Yale J. Biol. Med. 2016, 89, 375–382. [Google Scholar] [PubMed]
  202. Sun, C.; Chen, L.; Shen, Z. Mechanisms of Gastrointestinal Microflora on Drug Metabolism in Clinical Practice. Saudi Pharm. J. 2019, 27, 1146–1156. [Google Scholar] [CrossRef] [PubMed]
  203. Hou, Y.; Shan, C.; Zhuang, S.; Zhuang, Q.; Ghosh, A.; Zhu, K.; Kong, X.; Wang, S.; Gong, Y.; Yang, Y.; et al. Gut Microbiota-Derived Propionate Mediates the Neuroprotective Effect of Osteocalcin in a Mouse Model of Parkinson’s Disease. Microbiome 2021, 9, 34. [Google Scholar] [CrossRef]
  204. Wang, Y.; Tong, Q.; Ma, S.-R.; Zhao, Z.-X.; Pan, L.-B.; Cong, L.; Han, P.; Peng, R.; Yu, H.; Lin, Y.; et al. Oral Berberine Improves Brain Dopa/Dopamine Levels to Ameliorate Parkinson’s Disease by Regulating Gut Microbiota. Signal Transduct Target Ther. 2021, 6, 77. [Google Scholar] [CrossRef] [PubMed]
  205. Rothstein, J.D.; Patel, S.; Regan, M.R.; Haenggeli, C.; Huang, Y.H.; Bergles, D.E.; Jin, L.; Dykes Hoberg, M.; Vidensky, S.; Chung, D.S.; et al. β-Lactam Antibiotics Offer Neuroprotection by Increasing Glutamate Transporter Expression. Nature 2005, 433, 73–77. [Google Scholar] [CrossRef]
  206. Lipski, J.; Wan, C.K.; Bai, J.Z.; Pi, R.; Li, D.; Donnelly, D. Neuroprotective Potential of Ceftriaxone in in Vitro Models of Stroke. Neuroscience 2007, 146, 617–629. [Google Scholar] [CrossRef] [PubMed]
  207. Burton, A. Antibiotics for Neurological Ailments? Lancet Neurol. 2005, 4, 142. [Google Scholar] [CrossRef]
  208. Pradhan, S.; Madke, B.; Kabra, P.; Singh, A. Anti-Inflammatory and Immunomodulatory Effects of Antibiotics and Their Use in Dermatology. Indian J. Dermatol. 2016, 61, 469. [Google Scholar] [CrossRef] [PubMed]
  209. Stoilova, T.; Colombo, L.; Forloni, G.; Tagliavini, F.; Salmona, M. A New Face for Old Antibiotics: Tetracyclines in Treatment of Amyloidoses. J. Med. Chem. 2013, 56, 5987–6006. [Google Scholar] [CrossRef]
  210. Van Vuuren, M.J.; Nell, T.A.; Carr, J.A.; Kell, D.B.; Pretorius, E. Iron Dysregulation and Inflammagens Related to Oral and Gut Health Are Central to the Development of Parkinson’s Disease. Biomolecules 2020, 11, 30. [Google Scholar] [CrossRef] [PubMed]
  211. Bortolanza, M.; Nascimento, G.C.; Socias, S.B.; Ploper, D.; Chehín, R.N.; Raisman-Vozari, R.; Del-Bel, E. Tetracycline Repurposing in Neurodegeneration: Focus on Parkinson’s Disease. J. Neural. Transm. 2018, 125, 1403–1415. [Google Scholar] [CrossRef] [PubMed]
  212. Yadav, N.; Thakur, A.K.; Shekhar, N. Ayushi Potential of Antibiotics for the Treatment and Management of Parkinson’s Disease: An Overview. Curr. Drug Res. Rev. 2021, 13, 166–171. [Google Scholar] [CrossRef]
  213. Hurkacz, M.; Dobrek, L.; Wiela-Hojeńska, A. Antibiotics and the Nervous System—Which Face of Antibiotic Therapy Is Real, Dr. Jekyll (Neurotoxicity) or Mr. Hyde (Neuroprotection)? Molecules 2021, 26, 7456. [Google Scholar] [CrossRef] [PubMed]
  214. Balducci, C.; Forloni, G. Doxycycline for Alzheimer’s Disease: Fighting β-Amyloid Oligomers and Neuroinflammation. Front. Pharmacol. 2019, 10, 738. [Google Scholar] [CrossRef] [Green Version]
  215. Lorente-Picón, M.; Laguna, A. New Avenues for Parkinson’s Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021, 11, 433. [Google Scholar] [CrossRef]
  216. Pu, Y.; Chang, L.; Qu, Y.; Wang, S.; Zhang, K.; Hashimoto, K. Antibiotic-Induced Microbiome Depletion Protects against MPTP-Induced Dopaminergic Neurotoxicity in the Brain. Aging 2019, 11, 6915–6929. [Google Scholar] [CrossRef] [PubMed]
  217. Koutzoumis, D.N.; Vergara, M.; Pino, J.; Buddendorff, J.; Khoshbouei, H.; Mandel, R.J.; Torres, G.E. Alterations of the Gut Microbiota with Antibiotics Protects Dopamine Neuron Loss and Improve Motor Deficits in a Pharmacological Rodent Model of Parkinson’s Disease. Exp. Neurol. 2020, 325, 113159. [Google Scholar] [CrossRef]
  218. Cui, C.; Hong, H.; Shi, Y.; Zhou, Y.; Qiao, C.-M.; Zhao, W.-J.; Zhao, L.-P.; Wu, J.; Quan, W.; Niu, G.-Y.; et al. Vancomycin Pretreatment on MPTP-Induced Parkinson’s Disease Mice Exerts Neuroprotection by Suppressing Inflammation Both in Brain and Gut. J. Neuroimmune Pharmacol. 2022. [Google Scholar] [CrossRef]
  219. Baizabal-Carvallo, J.F.; Alonso-Juarez, M.; Fekete, R. Intestinal Decontamination Therapy for Dyskinesia and Motor Fluctuations in Parkinson’s Disease. Front. Neurol. 2021, 12, 729961. [Google Scholar] [CrossRef]
  220. Dănău, A.; Dumitrescu, L.; Lefter, A.; Tulbă, D.; Popescu, B.O. Small Intestinal Bacterial Overgrowth as Potential Therapeutic Target in Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 11663. [Google Scholar] [CrossRef] [PubMed]
  221. Ramprasad, C.; Douglas, J.Y.; Moshiree, B. Parkinson’s Disease and Current Treatments for Its Gastrointestinal Neurogastromotility Effects. Curr. Treat Options Gastroenterol 2018, 16, 489–510. [Google Scholar] [CrossRef] [PubMed]
  222. Fasano, A.; Bove, F.; Gabrielli, M.; Petracca, M.; Zocco, M.A.; Ragazzoni, E.; Barbaro, F.; Piano, C.; Fortuna, S.; Tortora, A.; et al. The Role of Small Intestinal Bacterial Overgrowth in Parkinson’s Disease. Mov. Disord. 2013, 28, 1241–1249. [Google Scholar] [CrossRef]
  223. Pekary, A.E.; Sattin, A. Rifaximin Modulates TRH and TRH-like Peptide Expression throughout the Brain and Peripheral Tissues of Male Rats. BMC Neurosci. 2022, 23, 9. [Google Scholar] [CrossRef]
  224. DiBaise, J.K.; Crowell, M.D.; Driver-Dunckley, E.; Mehta, S.H.; Hoffman-Snyder, C.; Lin, T.; Adler, C.H. Weight Loss in Parkinson’s Disease: No Evidence for Role of Small Intestinal Bacterial Overgrowth. J. Park. Dis. 2018, 8, 571–581. [Google Scholar] [CrossRef]
  225. Yimer, E.M.; Hishe, H.Z.; Tuem, K.B. Repurposing of the β-Lactam Antibiotic, Ceftriaxone for Neurological Disorders: A Review. Front. Neurosci. 2019, 13, 236. [Google Scholar] [CrossRef] [Green Version]
  226. Tai, C.-H.; Bellesi, M.; Chen, A.-C.; Lin, C.-L.; Li, H.-H.; Lin, P.-J.; Liao, W.-C.; Hung, C.-S.; Schwarting, R.K.; Ho, Y.-J. A New Avenue for Treating Neuronal Diseases: Ceftriaxone, an Old Antibiotic Demonstrating Behavioral Neuronal Effects. Behavioural. Brain Res. 2019, 364, 149–156. [Google Scholar] [CrossRef] [Green Version]
  227. Yamada, J.; Jinno, S. Alterations in Neuronal Survival and Glial Reactions after Axotomy by Ceftriaxone and Minocycline in the Mouse Hypoglossal Nucleus. Neurosci. Lett. 2011, 504, 295–300. [Google Scholar] [CrossRef]
  228. Zhang, Y.; Zhang, X.; Qu, S. Ceftriaxone Protects Astrocytes from MPP+ via Suppression of NF-ΚB/JNK/c-Jun Signaling. Mol. Neurobiol. 2015, 52, 78–92. [Google Scholar] [CrossRef] [PubMed]
  229. Ren, C.; He, K.-J.; Hu, H.; Zhang, J.-B.; Dong, L.-G.; Li, D.; Chen, J.; Mao, C.-J.; Wang, F.; Liu, C.-F. Induction of Parkinsonian-Like Changes via Targeted Downregulation of Astrocytic Glutamate Transporter GLT-1 in the Striatum. J. Park. Dis. 2022, 12, 295–314. [Google Scholar] [CrossRef]
  230. Ruzza, P.; Siligardi, G.; Hussain, R.; Marchiani, A.; Islami, M.; Bubacco, L.; Delogu, G.; Fabbri, D.; Dettori, M.A.; Sechi, M.; et al. Ceftriaxone Blocks the Polymerization of α-Synuclein and Exerts Neuroprotective Effects in Vitro. ACS Chem. Neurosci. 2014, 5, 30–38. [Google Scholar] [CrossRef] [Green Version]
  231. Smaga, I.; Fierro, D.; Mesa, J.; Filip, M.; Knackstedt, L.A. Molecular Changes Evoked by the Beta-Lactam Antibiotic Ceftriaxone across Rodent Models of Substance Use Disorder and Neurological Disease. Neurosci. Biobehav. Rev. 2020, 115, 116–130. [Google Scholar] [CrossRef]
  232. Zhou, X.; Lu, J.; Wei, K.; Wei, J.; Tian, P.; Yue, M.; Wang, Y.; Hong, D.; Li, F.; Wang, B.; et al. Neuroprotective Effect of Ceftriaxone on MPTP-Induced Parkinson’s Disease Mouse Model by Regulating Inflammation and Intestinal Microbiota. Oxid Med. Cell. Longev. 2021, 2021, 9424582. [Google Scholar] [CrossRef]
  233. Kaur, B.; Prakash, A. Ceftriaxone Attenuates Glutamate-Mediated Neuro-Inflammation and Restores BDNF in MPTP Model of Parkinson’s Disease in Rats. Pathophysiology 2017, 24, 71–79. [Google Scholar] [CrossRef]
  234. Bisht, R.; Kaur, B.; Gupta, H.; Prakash, A. Ceftriaxone Mediated Rescue of Nigral Oxidative Damage and Motor Deficits in MPTP Model of Parkinson’s Disease in Rats. Neurotoxicology 2014, 44, 71–79. [Google Scholar] [CrossRef]
  235. Brahmachari, S. Induction of Glial Fibrillary Acidic Protein Expression in Astrocytes by Nitric Oxide. J. Neurosci. 2006, 26, 4930–4939. [Google Scholar] [CrossRef] [Green Version]
  236. Jurga, A.M.; Paleczna, M.; Kuter, K.Z. Overview of General and Discriminating Markers of Differential Microglia Phenotypes. Front. Cell. Neurosci. 2020, 14, 198. [Google Scholar] [CrossRef]
  237. Ho, S.-C.; Hsu, C.-C.; Pawlak, C.R.; Tikhonova, M.A.; Lai, T.-J.; Amstislavskaya, T.G.; Ho, Y.-J. Effects of Ceftriaxone on the Behavioral and Neuronal Changes in an MPTP-Induced Parkinson’s Disease Rat Model. Behavioural. Brain Res. 2014, 268, 177–184. [Google Scholar] [CrossRef]
  238. Hsu, C.-Y.; Hung, C.-S.; Chang, H.-M.; Liao, W.-C.; Ho, S.-C.; Ho, Y.-J. Ceftriaxone Prevents and Reverses Behavioral and Neuronal Deficits in an MPTP-Induced Animal Model of Parkinson’s Disease Dementia. Neuropharmacology 2015, 91, 43–56. [Google Scholar] [CrossRef]
  239. Hsieh, M.-H.; Meng, W.-Y.; Liao, W.-C.; Weng, J.-C.; Li, H.-H.; Su, H.-L.; Lin, C.-L.; Hung, C.-S.; Ho, Y.-J. Ceftriaxone Reverses Deficits of Behavior and Neurogenesis in an MPTP-Induced Rat Model of Parkinson’s Disease Dementia. Brain Res. Bull 2017, 132, 129–138. [Google Scholar] [CrossRef]
  240. Chotibut, T.; Davis, R.W.; Arnold, J.C.; Frenchek, Z.; Gurwara, S.; Bondada, V.; Geddes, J.W.; Salvatore, M.F. Ceftriaxone Increases Glutamate Uptake and Reduces Striatal Tyrosine Hydroxylase Loss in 6-OHDA Parkinson’s Model. Mol. Neurobiol. 2014, 49, 1282–1292. [Google Scholar] [CrossRef] [Green Version]
  241. Weng, J.-C.; Tikhonova, M.A.; Chen, J.-H.; Shen, M.-S.; Meng, W.-Y.; Chang, Y.-T.; Chen, K.-H.; Liang, K.-C.; Hung, C.-S.; Amstislavskaya, T.G.; et al. Ceftriaxone Prevents the Neurodegeneration and Decreased Neurogenesis Seen in a Parkinson’s Disease Rat Model: An Immunohistochemical and MRI Study. Behav. Brain Res. 2016, 305, 126–139. [Google Scholar] [CrossRef]
  242. Leung, T.C.H.; Lui, C.N.P.; Chen, L.W.; Yung, W.H.; Chan, Y.S.; Yung, K.K.L. Ceftriaxone Ameliorates Motor Deficits and Protects Dopaminergic Neurons in 6-Hydroxydopamine-Lesioned Rats. ACS Chem. Neurosci. 2012, 3, 22–30. [Google Scholar] [CrossRef]
  243. Kelsey, J.E.; Neville, C. The Effects of the β-Lactam Antibiotic, Ceftriaxone, on Forepaw Stepping and l-DOPA-Induced Dyskinesia in a Rodent Model of Parkinson’s Disease. Psychopharmacology 2014, 231, 2405–2415. [Google Scholar] [CrossRef]
  244. Chotibut, T.; Meadows, S.; Kasanga, E.A.; McInnis, T.; Cantu, M.A.; Bishop, C.; Salvatore, M.F. Ceftriaxone Reduces L-Dopa-Induced Dyskinesia Severity in 6-Hydroxydopamine Parkinson’s Disease Model. Mov. Disord. 2017, 32, 1547–1556. [Google Scholar] [CrossRef]
  245. Karaman, I.; Kizilay-Ozfidan, G.; Karadag, C.H.; Ulugol, A. Lack of Effect of Ceftriaxone, a GLT-1 Transporter Activator, on Spatial Memory in Mice. Pharmacol. Biochem. Behav. 2013, 108, 61–65. [Google Scholar] [CrossRef]
  246. Huang, C.-K.; Chang, Y.-T.; Amstislavskaya, T.G.; Tikhonova, M.A.; Lin, C.-L.; Hung, C.-S.; Lai, T.-J.; Ho, Y.-J. Synergistic Effects of Ceftriaxone and Erythropoietin on Neuronal and Behavioral Deficits in an MPTP-Induced Animal Model of Parkinson’s Disease Dementia. Behav. Brain Res. 2015, 294, 198–207. [Google Scholar] [CrossRef]
  247. Garrido-Mesa, N.; Zarzuelo, A.; Gálvez, J. Minocycline: Far beyond an Antibiotic. Br. J. Pharmacol. 2013, 169, 337–352. [Google Scholar] [CrossRef] [Green Version]
  248. Cankaya, S.; Cankaya, B.; Kilic, U.; Kilic, E.; Yulug, B. The Therapeutic Role of Minocycline in Parkinson’s Disease. Drugs Context 2019, 8, 212553. [Google Scholar] [CrossRef] [Green Version]
  249. Peng, J.; Xie, L.; Stevenson, F.F.; Melov, S.; di Monte, D.A.; Andersen, J.K. Nigrostriatal Dopaminergic Neurodegeneration in the Weaver Mouse Is Mediated via Neuroinflammation and Alleviated by Minocycline Administration. J. Neurosci. 2006, 26, 11644–11651. [Google Scholar] [CrossRef] [Green Version]
  250. Inamdar, A.A.; Chaudhuri, A.; O’Donnell, J. The Protective Effect of Minocycline in a Paraquat-Induced Parkinson’s Disease Model in Drosophila Is Modified in Altered Genetic Backgrounds. Park. Dis. 2012, 2012, 212553. [Google Scholar] [CrossRef] [Green Version]
  251. Kim, H.-S.; Suh, Y.-H. Minocycline and Neurodegenerative Diseases. Behav. Brain Res. 2009, 196, 168–179. [Google Scholar] [CrossRef] [PubMed]
  252. Thomas, M.; Le, W.D.; Jankovic, J. Minocycline and Other Tetracycline Derivatives: A Neuroprotective Strategy in Parkinson’s Disease and Huntington’s Disease. Clin. Neuropharmacol 2003, 26, 18–23. [Google Scholar] [CrossRef] [PubMed]
  253. Yong, V.W.; Wells, J.; Giuliani, F.; Casha, S.; Power, C.; Metz, L.M. The Promise of Minocycline in Neurology. Lancet Neurol. 2004, 3, 744–751. [Google Scholar] [CrossRef]
  254. Fan, L.; Wang, T.-L.; Xu, Y.C.; Ma, Y.H.; Ye, W.G. Minocycline May Be Useful to Prevent/Treat Postoperative Cognitive Decline in Elderly Patients. Med. Hypotheses 2011, 76, 733–736. [Google Scholar] [CrossRef]
  255. Blum, D.; Chtarto, A.; Tenenbaum, L.; Brotchi, J.; Levivier, M. Clinical Potential of Minocycline for Neurodegenerative Disorders. Neurobiol. Dis. 2004, 17, 359–366. [Google Scholar] [CrossRef]
  256. Thomas, M.; Le, W. Minocycline: Neuroprotective Mechanisms in Parkinsons Disease. Curr. Pharm. Des. 2004, 10, 679–686. [Google Scholar] [CrossRef]
  257. Zemke, D.; Majid, A. The Potential of Minocycline for Neuroprotection in Human Neurologic Disease. Clin. Neuropharmacol 2004, 27, 293–298. [Google Scholar] [CrossRef]
  258. Ruan, Z.; Zhang, D.; Huang, R.; Sun, W.; Hou, L.; Zhao, J.; Wang, Q. Microglial Activation Damages Dopaminergic Neurons through MMP-2/-9-Mediated Increase of Blood-Brain Barrier Permeability in a Parkinson’s Disease Mouse Model. Int. J. Mol. Sci. 2022, 23, 2793. [Google Scholar] [CrossRef]
  259. Wang, Y.; Wang, Q.; Yu, R.; Zhang, Q.; Zhang, Z.; Li, H.; Ren, C.; Yang, R.; Niu, H. Minocycline Inhibition of Microglial Rescues Nigrostriatal Dopaminergic Neurodegeneration Caused by Mutant Alpha-Synuclein Overexpression. Aging 2020, 12, 14232–14243. [Google Scholar] [CrossRef]
  260. Zhang, D.; Li, S.; Hou, L.; Jing, L.; Ruan, Z.; Peng, B.; Zhang, X.; Hong, J.-S.; Zhao, J.; Wang, Q. Microglial Activation Contributes to Cognitive Impairments in Rotenone-Induced Mouse Parkinson’s Disease Model. J. Neuroinflamm. 2021, 18, 4. [Google Scholar] [CrossRef]
  261. Tomás-Camardiel, M.; Rite, I.; Herrera, A.J.; de Pablos, R.M.; Cano, J.; Machado, A.; Venero, J.L. Minocycline Reduces the Lipopolysaccharide-Induced Inflammatory Reaction, Peroxynitrite-Mediated Nitration of Proteins, Disruption of the Blood–Brain Barrier, and Damage in the Nigral Dopaminergic System. Neurobiol. Dis. 2004, 16, 190–201. [Google Scholar] [CrossRef]
  262. Wu, D.C.; Jackson-Lewis, V.; Vila, M.; Tieu, K.; Teismann, P.; Vadseth, C.; Choi, D.-K.; Ischiropoulos, H.; Przedborski, S. Blockade of Microglial Activation Is Neuroprotective in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Mouse Model of Parkinson Disease. J. Neurosci. 2002, 22, 1763–1771. [Google Scholar] [CrossRef]
  263. Kumar, V.; Singh, B.K.; Chauhan, A.K.; Singh, D.; Patel, D.K.; Singh, C. Minocycline Rescues from Zinc-Induced Nigrostriatal Dopaminergic Neurodegeneration: Biochemical and Molecular Interventions. Mol. Neurobiol. 2016, 53, 2761–2777. [Google Scholar] [CrossRef] [PubMed]
  264. Du, Y.; Ma, Z.; Lin, S.; Dodel, R.C.; Gao, F.; Bales, K.R.; Triarhou, L.C.; Chernet, E.; Perry, K.W.; Nelson, D.L.G.; et al. Minocycline Prevents Nigrostriatal Dopaminergic Neurodegeneration in the MPTP Model of Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2001, 98, 14669–14674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Jiang, B.-P.; Le, L.; Xu, L.-J.; Xiao, P.-G. Minocycline Inhibits ICAD Degradation and the NF-ΚB Activation Induced by 6-OHDA in PC12 Cells. Brain Res. 2014, 1586, 1–11. [Google Scholar] [CrossRef]
  266. Radad, K.; Moldzio, R.; Rausch, W.-D. Minocycline Protects Dopaminergic Neurons Against Long-Term Rotenone Toxicity. Can. J. Neurol. Sci. J. Can. Des Sci. Neurol. 2010, 37, 81–85. [Google Scholar] [CrossRef] [Green Version]
  267. Lin, S.; Wei, X.; Xu, Y.; Yan, C.; Dodel, R.; Zhang, Y.; Liu, J.; Klaunig, J.E.; Farlow, M.; Du, Y. Minocycline Blocks 6-Hydroxydopamine-Induced Neurotoxicity and Free Radical Production in Rat Cerebellar Granule Neurons. Life Sci. 2003, 72, 1635–1641. [Google Scholar] [CrossRef]
  268. Cronin, A.; Grealy, M. Neuroprotective and Neuro-Restorative Effects of Minocycline and Rasagiline in a Zebrafish 6-Hydroxydopamine Model of Parkinson’s Disease. Neuroscience 2017, 367, 34–46. [Google Scholar] [CrossRef]
  269. Maneshian, M.; Nasirinezhad, F.; Mohammadi, F.; Behzadi, M.; Asadi-Shekaari, M.; Shabani, M. Minocycline Mitigation of Tremor Syndrome and Defect of Cognitive and Balance Induced by Harmaline. Basic Clin. Neurosci. J. 2021, 12, 255–268. [Google Scholar] [CrossRef]
  270. Quintero, E.M.; Willis, L.; Singleton, R.; Harris, N.; Huang, P.; Bhat, N.; Granholm, A.-C. Behavioral and Morphological Effects of Minocycline in the 6-Hydroxydopamine Rat Model of Parkinson’s Disease. Brain Res. 2006, 1093, 198–207. [Google Scholar] [CrossRef]
  271. Sun, C.; Wang, Y.; Mo, M.; Song, C.; Wang, X.; Chen, S.; Liu, Y. Minocycline Protects against Rotenone-Induced Neurotoxicity Correlating with Upregulation of Nurr1 in a Parkinson’s Disease Rat Model. Biomed Res. Int. 2019, 2019, 6843265. [Google Scholar] [CrossRef] [Green Version]
  272. Huang, C.-L.; Lee, Y.-C.; Yang, Y.-C.; Kuo, T.-Y.; Huang, N.-K. Minocycline Prevents Paraquat-Induced Cell Death through Attenuating Endoplasmic Reticulum Stress and Mitochondrial Dysfunction. Toxicol. Lett. 2012, 209, 203–210. [Google Scholar] [CrossRef] [PubMed]
  273. Fernandez-Gomez, F.J.; Galindo, M.F.; Gomez-Lazaro, M.; González-García, C.; Ceña, V.; Aguirre, N.; Jordán, J. Involvement of Mitochondrial Potential and Calcium Buffering Capacity in Minocycline Cytoprotective Actions. Neuroscience 2005, 133, 959–967. [Google Scholar] [CrossRef]
  274. Dixit, A.; Srivastava, G.; Verma, D.; Mishra, M.; Singh, P.K.; Prakash, O.; Singh, M.P. Minocycline, Levodopa and MnTMPyP Induced Changes in the Mitochondrial Proteome Profile of MPTP and Maneb and Paraquat Mice Models of Parkinson’s Disease. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2013, 1832, 1227–1240. [Google Scholar] [CrossRef] [Green Version]
  275. Yang, L.; Sugama, S.; Chirichigno, J.W.; Gregorio, J.; Lorenzl, S.; Shin, D.H.; Browne, S.E.; Shimizu, Y.; Joh, T.H.; Beal, M.F.; et al. Minocycline Enhances MPTP Toxicity to Dopaminergic Neurons. J. Neurosci. Res. 2003, 74, 278–285. [Google Scholar] [CrossRef]
  276. Diguet, E.; Fernagut, P.-O.; Wei, X.; Du, Y.; Rouland, R.; Gross, C.; Bezard, E.; Tison, F. Deleterious Effects of Minocycline in Animal Models of Parkinson’s Disease and Huntington’s Disease. Eur. J. Neurosci. 2004, 19, 3266–3276. [Google Scholar] [CrossRef]
  277. NINDS NET-PD Investigators. A Pilot Clinical Trial of Creatine and Minocycline in Early Parkinson Disease. Clin. Neuropharmacol 2008, 31, 141–150. [Google Scholar] [CrossRef] [Green Version]
  278. NINDS NET-PD Investigators. A Randomized, Double-Blind, Futility Clinical Trial of Creatine and Minocycline in Early Parkinson Disease. Neurology 2006, 66, 664–671. [Google Scholar] [CrossRef]
  279. Rahmani, M.; Negro Álvarez, S.E.; Hernández, E.B. The Potential Use of Tetracyclines in Neurodegenerative Diseases and the Role of Nano-Based Drug Delivery Systems. Eur. J. Pharm. Sci. 2022, 175, 106237. [Google Scholar] [CrossRef]
  280. Santa-Cecília, F.V.; Socias, B.; Ouidja, M.O.; Sepulveda-Diaz, J.E.; Acuña, L.; Silva, R.L.; Michel, P.P.; Del-Bel, E.; Cunha, T.M.; Raisman-Vozari, R. Doxycycline Suppresses Microglial Activation by Inhibiting the P38 MAPK and NF-KB Signaling Pathways. Neurotox. Res. 2016, 29, 447–459. [Google Scholar] [CrossRef]
  281. Cho, Y.; Son, H.J.; Kim, E.-M.; Choi, J.H.; Kim, S.T.; Ji, I.J.; Choi, D.H.; Joh, T.H.; Kim, Y.S.; Hwang, O. Doxycycline Is Neuroprotective Against Nigral Dopaminergic Degeneration by a Dual Mechanism Involving MMP-3. Neurotox. Res. 2009, 16, 361–371. [Google Scholar] [CrossRef] [PubMed]
  282. Dominguez-Meijide, A.; Parrales, V.; Vasili, E.; González-Lizárraga, F.; König, A.; Lázaro, D.F.; Lannuzel, A.; Haik, S.; del Bel, E.; Chehín, R.; et al. Doxycycline Inhibits α-Synuclein-Associated Pathologies in Vitro and in Vivo. Neurobiol. Dis. 2021, 151, 105256. [Google Scholar] [CrossRef]
  283. Do Amaral, L.; dos Santos, N.A.G.; Sisti, F.M.; del Bel, E.; Santos, A.C. dos The Antibiotic Doxycycline Mimics the NGF Signaling in PC12 Cells: A Relevant Mechanism for Neuroprotection. Chem. Biol. Interact 2021, 341, 109454. [Google Scholar] [CrossRef] [PubMed]
  284. González-Lizárraga, F.; Socías, S.B.; Ávila, C.L.; Torres-Bugeau, C.M.; Barbosa, L.R.S.; Binolfi, A.; Sepúlveda-Díaz, J.E.; Del-Bel, E.; Fernandez, C.O.; Papy-Garcia, D.; et al. Repurposing Doxycycline for Synucleinopathies: Remodelling of α-Synuclein Oligomers towards Non-Toxic Parallel Beta-Sheet Structured Species. Sci. Rep. 2017, 7, 41755. [Google Scholar] [CrossRef] [Green Version]
  285. Lazzarini, M.; Martin, S.; Mitkovski, M.; Vozari, R.R.; Stühmer, W.; Bel, E. del Doxycycline Restrains Glia and Confers Neuroprotection in a 6-OHDA Parkinson Model. Glia 2013, 61, 1084–1100. [Google Scholar] [CrossRef]
  286. Zhang, G.-B.; Feng, Y.-H.; Wang, P.-Q.; Song, J.-H.; Wang, P.; Wang, S.-A. A Study on the Protective Role of Doxycycline upon Dopaminergic Neuron of LPS-PD Rat Model Rat. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 3468–3474. [Google Scholar]
  287. Bortolanza, M.; Nascimento, G.C.; Raisman-Vozari, R.; Del-Bel, E. Doxycycline and Its Derivative, COL-3, Decrease Dyskinesia Induced by L-DOPA in Hemiparkinsonian Rats. Br. J. Pharmacol. 2021, 178, 2595–2616. [Google Scholar] [CrossRef]
  288. Bi, W.; Zhu, L.; Jing, X.; Liang, Y.; Tao, E. Rifampicin and Parkinson’s Disease. Neurol. Sci. 2013, 34, 137–141. [Google Scholar] [CrossRef]
  289. Yulug, B.; Hanoglu, L.; Kilic, E.; Schabitz, W.R. Rifampicin: An Antibiotic with Brain Protective Function. Brain Res. Bull 2014, 107, 37–42. [Google Scholar] [CrossRef] [PubMed]
  290. Lin, D.; Jing, X.; Chen, Y.; Liang, Y.; Lei, M.; Peng, S.; Zhou, T.; Zheng, D.; Zeng, Z.; Wu, X.; et al. Rifampicin Pre-Treatment Inhibits the Toxicity of Rotenone-Induced PC12 Cells by Enhancing Sumoylation Modification of α-Synuclein. Biochem. Biophys. Res. Commun. 2017, 485, 23–29. [Google Scholar] [CrossRef] [PubMed]
  291. Li, J.; Zhu, M.; Rajamani, S.; Uversky, V.N.; Fink, A.L. Rifampicin Inhibits α-Synuclein Fibrillation and Disaggregates Fibrils. Chem. Biol. 2004, 11, 1513–1521. [Google Scholar] [CrossRef] [Green Version]
  292. Xu, J.; Wei, C.; Xu, C.; Bennett, M.C.; Zhang, G.; Li, F.; Tao, E. Rifampicin Protects PC12 Cells against MPP+-Induced Apoptosis and Inhibits the Expression of an α-Synuclein Multimer. Brain Res. 2007, 1139, 220–225. [Google Scholar] [CrossRef] [PubMed]
  293. Liang, Y.; Zhou, T.; Chen, Y.; Lin, D.; Jing, X.; Peng, S.; Zheng, D.; Zeng, Z.; Lei, M.; Wu, X.; et al. Rifampicin Inhibits Rotenone-Induced Microglial Inflammation via Enhancement of Autophagy. Neurotoxicology 2017, 63, 137–145. [Google Scholar] [CrossRef]
  294. Acuña, L.; Hamadat, S.; Corbalán, N.S.; González-Lizárraga, F.; Dos-Santos-Pereira, M.; Rocca, J.; Díaz, J.S.; Del-Bel, E.; Papy-García, D.; Chehín, R.N.; et al. Rifampicin and Its Derivative Rifampicin Quinone Reduce Microglial Inflammatory Responses and Neurodegeneration Induced In Vitro by α-Synuclein Fibrillary Aggregates. Cells 2019, 8, 776. [Google Scholar] [CrossRef] [Green Version]
  295. Liang, Y.; Zheng, D.; Peng, S.; Lin, D.; Jing, X.; Zeng, Z.; Chen, Y.; Huang, K.; Xie, Y.; Zhou, T.; et al. Rifampicin Attenuates Rotenone-Treated Microglia Inflammation via Improving Lysosomal Function. Toxicol. Vitr. 2020, 63, 104690. [Google Scholar] [CrossRef]
  296. Wu, X.; Liang, Y.; Jing, X.; Lin, D.; Chen, Y.; Zhou, T.; Peng, S.; Zheng, D.; Zeng, Z.; Lei, M.; et al. Rifampicin Prevents SH-SY5Y Cells from Rotenone-Induced Apoptosis via the PI3K/Akt/GSK-3β/CREB Signaling Pathway. Neurochem. Res. 2018, 43, 886–893. [Google Scholar] [CrossRef]
  297. Jing, X.; Shi, Q.; Bi, W.; Zeng, Z.; Liang, Y.; Wu, X.; Xiao, S.; Liu, J.; Yang, L.; Tao, E. Rifampicin Protects PC12 Cells from Rotenone-Induced Cytotoxicity by Activating GRP78 via PERK-EIF2α-ATF4 Pathway. PLoS ONE 2014, 9, e92110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  298. Oida, Y.; Kitaichi, K.; Nakayama, H.; Ito, Y.; Fujimoto, Y.; Shimazawa, M.; Nagai, H.; Hara, H. Rifampicin Attenuates the MPTP-Induced Neurotoxicity in Mouse Brain. Brain Res. 2006, 1082, 196–204. [Google Scholar] [CrossRef]
  299. Yurtsever, İ.; Üstündağ, Ü.V.; Ünal, İ.; Ateş, P.S.; Emekli-Alturfan, E. Rifampicin Decreases Neuroinflammation to Maintain Mitochondrial Function and Calcium Homeostasis in Rotenone-Treated Zebrafish. Drug Chem. Toxicol. 2022, 45, 1544–1551. [Google Scholar] [CrossRef]
  300. Modi, S.R.; Collins, J.J.; Relman, D.A. Antibiotics and the Gut Microbiota. J. Clin. Investig. 2014, 124, 4212–4218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  301. Markulin, I.; Matasin, M.; Turk, V.E.; Salković-Petrisic, M. Challenges of Repurposing Tetracyclines for the Treatment of Alzheimer’s and Parkinson’s Disease. J. Neural. Transm. 2022, 129, 773–804. [Google Scholar] [CrossRef] [PubMed]
  302. Avagliano, C.; Coretti, L.; Lama, A.; Pirozzi, C.; de Caro, C.; de Biase, D.; Turco, L.; Mollica, M.P.; Paciello, O.; Calignano, A.; et al. Dual-Hit Model of Parkinson’s Disease: Impact of Dysbiosis on 6-Hydroxydopamine-Insulted Mice—Neuroprotective and Anti-Inflammatory Effects of Butyrate. Int. J. Mol. Sci. 2022, 23, 6367. [Google Scholar] [CrossRef] [PubMed]
  303. Ternák, G.; Kuti, D.; Kovács, K.J. Dysbiosis in Parkinson’s Disease Might Be Triggered by Certain Antibiotics. Med. Hypotheses 2020, 137, 109564. [Google Scholar] [CrossRef] [PubMed]
  304. Wang, C.; Lau, C.Y.; Ma, F.; Zheng, C. Genome-Wide Screen Identifies Curli Amyloid Fibril as a Bacterial Component Promoting Host Neurodegeneration. Proc. Natl. Acad. Sci. USA 2021, 118, e2106504118. [Google Scholar] [CrossRef]
  305. Roe, K. An Alternative Explanation for Alzheimer’s Disease and Parkinson’s Disease Initiation from Specific Antibiotics, Gut Microbiota Dysbiosis and Neurotoxins. Neurochem. Res. 2022, 47, 517–530. [Google Scholar] [CrossRef]
  306. Mertsalmi, T.H.; Pekkonen, E.; Scheperjans, F. Antibiotic Exposure and Risk of Parkinson’s Disease in Finland: A Nationwide Case-Control Study. Mov. Disord. 2020, 35, 431–442. [Google Scholar] [CrossRef]
  307. Palacios, N.; O’Reilly, É.J.; Schwarzschild, M.A.; Ascherio, A. Long-Term Use of Antibiotics and Risk of Parkinson’s Disease in the Nurses’ Health Study. Park. Dis. 2020, 2020, 4038375. [Google Scholar] [CrossRef] [Green Version]
  308. Morelli, L.; Capurso, L. FAO/WHO Guidelines on Probiotics. J. Clin. Gastroenterol. 2012, 46, S1–S2. [Google Scholar] [CrossRef]
  309. Alfonsetti, M.; Castelli, V.; d’Angelo, M. Are We What We Eat? Impact of Diet on the Gut–Brain Axis in Parkinson’s Disease. Nutrients 2022, 14, 380. [Google Scholar] [CrossRef]
  310. Cristofori, F.; Dargenio, V.N.; Dargenio, C.; Miniello, V.L.; Barone, M.; Francavilla, R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front. Immunol. 2021, 12, 578386. [Google Scholar] [CrossRef]
  311. Gazerani, P. Probiotics for Parkinson’s Disease. Int. J. Mol. Sci. 2019, 20, 4121. [Google Scholar] [CrossRef] [Green Version]
  312. Gupta, V.; Garg, R. PROBIOTICS. Indian J. Med. Microbiol. 2009, 27, 202–209. [Google Scholar] [CrossRef]
  313. Reid, G.; Younes, J.A.; van der Mei, H.C.; Gloor, G.B.; Knight, R.; Busscher, H.J. Microbiota Restoration: Natural and Supplemented Recovery of Human Microbial Communities. Nat. Rev. Microbiol. 2011, 9, 27–38. [Google Scholar] [CrossRef] [PubMed]
  314. Nandwana, V.; Nandwana, N.K.; Das, Y.; Saito, M.; Panda, T.; Das, S.; Almaguel, F.; Hosmane, N.S.; Das, B.C. The Role of Microbiome in Brain Development and Neurodegenerative Diseases. Molecules 2022, 27, 3402. [Google Scholar] [CrossRef] [PubMed]
  315. Peterson, C.T. Dysfunction of the Microbiota-Gut-Brain Axis in Neurodegenerative Disease: The Promise of Therapeutic Modulation With Prebiotics, Medicinal Herbs, Probiotics, and Synbiotics. J. Evid Based Integr. Med. 2020, 25, 2515690X20957225. [Google Scholar] [CrossRef]
  316. Becker, A.; Pierre Schmartz, G.; Gröger, L.; Grammes, N.; Galata, V.; Philippeit, H.; Weiland, J.; Ludwig, N.; Meese, E.; Tierling, S.; et al. Effects of Resistant Starch on Symptoms, Fecal Markers and Gut Microbiota in Parkinson’s Disease–the RESISTA-PD Trial. Genom. Proteom. Bioinform. 2021. [Google Scholar] [CrossRef]
  317. Jiang, J.; Chu, C.; Wu, C.; Wang, C.; Zhang, C.; Li, T.; Zhai, Q.; Yu, L.; Tian, F.; Chen, W. Efficacy of Probiotics in Multiple Sclerosis: A Systematic Review of Preclinical Trials and Meta-Analysis of Randomized Controlled Trials. Food Funct. 2021, 12, 2354–2377. [Google Scholar] [CrossRef]
  318. Mirzaei, H.; Sedighi, S.; Kouchaki, E.; Barati, E.; Dadgostar, E.; Aschner, M.; Tamtaji, O.R. Probiotics and the Treatment of Parkinson’s Disease: An Update. Cell. Mol. Neurobiol. 2021. [Google Scholar] [CrossRef]
  319. Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The Role of Microbiota-Gut-Brain Axis in Neuropsychiatric and Neurological Disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
  320. Xiang, S.; Ji, J.-L.; Li, S.; Cao, X.-P.; Xu, W.; Tan, L.; Tan, C.-C. Efficacy and Safety of Probiotics for the Treatment of Alzheimer’s Disease, Mild Cognitive Impairment, and Parkinson’s Disease: A Systematic Review and Meta-Analysis. Front. Aging Neurosci. 2022, 14. [Google Scholar] [CrossRef]
  321. Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota Modulate Behavioral and Physiological Abnormalities Associated with Neurodevelopmental Disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  322. Bonfili, L.; Cecarini, V.; Berardi, S.; Scarpona, S.; Suchodolski, J.S.; Nasuti, C.; Fiorini, D.; Boarelli, M.C.; Rossi, G.; Eleuteri, A.M. Microbiota Modulation Counteracts Alzheimer’s Disease Progression Influencing Neuronal Proteolysis and Gut Hormones Plasma Levels. Sci. Rep. 2017, 7, 2426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  323. Akbari, E.; Asemi, Z.; Daneshvar Kakhaki, R.; Bahmani, F.; Kouchaki, E.; Tamtaji, O.R.; Hamidi, G.A.; Salami, M. Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer’s Disease: A Randomized, Double-Blind and Controlled Trial. Front. Aging Neurosci. 2016, 8, 256. [Google Scholar] [CrossRef]
  324. Abildgaard, A.; Elfving, B.; Hokland, M.; Wegener, G.; Lund, S. Probiotic Treatment Reduces Depressive-like Behaviour in Rats Independently of Diet. Psychoneuroendocrinology 2017, 79, 40–48. [Google Scholar] [CrossRef]
  325. Srivastav, S.; Neupane, S.; Bhurtel, S.; Katila, N.; Maharjan, S.; Choi, H.; Hong, J.T.; Choi, D.-Y. Probiotics Mixture Increases Butyrate, and Subsequently Rescues the Nigral Dopaminergic Neurons from MPTP and Rotenone-Induced Neurotoxicity. J. Nutr. Biochem. 2019, 69, 73–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  326. Wallace, C.J.K.; Milev, R. The Effects of Probiotics on Depressive Symptoms in Humans: A Systematic Review. Ann. Gen. Psychiatry 2017, 16. [Google Scholar] [CrossRef] [Green Version]
  327. Wang, H.; Lee, I.-S.; Braun, C.; Enck, P. Effect of Probiotics on Central Nervous System Functions in Animals and Humans: A Systematic Review. J. Neurogastroenterol. Motil. 2016, 22, 589–605. [Google Scholar] [CrossRef] [Green Version]
  328. Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello, R.; Pane, M.; Comi, C. Probiotics May Have Beneficial Effects in Parkinson’s Disease: In Vitro Evidence. Front. Immunol. 2019, 10, 969. [Google Scholar] [CrossRef] [Green Version]
  329. Ghyselinck, J.; Verstrepen, L.; Moens, F.; van den Abbeele, P.; Bruggeman, A.; Said, J.; Smith, B.; Barker, L.A.; Jordan, C.; Leta, V.; et al. Influence of Probiotic Bacteria on Gut Microbiota Composition and Gut Wall Function in an In-Vitro Model in Patients with Parkinson’s Disease. Int. J. Pharm. X 2021, 3, 100087. [Google Scholar] [CrossRef] [PubMed]
  330. Cheon, M.-J.; Lee, N.-K.; Paik, H.-D. Neuroprotective Effects of Heat-Killed Lactobacillus Plantarum 200655 Isolated from Kimchi Against Oxidative Stress. Probiotics Antimicrob Proteins 2021, 13, 788–795. [Google Scholar] [CrossRef]
  331. Castelli, V.; d’Angelo, M.; Lombardi, F.; Alfonsetti, M.; Antonosante, A.; Catanesi, M.; Benedetti, E.; Palumbo, P.; Cifone, M.G.; Giordano, A.; et al. Effects of the Probiotic Formulation SLAB51 in in Vitro and in Vivo Parkinson’s Disease Models. Aging 2020, 12, 4641–4659. [Google Scholar] [CrossRef] [PubMed]
  332. Liao, J.-F.; Cheng, Y.-F.; You, S.-T.; Kuo, W.-C.; Huang, C.-W.; Chiou, J.-J.; Hsu, C.-C.; Hsieh-Li, H.-M.; Wang, S.; Tsai, Y.-C. Lactobacillus Plantarum PS128 Alleviates Neurodegenerative Progression in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Induced Mouse Models of Parkinson’s Disease. Brain Behav. Immun. 2020, 90, 26–46. [Google Scholar] [CrossRef] [PubMed]
  333. Hsieh, T.-H.; Kuo, C.-W.; Hsieh, K.-H.; Shieh, M.-J.; Peng, C.-W.; Chen, Y.-C.; Chang, Y.-L.; Huang, Y.-Z.; Chen, C.-C.; Chang, P.-K.; et al. Probiotics Alleviate the Progressive Deterioration of Motor Functions in a Mouse Model of Parkinson’s Disease. Brain Sci. 2020, 10, 206. [Google Scholar] [CrossRef]
  334. Sun, J.; Li, H.; Jin, Y.; Yu, J.; Mao, S.; Su, K.-P.; Ling, Z.; Liu, J. Probiotic Clostridium Butyricum Ameliorated Motor Deficits in a Mouse Model of Parkinson’s Disease via Gut Microbiota-GLP-1 Pathway. Brain Behav. Immun. 2021, 91, 703–715. [Google Scholar] [CrossRef] [PubMed]
  335. Wang, L.; Zhao, Z.; Zhao, L.; Zhao, Y.; Yang, G.; Wang, C.; Gao, L.; Niu, C.; Li, S. Lactobacillus Plantarum DP189 Reduces α-SYN Aggravation in MPTP-Induced Parkinson’s Disease Mice via Regulating Oxidative Damage, Inflammation, and Gut Microbiota Disorder. J. Agric. Food Chem. 2022, 70, 1163–1173. [Google Scholar] [CrossRef] [PubMed]
  336. Cassani, E.; Privitera, G.; Pezzoli, G.; Pusani, C.; Madio, C.; Iorio, L.; Barichella, M. Use of Probiotics for the Treatment of Constipation in Parkinson’s Disease Patients. Minerva Gastroenterol. Dietol. 2011, 57, 117–121. [Google Scholar] [PubMed]
  337. Georgescu, D.; Ancusa, O.; Georgescu, L.; Ionita, I.; Reisz, D. Nonmotor Gastrointestinal Disorders in Older Patients with Parkinson’s Disease: Is There Hope? Clin. Interv. Aging 2016, 11, 1601–1608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  338. Borzabadi, S.; Oryan, S.; Eidi, A.; Aghadavod, E.; Daneshvar Kakhaki, R.; Tamtaji, O.R.; Taghizadeh, M.; Asemi, Z. The Effects of Probiotic Supplementation on Gene Expression Related to Inflammation, Insulin and Lipid in Patients with Parkinson’s Disease: A Randomized, Double-Blind, PlaceboControlled Trial. Arch. Iran Med. 2018, 21, 289–295. [Google Scholar]
  339. Tamtaji, O.R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and Metabolic Response to Probiotic Administration in People with Parkinson’s Disease: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Nutr. 2019, 38, 1031–1035. [Google Scholar] [CrossRef]
  340. Ibrahim, A.; Ali, R.A.R.; Manaf, M.R.A.; Ahmad, N.; Tajurruddin, F.W.; Qin, W.Z.; Desa, S.H.M.; Ibrahim, N.M. Multi-Strain Probiotics (Hexbio) Containing MCP BCMC Strains Improved Constipation and Gut Motility in Parkinson’s Disease: A Randomised Controlled Trial. PLoS ONE 2020, 15, e0244680. [Google Scholar] [CrossRef]
  341. Tan, A.H.; Lim, S.-Y.; Chong, K.K.; Azhan A Manap, M.A.; Hor, J.W.; Lim, J.L.; Low, S.C.; Chong, C.W.; Mahadeva, S.; Lang, A.E. Probiotics for Constipation in Parkinson’s Disease: A Randomized Placebo-Controlled Study. Neurology 2020, 96, e772–e782. [Google Scholar] [CrossRef] [PubMed]
  342. Sun, H.; Zhao, F.; Liu, Y.; Ma, T.; Jin, H.; Quan, K.; Leng, B.; Zhao, J.; Yuan, X.; Li, Z.; et al. Probiotics Synergized with Conventional Regimen in Managing Parkinson’s Disease. NPJ Park. Dis. 2022, 8. [Google Scholar] [CrossRef]
  343. Surwase, S.N.; Jadhav, J.P. Bioconversion of L-Tyrosine to l-DOPA by a Novel Bacterium Bacillus sp. JPJ. Amino Acids 2011, 41, 495–506. [Google Scholar] [CrossRef]
  344. Goya, M.E.; Xue, F.; Sampedro-Torres-Quevedo, C.; Arnaouteli, S.; Riquelme-Dominguez, L.; Romanowski, A.; Brydon, J.; Ball, K.L.; Stanley-Wall, N.R.; Doitsidou, M. Probiotic Bacillus Subtilis Protects against α-Synuclein Aggregation in C. Elegans. Cell Rep. 2020, 30, 367–380.e7. [Google Scholar] [CrossRef] [PubMed]
  345. Perez Visñuk, D.; Savoy de Giori, G.; LeBlanc, J.G.; de Moreno de LeBlanc, A. Neuroprotective Effects Associated with Immune Modulation by Selected Lactic Acid Bacteria in a Parkinson’s Disease Model. Nutrition 2020, 79–80, 110995. [Google Scholar] [CrossRef] [PubMed]
  346. Tsao, S.-P.; Nurrahma, B.A.; Kumar, R.; Wu, C.-H.; Yeh, T.-H.; Chiu, C.-C.; Lee, Y.-P.; Liao, Y.-C.; Huang, C.-H.; Yeh, Y.-T.; et al. Probiotic Enhancement of Antioxidant Capacity and Alterations of Gut Microbiota Composition in 6-Hydroxydopamin-Induced Parkinson’s Disease Rats. Antioxidants 2021, 10, 1823. [Google Scholar] [CrossRef] [PubMed]
  347. Alipour Nosrani, E.; Tamtaji, O.R.; Alibolandi, Z.; Sarkar, P.; Ghazanfari, M.; Azami Tameh, A.; Taghizadeh, M.; Banikazemi, Z.; Hadavi, R.; Naderi Taheri, M. Neuroprotective Effects of Probiotics Bacteria on Animal Model of Parkinson’s Disease Induced by 6-Hydroxydopamine: A Behavioral, Biochemical, and Histological Study. J. Immunoass. Immunochem. 2021, 42, 106–120. [Google Scholar] [CrossRef] [PubMed]
  348. Xie, C.; Prasad, A.A. Probiotics Treatment Improves Hippocampal Dependent Cognition in a Rodent Model of Parkinson’s Disease. Microorganisms 2020, 8, 1661. [Google Scholar] [CrossRef]
  349. Fang, X.; Tian, P.; Zhao, X.; Jiang, C.; Chen, T. Neuroprotective Effects of an Engineered Commensal Bacterium in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine Parkinson Disease Mouse Model via Producing Glucagon-like Peptide-1. J. Neurochem. 2019, 150, 441–452. [Google Scholar] [CrossRef]
  350. Fang, X.; Zhou, X.; Miao, Y.; Han, Y.; Wei, J.; Chen, T. Therapeutic Effect of GLP-1 Engineered Strain on Mice Model of Alzheimer’s Disease and Parkinson’s Disease. AMB Express 2020, 10, 13–80. [Google Scholar] [CrossRef]
  351. Tan, A.H.; Hor, J.W.; Chong, C.W.; Lim, S. Probiotics for Parkinson’s Disease: Current Evidence and Future Directions. JGH Open 2021, 5, 414–419. [Google Scholar] [CrossRef] [PubMed]
  352. Xu, Y.; Xie, M.; Xue, J.; Xiang, L.; Li, Y.; Xiao, J.; Xiao, G.; Wang, H. EGCG Ameliorates Neuronal and Behavioral Defects by Remodeling Gut Microbiota and TotM Expression in Drosophila Models of Parkinson’s Disease. FASEB J. 2020, 34, 5931–5950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. Dwyer, Z.; Chaiquin, M.; Landrigan, J.; Ayoub, K.; Shail, P.; Rocha, J.; Childers, C.L.; Storey, K.B.; Philpott, D.J.; Sun, H.; et al. The Impact of Dextran Sodium Sulphate and Probiotic Pre-Treatment in a Murine Model of Parkinson’s Disease. J. Neuroinflamm. 2021, 18, 20. [Google Scholar] [CrossRef]
  354. Lu, C.-S.; Chang, H.-C.; Weng, Y.-H.; Chen, C.-C.; Kuo, Y.-S.; Tsai, Y.-C. The Add-On Effect of Lactobacillus Plantarum PS128 in Patients With Parkinson’s Disease: A Pilot Study. Front. Nutr. 2021, 8, 5931–5950. [Google Scholar] [CrossRef] [PubMed]
  355. Yahfoufi, N.; Matar, C.; Ismail, N. Adolescence and Aging: Impact of Adolescence Inflammatory Stress and Microbiota Alterations on Brain Development, Aging, and Neurodegeneration. J. Gerontol. Ser. A 2020, 75, 1251–1257. [Google Scholar] [CrossRef] [Green Version]
  356. Enck, K.; Banks, S.; Yadav, H.; Welker, M.E.; Opara, E.C. Development of a Novel Oral Delivery Vehicle for Probiotics. Curr. Pharm. Des. 2020, 26, 3134–3140. [Google Scholar] [CrossRef] [PubMed]
  357. Pan, H.; Sun, T.; Cui, M.; Ma, N.; Yang, C.; Liu, J.; Pang, G.; Liu, B.; Li, L.; Zhang, X.; et al. Light-Sensitive Lactococcus Lactis for Microbe–Gut–Brain Axis Regulating via Upconversion Optogenetic Micro-Nano System. ACS Nano 2022, 16, 6049–6063. [Google Scholar] [CrossRef] [PubMed]
  358. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert Consensus Document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
  359. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, Prebiotics and Synbiotics- a Review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef] [Green Version]
  360. Wang, Q.; Luo, Y.; Ray Chaudhuri, K.; Reynolds, R.; Tan, E.-K.; Pettersson, S. The Role of Gut Dysbiosis in Parkinson’s Disease: Mechanistic Insights and Therapeutic Options. Brain 2021, 144, 2571–2593. [Google Scholar] [CrossRef]
  361. Wiciński, M.; Gębalski, J.; Mazurek, E.; Podhorecka, M.; Śniegocki, M.; Szychta, P.; Sawicka, E.; Malinowski, B. The Influence of Polyphenol Compounds on Human Gastrointestinal Tract Microbiota. Nutrients 2020, 12, 350. [Google Scholar] [CrossRef] [Green Version]
  362. de Vrese, M.; Schrezenmeir, J. Probiotics, Prebiotics, and Synbiotics. Adv. Biochem. Eng. Biotechnol. 2008, 111, 1–66. [Google Scholar] [CrossRef] [PubMed]
  363. Manzoor, S.; Wani, S.M.; Ahmad Mir, S.; Rizwan, D. Role of Probiotics and Prebiotics in Mitigation of Different Diseases. Nutrition 2022, 96, 111602. [Google Scholar] [CrossRef] [PubMed]
  364. Raval, U.; Harary, J.M.; Zeng, E.; Pasinetti, G.M. The Dichotomous Role of the Gut Microbiome in Exacerbating and Ameliorating Neurodegenerative Disorders. Expert Rev. Neurother. 2020, 20, 673–686. [Google Scholar] [CrossRef] [PubMed]
  365. Cantu-Jungles, T.M.; Rasmussen, H.E.; Hamaker, B.R. Potential of Prebiotic Butyrogenic Fibers in Parkinson’s Disease. Front. Neurol. 2019, 10, 663. [Google Scholar] [CrossRef] [Green Version]
  366. Guo, T.; Chen, L. Gut Microbiota and Inflammation in Parkinson’s Disease: Pathogenetic and Therapeutic Insights. Eur. J. Inflamm. 2022, 20. [Google Scholar] [CrossRef]
  367. Yang, H.; Liu, Y.; Cai, R.; Li, Y.; Gu, B. A Narrative Review of Relationship between Gut Microbiota and Neuropsychiatric Disorders: Mechanisms and Clinical Application of Probiotics and Prebiotics. Ann. Palliat. Med. 2021, 10, 2304–2313. [Google Scholar] [CrossRef]
  368. Lee, Y.-S.; Lai, D.-M.; Huang, H.-J.; Lee-Chen, G.-J.; Chang, C.-H.; Hsieh-Li, H.M.; Lee, G.-C. Prebiotic Lactulose Ameliorates the Cognitive Deficit in Alzheimer’s Disease Mouse Model through Macroautophagy and Chaperone-Mediated Autophagy Pathways. J. Agric. Food Chem. 2021, 69, 2422–2437. [Google Scholar] [CrossRef]
  369. Grimaldi, R.; Gibson, G.R.; Vulevic, J.; Giallourou, N.; Castro-Mejía, J.L.; Hansen, L.H.; Leigh Gibson, E.; Nielsen, D.S.; Costabile, A. A Prebiotic Intervention Study in Children with Autism Spectrum Disorders (ASDs). Microbiome 2018, 6, 133. [Google Scholar] [CrossRef] [Green Version]
  370. Chen, D.; Yang, X.; Yang, J.; Lai, G.; Yong, T.; Tang, X.; Shuai, O.; Zhou, G.; Xie, Y.; Wu, Q. Prebiotic Effect of Fructooligosaccharides from Morinda Officinalis on Alzheimer’s Disease in Rodent Models by Targeting the Microbiota-Gut-Brain Axis. Front. Aging Neurosci. 2017, 9, 403. [Google Scholar] [CrossRef] [Green Version]
  371. Xin, Y.; Diling, C.; Jian, Y.; Ting, L.; Guoyan, H.; Hualun, L.; Xiaocui, T.; Guoxiao, L.; Ou, S.; Chaoqun, Z.; et al. Effects of Oligosaccharides From Morinda Officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice. Front. Neurol. 2018, 9, 412. [Google Scholar] [CrossRef] [PubMed]
  372. Perez-Pardo, P.; de Jong, E.M.; Broersen, L.M.; van Wijk, N.; Attali, A.; Garssen, J.; Kraneveld, A.D. Promising Effects of Neurorestorative Diets on Motor, Cognitive, and Gastrointestinal Dysfunction after Symptom Development in a Mouse Model of Parkinson’s Disease. Front. Aging Neurosci. 2017, 9, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  373. Liu, X.; Du, Z.R.; Wang, X.; Sun, X.R.; Zhao, Q.; Zhao, F.; Wong, W.T.; Wong, K.H.; Dong, X.-L. Polymannuronic Acid Prebiotic plus Lacticaseibacillus Rhamnosus GG Probiotic as a Novel Synbiotic Promoted Their Separate Neuroprotection against Parkinson’s Disease. Food Res. Int. 2022, 155, 111067. [Google Scholar] [CrossRef] [PubMed]
  374. Astarloa, R.; Mena, M.A.; Sánchez, V.; de la Vega, L.; de Yébenes, J.G. Clinical and Pharmacokinetic Effects of a Diet Rich in Insoluble Fiber on Parkinson Disease. Clin. Neuropharmacol. 1992, 15, 375–380. [Google Scholar] [CrossRef] [PubMed]
  375. Ashraf, W.; Pfeiffer, R.F.; Park, F.; Lof, J.; Quigley, E.M.M. Constipation in Parkinson’s Disease: Objective Assessment and Response to Psyllium. Mov. Disord. 1997, 12, 946–951. [Google Scholar] [CrossRef] [PubMed]
  376. Nurrahma, B.A.; Tsao, S.-P.; Wu, C.-H.; Yeh, T.-H.; Hsieh, P.-S.; Panunggal, B.; Huang, H.-Y. Probiotic Supplementation Facilitates Recovery of 6-OHDA-Induced Motor Deficit via Improving Mitochondrial Function and Energy Metabolism. Front. Aging Neurosci. 2021, 13, 668775. [Google Scholar] [CrossRef]
  377. Savignac, H.M.; Corona, G.; Mills, H.; Chen, L.; Spencer, J.P.E.; Tzortzis, G.; Burnet, P.W.J. Prebiotic Feeding Elevates Central Brain Derived Neurotrophic Factor, N-Methyl-d-Aspartate Receptor Subunits and d-Serine. Neurochem. Int. 2013, 63, 756–764. [Google Scholar] [CrossRef] [Green Version]
  378. Bathina, S.; Das, U.N. Brain-Derived Neurotrophic Factor and Its Clinical Implications. Arch. Med. Sci. 2015, 6, 1164–1178. [Google Scholar] [CrossRef]
  379. St. Laurent, R.; O’Brien, L.M.; Ahmad, S.T. Sodium Butyrate Improves Locomotor Impairment and Early Mortality in a Rotenone-Induced Drosophila Model of Parkinson’s Disease. Neuroscience 2013, 246, 382–390. [Google Scholar] [CrossRef] [Green Version]
  380. Zhou, W.; Bercury, K.; Cummiskey, J.; Luong, N.; Lebin, J.; Freed, C.R. Phenylbutyrate Up-Regulates the DJ-1 Protein and Protects Neurons in Cell Culture and in Animal Models of Parkinson Disease. J. Biol. Chem. 2011, 286, 14941–14951. [Google Scholar] [CrossRef] [Green Version]
  381. Bianchi, V.E.; Herrera, P.F.; Laura, R. Effect of Nutrition on Neurodegenerative Diseases. A Systematic Review. Nutr. Neurosci. 2021, 24, 810–834. [Google Scholar] [CrossRef]
  382. Solfrizzi, V.; Custodero, C.; Lozupone, M.; Imbimbo, B.P.; Valiani, V.; Agosti, P.; Schilardi, A.; D’Introno, A.; la Montagna, M.; Calvani, M.; et al. Relationships of Dietary Patterns, Foods, and Micro- and Macronutrients with Alzheimer’s Disease and Late-Life Cognitive Disorders: A Systematic Review. J. Alzheimer’s Dis. 2017, 59, 815–849. [Google Scholar] [CrossRef] [Green Version]
  383. Metcalfe-Roach, A.; Yu, A.C.; Golz, E.; Cirstea, M.; Sundvick, K.; Kliger, D.; Foulger, L.H.; Mackenzie, M.; Finlay, B.B.; Appel-Cresswell, S. MIND and Mediterranean Diets Associated with Later Onset of Parkinson’s Disease. Mov. Disord. 2021, 36, 977–984. [Google Scholar] [CrossRef] [PubMed]
  384. Paknahad, Z.; Sheklabadi, E.; Derakhshan, Y.; Bagherniya, M.; Chitsaz, A. The Effect of the Mediterranean Diet on Cognitive Function in Patients with Parkinson’s Disease: A Randomized Clinical Controlled Trial. Complement. Ther. Med. 2020, 50, 102366. [Google Scholar] [CrossRef] [PubMed]
  385. Rusch, C.; Beke, M.; Tucciarone, L.; Nieves, C.; Ukhanova, M.; Tagliamonte, M.S.; Mai, V.; Suh, J.H.; Wang, Y.; Chiu, S.; et al. Mediterranean Diet Adherence in People With Parkinson’s Disease Reduces Constipation Symptoms and Changes Fecal Microbiota After a 5-Week Single-Arm Pilot Study. Front. Neurol. 2021, 12, 794640. [Google Scholar] [CrossRef]
  386. Cassani, E.; Barichella, M.; Ferri, V.; Pinelli, G.; Iorio, L.; Bolliri, C.; Caronni, S.; Faierman, S.A.; Mottolese, A.; Pusani, C.; et al. Dietary Habits in Parkinson’s Disease: Adherence to Mediterranean Diet. Park. Relat. Disord. 2017, 42, 40–46. [Google Scholar] [CrossRef] [PubMed]
  387. Maraki, M.I.; Yannakoulia, M.; Stamelou, M.; Stefanis, L.; Xiromerisiou, G.; Kosmidis, M.H.; Dardiotis, E.; Hadjigeorgiou, G.M.; Sakka, P.; Anastasiou, C.A.; et al. Mediterranean Diet Adherence Is Related to Reduced Probability of Prodromal Parkinson’s Disease. Mov. Disord. 2019, 34, 48–57. [Google Scholar] [CrossRef]
  388. Paknahad, Z.; Sheklabadi, E.; Moravejolahkami, A.R.; Chitsaz, A.; Hassanzadeh, A. The Effects of Mediterranean Diet on Severity of Disease and Serum Total Antioxidant Capacity (TAC) in Patients with Parkinson’s Disease: A Single Center, Randomized Controlled Trial. Nutr. Neurosci. 2022, 25, 313–320. [Google Scholar] [CrossRef] [PubMed]
  389. Alcalay, R.N.; Gu, Y.; Mejia-Santana, H.; Cote, L.; Marder, K.S.; Scarmeas, N. The Association between Mediterranean Diet Adherence and Parkinson’s Disease. Mov. Disord. 2012, 27, 771–774. [Google Scholar] [CrossRef] [Green Version]
  390. Strikwerda, A.J.; Dommershuijsen, L.J.; Ikram, M.K.; Voortman, T. Diet Quality and Risk of Parkinson’s Disease: The Rotterdam Study. Nutrients 2021, 13, 3970. [Google Scholar] [CrossRef]
  391. Yin, W.; Löf, M.; Pedersen, N.L.; Sandin, S.; Fang, F. Mediterranean Dietary Pattern at Middle Age and Risk of Parkinson’s Disease: A Swedish Cohort Study. Mov. Disord. 2021, 36, 255–260. [Google Scholar] [CrossRef] [PubMed]
  392. Agarwal, P.; Wang, Y.; Buchman, A.S.; Holland, T.M.; Bennett, D.A.; Morris, M.C. MIND Diet Associated with Reduced Incidence and Delayed Progression of Parkinsonism in Old Age. J. Nutr. Health Aging 2018, 22, 1211–1215. [Google Scholar] [CrossRef] [PubMed]
  393. Lawrie, S.; Coe, S.; Mansoubi, M.; Welch, J.; Razzaque, J.; Hu, M.T.; Dawes, H. Dietary Patterns and Nonmotor Symptoms in Parkinson’s Disease: A Cross-Sectional Analysis. J. Am. Nutr. Assoc. 2022, 1–10. [Google Scholar] [CrossRef] [PubMed]
  394. Koyuncu, H.; Fidan, V.; Toktas, H.; Binay, O.; Celik, H. Effect of Ketogenic Diet versus Regular Diet on Voice Quality of Patients with Parkinson’s Disease. Acta Neurol. Belg. 2021, 121, 1729–1732. [Google Scholar] [CrossRef]
  395. VanItallie, T.B.; Nonas, C.; di Rocco, A.; Boyar, K.; Hyams, K.; Heymsfield, S.B. Treatment of Parkinson Disease with Diet-Induced Hyperketonemia: A Feasibility Study. Neurology 2005, 64, 728–730. [Google Scholar] [CrossRef]
  396. Phillips, M.C.L.; Murtagh, D.K.J.; Gilbertson, L.J.; Asztely, F.J.S.; Lynch, C.D.P. Low-Fat versus Ketogenic Diet in Parkinson’s Disease: A Pilot Randomized Controlled Trial. Mov. Disord. 2018, 33, 1306–1314. [Google Scholar] [CrossRef] [Green Version]
  397. Tidman, M. Effects of a Ketogenic Diet on Symptoms, Biomarkers, Depression, and Anxiety in Parkinson’s Disease: A Case Study. Cureus 2022, 14, e23684. [Google Scholar] [CrossRef]
  398. Mischley, L.K.; Lau, R.C.; Bennett, R.D. Role of Diet and Nutritional Supplements in Parkinson’s Disease Progression. Oxid Med. Cell Longev. 2017, 2017, 6405278. [Google Scholar] [CrossRef] [Green Version]
  399. Kalampokini, S.; Becker, A.; Fassbender, K.; Lyros, E.; Unger, M.M. Nonpharmacological Modulation of Chronic Inflammation in Parkinson’s Disease: Role of Diet Interventions. Park. Dis. 2019, 2019, 7535472. [Google Scholar] [CrossRef] [Green Version]
  400. Jackson, A.; Forsyth, C.B.; Shaikh, M.; Voigt, R.M.; Engen, P.A.; Ramirez, V.; Keshavarzian, A. Diet in Parkinson’s Disease: Critical Role for the Microbiome. Front. Neurol. 2019, 10, 1245. [Google Scholar] [CrossRef]
  401. Sofi, F.; Cesari, F.; Abbate, R.; Gensini, G.F.; Casini, A. Adherence to Mediterranean Diet and Health Status: Meta-Analysis. BMJ 2008, 337, a1344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  402. Solch, R.J.; Aigbogun, J.O.; Voyiadjis, A.G.; Talkington, G.M.; Darensbourg, R.M.; O’Connell, S.; Pickett, K.M.; Perez, S.R.; Maraganore, D.M. Mediterranean Diet Adherence, Gut Microbiota, and Alzheimer’s or Parkinson’s Disease Risk: A Systematic Review. J. Neurol. Sci. 2022, 434, 120166. [Google Scholar] [CrossRef] [PubMed]
  403. Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet; A Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  404. Ma, W.; Nguyen, L.H.; Song, M.; Wang, D.D.; Franzosa, E.A.; Cao, Y.; Joshi, A.; Drew, D.A.; Mehta, R.; Ivey, K.L.; et al. Dietary Fiber Intake, the Gut Microbiome, and Chronic Systemic Inflammation in a Cohort of Adult Men. Genome Med. 2021, 13, 102. [Google Scholar] [CrossRef]
  405. Koh, A.; de Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [Green Version]
  406. Garcia-Mantrana, I.; Selma-Royo, M.; Alcantara, C.; Collado, M.C. Shifts on Gut Microbiota Associated to Mediterranean Diet Adherence and Specific Dietary Intakes on General Adult Population. Front. Microbiol. 2018, 9, 890. [Google Scholar] [CrossRef] [Green Version]
  407. Baert, F.; Matthys, C.; Mellaerts, R.; Lemaître, D.; Vlaemynck, G.; Foulon, V. Dietary Intake of Parkinson’s Disease Patients. Front. Nutr. 2020, 7, 105. [Google Scholar] [CrossRef]
  408. Yang, X.; Feng, P.; Ji, R.; Ren, Y.; Wei, W.; Hölscher, C. Therapeutic Application of GLP-1 and GIP Receptor Agonists in Parkinson’s Disease. Expert Opin. Ther. Targets 2022, 26, 445–460. [Google Scholar] [CrossRef]
  409. Palasz, E.; Wysocka, A.; Gasiorowska, A.; Chalimoniuk, M.; Niewiadomski, W.; Niewiadomska, G. BDNF as a Promising Therapeutic Agent in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 1170. [Google Scholar] [CrossRef] [Green Version]
  410. Dong, Y.; Chen, L.; Gutin, B.; Zhu, H. Total, Insoluble, and Soluble Dietary Fiber Intake and Insulin Resistance and Blood Pressure in Adolescents. Eur. J. Clin. Nutr. 2019, 73, 1172–1178. [Google Scholar] [CrossRef]
  411. Namsi, A.; Nury, T.; Hamdouni, H.; Yammine, A.; Vejux, A.; Vervandier-Fasseur, D.; Latruffe, N.; Masmoudi-Kouki, O.; Lizard, G. Induction of Neuronal Differentiation of Murine N2a Cells by Two Polyphenols Present in the Mediterranean Diet Mimicking Neurotrophins Activities: Resveratrol and Apigenin. Diseases 2018, 6, 67. [Google Scholar] [CrossRef] [Green Version]
  412. Meng, X.; Munishkina, L.A.; Fink, A.L.; Uversky, V.N. Molecular Mechanisms Underlying the Flavonoid-Induced Inhibition of α-Synuclein Fibrillation. Biochemistry 2009, 48, 8206–8224. [Google Scholar] [CrossRef] [PubMed]
  413. Gao, X.; Cassidy, A.; Schwarzschild, M.A.; Rimm, E.B.; Ascherio, A. Habitual Intake of Dietary Flavonoids and Risk of Parkinson Disease. Neurology 2012, 78, 1138–1145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  414. Jung, U.J.; Kim, S.R. Beneficial Effects of Flavonoids Against Parkinson’s Disease. J. Med. Food 2018, 21, 421–432. [Google Scholar] [CrossRef] [PubMed]
  415. Giuliano, C.; Cerri, S.; Blandini, F. Potential Therapeutic Effects of Polyphenols in Parkinson’s Disease: In Vivo and in Vitro Pre-Clinical Studies. Neural Regen Res. 2021, 16, 234. [Google Scholar] [CrossRef] [PubMed]
  416. di Rosa, G.; Brunetti, G.; Scuto, M.; Trovato Salinaro, A.; Calabrese, E.J.; Crea, R.; Schmitz-Linneweber, C.; Calabrese, V.; Saul, N. Healthspan Enhancement by Olive Polyphenols in C. Elegans Wild Type and Parkinson’s Models. Int. J. Mol. Sci. 2020, 21, 3893. [Google Scholar] [CrossRef]
  417. Gallardo-Fernández, M.; Hornedo-Ortega, R.; Alonso-Bellido, I.M.; Rodríguez-Gómez, J.A.; Troncoso, A.M.; García-Parrilla, M.C.; Venero, J.L.; Espinosa-Oliva, A.M.; de Pablos, R.M. Hydroxytyrosol Decreases LPS- and α-Synuclein-Induced Microglial Activation In Vitro. Antioxidants 2019, 9, 36. [Google Scholar] [CrossRef] [Green Version]
  418. Brunetti, G.; di Rosa, G.; Scuto, M.; Leri, M.; Stefani, M.; Schmitz-Linneweber, C.; Calabrese, V.; Saul, N. Healthspan Maintenance and Prevention of Parkinson’s-like Phenotypes with Hydroxytyrosol and Oleuropein Aglycone in C. Elegans. Int. J. Mol. Sci. 2020, 21, 2588. [Google Scholar] [CrossRef] [Green Version]
  419. Godos, J.; Tieri, M.; Ghelfi, F.; Titta, L.; Marventano, S.; Lafranconi, A.; Gambera, A.; Alonzo, E.; Sciacca, S.; Buscemi, S.; et al. Dairy Foods and Health: An Umbrella Review of Observational Studies. Int. J. Food Sci. Nutr. 2020, 71, 138–151. [Google Scholar] [CrossRef] [PubMed]
  420. Comerford, K.; Pasin, G. Gene–Dairy Food Interactions and Health Outcomes: A Review of Nutrigenetic Studies. Nutrients 2017, 9, 710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  421. Paganoni, S.; Schwarzschild, M.A. Urate as a Marker of Risk and Progression of Neurodegenerative Disease. Neurotherapeutics 2017, 14, 148–153. [Google Scholar] [CrossRef] [PubMed]
  422. Dardiotis, E.; Xiromerisiou, G.; Hadjichristodoulou, C.; Tsatsakis, A.M.; Wilks, M.F.; Hadjigeorgiou, G.M. The Interplay between Environmental and Genetic Factors in Parkinson’s Disease Susceptibility: The Evidence for Pesticides. Toxicology 2013, 307, 17–23. [Google Scholar] [CrossRef] [PubMed]
  423. Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Barnes, L.L.; Bennett, D.A.; Aggarwal, N.T. MIND Diet Slows Cognitive Decline with Aging. Alzheimer’s Dement. 2015, 11, 1015–1022. [Google Scholar] [CrossRef] [Green Version]
  424. Pietrzak, D.; Kasperek, K.; Rękawek, P.; Piątkowska-Chmiel, I. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients 2022, 14, 1952. [Google Scholar] [CrossRef]
  425. Włodarek, D. Role of Ketogenic Diets in Neurodegenerative Diseases (Alzheimer’s Disease and Parkinson’s Disease). Nutrients 2019, 11, 169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  426. Hartman, A.L.; Vining, E.P.G. Clinical Aspects of the Ketogenic Diet. Epilepsia 2007, 48, 31–42. [Google Scholar] [CrossRef]
  427. Cheng, B.; Yang, X.; An, L.; Gao, B.; Liu, X.; Liu, S. Ketogenic Diet Protects Dopaminergic Neurons against 6-OHDA Neurotoxicity via up-Regulating Glutathione in a Rat Model of Parkinson’s Disease. Brain Res. 2009, 1286, 25–31. [Google Scholar] [CrossRef] [PubMed]
  428. Yang, X.; Cheng, B. Neuroprotective and Anti-Inflammatory Activities of Ketogenic Diet on MPTP-Induced Neurotoxicity. J. Mol. Neurosci. 2010, 42, 145–153. [Google Scholar] [CrossRef] [PubMed]
  429. Shaafi, S.; Najmi, S.; Aliasgharpour, H.; Mahmoudi, J.; Sadigh-Etemad, S.; Farhoudi, M.; Baniasadi, N. The Efficacy of the Ketogenic Diet on Motor Functions in Parkinson’s Disease: A Rat Model. Iran J. Neurol. 2016, 15, 63–69. [Google Scholar]
  430. Tieu, K.; Perier, C.; Caspersen, C.; Teismann, P.; Wu, D.-C.; Yan, S.-D.; Naini, A.; Vila, M.; Jackson-Lewis, V.; Ramasamy, R.; et al. D-β-Hydroxybutyrate Rescues Mitochondrial Respiration and Mitigates Features of Parkinson Disease. J. Clin. Investig. 2003, 112, 892–901. [Google Scholar] [CrossRef] [Green Version]
  431. Cornille, E.; Abou-Hamdan, M.; Khrestchatisky, M.; Nieoullon, A.; de Reggi, M.; Gharib, B. Enhancement of L-3-Hydroxybutyryl-CoA Dehydrogenase Activity and Circulating Ketone Body Levels by Pantethine. Relevance to Dopaminergic Injury. BMC Neurosci. 2010, 11, 51. [Google Scholar] [CrossRef]
  432. Pinto, A.; Bonucci, A.; Maggi, E.; Corsi, M.; Businaro, R. Anti-Oxidant and Anti-Inflammatory Activity of Ketogenic Diet: New Perspectives for Neuroprotection in Alzheimer’s Disease. Antioxidants 2018, 7, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  433. Jensen, N.J.; Wodschow, H.Z.; Nilsson, M.; Rungby, J. Effects of Ketone Bodies on Brain Metabolism and Function in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 8767. [Google Scholar] [CrossRef]
  434. Huang, C.; Wang, P.; Xu, X.; Zhang, Y.; Gong, Y.; Hu, W.; Gao, M.; Wu, Y.; Ling, Y.; Zhao, X.; et al. The Ketone Body Metabolite β-Hydroxybutyrate Induces an Antidepression-Associated Ramification of Microglia via HDACs Inhibition-Triggered Akt-Small RhoGTPase Activation. Glia 2018, 66, 256–278. [Google Scholar] [CrossRef] [PubMed]
  435. Youm, Y.-H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The Ketone Metabolite β-Hydroxybutyrate Blocks NLRP3 Inflammasome–Mediated Inflammatory Disease. Nat. Med. 2015, 21, 263–269. [Google Scholar] [CrossRef] [PubMed]
  436. Shimazu, T.; Hirschey, M.D.; Newman, J.; He, W.; Shirakawa, K.; le Moan, N.; Grueter, C.A.; Lim, H.; Saunders, L.R.; Stevens, R.D.; et al. Suppression of Oxidative Stress by β-Hydroxybutyrate, an Endogenous Histone Deacetylase Inhibitor. Science (1979) 2013, 339, 211–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  437. Fu, S.-P.; Wang, J.-F.; Xue, W.-J.; Liu, H.-M.; Liu, B.; Zeng, Y.-L.; Li, S.-N.; Huang, B.-X.; Lv, Q.-K.; Wang, W.; et al. Anti-Inflammatory Effects of BHBA in Both in Vivo and in Vitro Parkinson’s Disease Models Are Mediated by GPR109A-Dependent Mechanisms. J. Neuroinflamm. 2015, 12, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  438. Qiao, G.; Lv, T.; Zhang, M.; Chen, P.; Sun, Q.; Zhang, J.; Li, Q. β-Hydroxybutyrate (β-HB) Exerts Anti-Inflammatory and Antioxidant Effects in Lipopolysaccharide (LPS)-Stimulated Macrophages in Liza Haematocheila. Fish Shellfish. Immunol. 2020, 107, 444–451. [Google Scholar] [CrossRef]
  439. Elbarbry, F.; Nguyen, V.; Mirka, A.; Zwickey, H.; Rosenbaum, R. A New Validated HPLC Method for the Determination of Levodopa: Application to Study the Impact of Ketogenic Diet on the Pharmacokinetics of Levodopa in Parkinson’s Participants. Biomed. Chromatogr. 2019, 33, e4382. [Google Scholar] [CrossRef] [Green Version]
  440. Calder, P.C. Omega-3 Fatty Acids and Inflammatory Processes. Nutrients 2010, 2, 355–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  441. Avallone, R.; Vitale, G.; Bertolotti, M. Omega-3 Fatty Acids and Neurodegenerative Diseases: New Evidence in Clinical Trials. Int. J. Mol. Sci. 2019, 20, 4256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  442. de Lau, L.M.L.; Bornebroek, M.; Witteman, J.C.M.; Hofman, A.; Koudstaal, P.J.; Breteler, M.M.B. Dietary Fatty Acids and the Risk of Parkinson Disease. Neurology 2005, 64, 2040–2045. [Google Scholar] [CrossRef] [PubMed]
  443. Abbott, R.D.; Webster Ross, G.; White, L.R.; Sanderson, W.T.; Burchfiel, C.M.; Kashon, M.; Sharp, D.S.; Masaki, K.H.; Curb, J.D.; Petrovitch, H. Environmental, Life-Style, and Physical Precursors of Clinical Parkinson?S Disease: Recent Findings from the Honolulu-Asia Aging Study. J. Neurol. 2003, 250, iii30–iii39. [Google Scholar] [CrossRef] [PubMed]
  444. Pantzaris, M.; Loukaides, G.; Paraskevis, D.; Kostaki, E.-G.; Patrikios, I. Neuroaspis PLP10TM, a Nutritional Formula Rich in Omega-3 and Omega-6 Fatty Acids with Antioxidant Vitamins Including Gamma-Tocopherol in Early Parkinson’s Disease: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Neurol. Neurosurg. 2021, 210, 106954. [Google Scholar] [CrossRef] [PubMed]
  445. Reijnders, J.S.A.M.; Ehrt, U.; Weber, W.E.J.; Aarsland, D.; Leentjens, A.F.G. A Systematic Review of Prevalence Studies of Depression in Parkinson’s Disease. Mov. Disord. 2008, 23, 183–189. [Google Scholar] [CrossRef] [PubMed]
  446. Murakami, K.; Sasaki, S. Dietary Intake and Depressive Symptoms: A Systematic Review of Observational Studies. Mol. Nutr. Food Res. 2010, 54, 471–488. [Google Scholar] [CrossRef] [PubMed]
  447. Grosso, G.; Micek, A.; Marventano, S.; Castellano, S.; Mistretta, A.; Pajak, A.; Galvano, F. Dietary N-3 PUFA, Fish Consumption and Depression: A Systematic Review and Meta-Analysis of Observational Studies. J. Affect Disord. 2016, 205, 269–281. [Google Scholar] [CrossRef] [PubMed]
  448. Assies, J.; Lok, A.; Bockting, C.L.; Weverling, G.J.; Lieverse, R.; Visser, I.; Abeling, N.G.G.M.; Duran, M.; Schene, A.H. Fatty Acids and Homocysteine Levels in Patients with Recurrent Depression: An Explorative Pilot Study. Prostaglandins Leukot Essent Fat. Acids 2004, 70, 349–356. [Google Scholar] [CrossRef]
  449. Sánchez-Villegas, A.; Álvarez-Pérez, J.; Toledo, E.; Salas-Salvadó, J.; Ortega-Azorín, C.; Zomeño, M.; Vioque, J.; Martínez, J.; Romaguera, D.; Pérez-López, J.; et al. Seafood Consumption, Omega-3 Fatty Acids Intake, and Life-Time Prevalence of Depression in the PREDIMED-Plus Trial. Nutrients 2018, 10, 2000. [Google Scholar] [CrossRef] [Green Version]
  450. da Silva, T.M.; Munhoz, R.P.; Alvarez, C.; Naliwaiko, K.; Kiss, Á.; Andreatini, R.; Ferraz, A.C. Depression in Parkinson’s Disease: A Double-Blind, Randomized, Placebo-Controlled Pilot Study of Omega-3 Fatty-Acid Supplementation. J. Affect Disord. 2008, 111, 351–359. [Google Scholar] [CrossRef]
  451. Li, P.; Song, C. Potential Treatment of Parkinson’s Disease with Omega-3 Polyunsaturated Fatty Acids. Nutr. Neurosci. 2022, 25, 180–191. [Google Scholar] [CrossRef]
  452. Kujawska, M.; Domanskyi, A.; Kreiner, G. Editorial: Common Pathways Linking Neurodegenerative Diseases—The Role of Inflammation. Front. Cell. Neurosci. 2021, 15, 754051. [Google Scholar] [CrossRef] [PubMed]
  453. Subramaniam, S.R.; Federoff, H.J. Targeting Microglial Activation States as a Therapeutic Avenue in Parkinson’s Disease. Front. Aging Neurosci. 2017, 9, 176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  454. Wijeyekoon, R.S.; Moore, S.F.; Farrell, K.; Breen, D.P.; Barker, R.A.; Williams-Gray, C.H. Cerebrospinal Fluid Cytokines and Neurodegeneration-Associated Proteins in Parkinson’s Disease. Mov. Disord. 2020, 35, 1062–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  455. Tansey, M.G.; Goldberg, M.S. Neuroinflammation in Parkinson’s Disease: Its Role in Neuronal Death and Implications for Therapeutic Intervention. Neurobiol. Dis. 2010, 37, 510–518. [Google Scholar] [CrossRef]
  456. Hirsch, E.C.; Hunot, S. Neuroinflammation in Parkinson’s Disease: A Target for Neuroprotection? Lancet Neurol. 2009, 8, 382–397. [Google Scholar] [CrossRef]
  457. Teleanu, D.M.; Niculescu, A.-G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef]
  458. Taghizadeh, M.; Tamtaji, O.R.; Dadgostar, E.; Daneshvar Kakhaki, R.; Bahmani, F.; Abolhassani, J.; Aarabi, M.H.; Kouchaki, E.; Memarzadeh, M.R.; Asemi, Z. The Effects of Omega-3 Fatty Acids and Vitamin E Co-Supplementation on Clinical and Metabolic Status in Patients with Parkinson’s Disease: A Randomized, Double-Blind, Placebo-Controlled Trial. Neurochem. Int. 2017, 108, 183–189. [Google Scholar] [CrossRef]
  459. Cardoso, H.D.; dos Santos Junior, E.F.; de Santana, D.F.; Gonçalves-Pimentel, C.; Angelim, M.K.; Isaac, A.R.; Lagranha, C.J.; Guedes, R.C.A.; Beltrão, E.I.; Morya, E.; et al. Omega-3 Deficiency and Neurodegeneration in the Substantia Nigra: Involvement of Increased Nitric Oxide Production and Reduced BDNF Expression. Biochim. Et Biophys. Acta (BBA)-Gen. Subj. 2014, 1840, 1902–1912. [Google Scholar] [CrossRef]
  460. Mori, M.A.; Delattre, A.M.; Carabelli, B.; Pudell, C.; Bortolanza, M.; Staziaki, P.V.; Visentainer, J.V.; Montanher, P.F.; del Bel, E.A.; Ferraz, A.C. Neuroprotective Effect of Omega-3 Polyunsaturated Fatty Acids in the 6-OHDA Model of Parkinson’s Disease Is Mediated by a Reduction of Inducible Nitric Oxide Synthase. Nutr. Neurosci. 2018, 21, 341–351. [Google Scholar] [CrossRef]
  461. Healy-Stoffel, M.; Levant, B. N-3 (Omega-3) Fatty Acids: Effects on Brain Dopamine Systems and Potential Role in the Etiology and Treatment of Neuropsychiatric Disorders. CNS Neurol. Disord. Drug Targets 2018, 17, 216–232. [Google Scholar] [CrossRef] [PubMed]
  462. Gupta, A.; Khanna, S. Fecal Microbiota Transplantation. JAMA 2017, 318, 102. [Google Scholar] [CrossRef] [PubMed]
  463. Tan, P.; Li, X.; Shen, J.; Feng, Q. Fecal Microbiota Transplantation for the Treatment of Inflammatory Bowel Disease: An Update. Front. Pharmacol. 2020, 11, 574533. [Google Scholar] [CrossRef] [PubMed]
  464. Ramai, D. Fecal Microbiota Transplantation: Donor Relation, Fresh or Frozen, Delivery Methods, Cost-Effectiveness. Ann. Gastroenterol. 2018, 32, 30–38. [Google Scholar] [CrossRef]
  465. Vendrik, K.E.W.; Ooijevaar, R.E.; de Jong, P.R.C.; Laman, J.D.; van Oosten, B.W.; van Hilten, J.J.; Ducarmon, Q.R.; Keller, J.J.; Kuijper, E.J.; Contarino, M.F. Fecal Microbiota Transplantation in Neurological Disorders. Front. Cell. Infect Microbiol. 2020, 10, 98. [Google Scholar] [CrossRef]
  466. Kang, D.-W.; Adams, J.B.; Gregory, A.C.; Borody, T.; Chittick, L.; Fasano, A.; Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S.; et al. Microbiota Transfer Therapy Alters Gut Ecosystem and Improves Gastrointestinal and Autism Symptoms: An Open-Label Study. Microbiome 2017, 5, 10. [Google Scholar] [CrossRef]
  467. Evrensel, A.; Ceylan, M.E. Fecal Microbiota Transplantation and Its Usage in Neuropsychiatric Disorders. Clin. Psychopharmacol. Neurosci. 2016, 14, 231–237. [Google Scholar] [CrossRef] [Green Version]
  468. Xu, M.-Q. Fecal Microbiota Transplantation Broadening Its Application beyond Intestinal Disorders. World J. Gastroenterol. 2015, 21, 102. [Google Scholar] [CrossRef]
  469. Sun, M.-F.; Zhu, Y.-L.; Zhou, Z.-L.; Jia, X.-B.; Xu, Y.-D.; Yang, Q.; Cui, C.; Shen, Y.-Q. Neuroprotective Effects of Fecal Microbiota Transplantation on MPTP-Induced Parkinson’s Disease Mice: Gut Microbiota, Glial Reaction and TLR4/TNF-α Signaling Pathway. Brain Behav. Immun. 2018, 70, 48–60. [Google Scholar] [CrossRef]
  470. Zhao, Z.; Ning, J.; Bao, X.; Shang, M.; Ma, J.; Li, G.; Zhang, D. Fecal Microbiota Transplantation Protects Rotenone-Induced Parkinson’s Disease Mice via Suppressing Inflammation Mediated by the Lipopolysaccharide-TLR4 Signaling Pathway through the Microbiota-Gut-Brain Axis. Microbiome 2021, 9, 226. [Google Scholar] [CrossRef]
  471. Zhong, Z.; Chen, W.; Gao, H.; Che, N.; Xu, M.; Yang, L.; Zhang, Y.; Ye, M. Fecal Microbiota Transplantation Exerts a Protective Role in MPTP-Induced Parkinson’s Disease via the TLR4/PI3K/AKT/NF-ΚB Pathway Stimulated by α-Synuclein. Neurochem. Res. 2021, 46, 3050–3058. [Google Scholar] [CrossRef] [PubMed]
  472. Zhang, T.; Wang, T.; Chen, X.; Zhao, Z.; Chen, Z. Gut Microbiota Relieves Inflammation in the Substantia Nigra of Chronic Parkinson’s Disease by Protecting the Function of Dopamine Neurons. Exp. Ther. Med. 2021, 23, 52. [Google Scholar] [CrossRef]
  473. Zhou, Z.-L.; Jia, X.-B.; Sun, M.-F.; Zhu, Y.-L.; Qiao, C.-M.; Zhang, B.-P.; Zhao, L.-P.; Yang, Q.; Cui, C.; Chen, X.; et al. Neuroprotection of Fasting Mimicking Diet on MPTP-Induced Parkinson’s Disease Mice via Gut Microbiota and Metabolites. Neurotherapeutics 2019, 16, 741–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  474. Huang, H.; Xu, H.; Luo, Q.; He, J.; Li, M.; Chen, H.; Tang, W.; Nie, Y.; Zhou, Y. Fecal Microbiota Transplantation to Treat Parkinson’s Disease with Constipation. Medicine 2019, 98, e16163. [Google Scholar] [CrossRef] [PubMed]
  475. Kuai, X.; Yao, X.; Xu, L.; Zhou, Y.; Zhang, L.; Liu, Y.; Pei, S.; Zhou, C. Evaluation of Fecal Microbiota Transplantation in Parkinson’s Disease Patients with Constipation. Microb. Cell. Fact 2021, 20, 98. [Google Scholar] [CrossRef] [PubMed]
  476. Xue, L.-J.; Yang, X.-Z.; Tong, Q.; Shen, P.; Ma, S.-J.; Wu, S.-N.; Zheng, J.-L.; Wang, H.-G. Fecal Microbiota Transplantation Therapy for Parkinson’s Disease. Medicine 2020, 99, e22035. [Google Scholar] [CrossRef] [PubMed]
  477. Segal, A.; Zlotnik, Y.; Moyal-Atias, K.; Abuhasira, R.; Ifergane, G. Fecal Microbiota Transplant as a Potential Treatment for Parkinson’s Disease—A Case Series. Clin. Neurol. Neurosurg. 2021, 207, 106791. [Google Scholar] [CrossRef]
  478. Knott, C.; Stern, G.; Wilkin, G.P. Inflammatory Regulators in Parkinson’s Disease: INOS, Lipocortin-1, and Cyclooxygenases-1 and -2. Mol. Cell. Neurosci. 2000, 16, 724–739. [Google Scholar] [CrossRef]
  479. Li, J.; Ma, S.; Chen, J.; Hu, K.; Li, Y.; Zhang, Z.; Su, Z.; Woodgett, J.R.; Li, M.; Huang, Q. GSK-3β Contributes to Parkinsonian Dopaminergic Neuron Death: Evidence From Conditional Knockout Mice and Tideglusib. Front. Mol. Neurosci. 2020, 13, 81. [Google Scholar] [CrossRef]
  480. 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] [Green Version]
  481. Choi, H.H.; Cho, Y.-S. Fecal Microbiota Transplantation: Current Applications, Effectiveness, and Future Perspectives. Clin. Endosc. 2016, 49, 257–265. [Google Scholar] [CrossRef] [PubMed]
  482. Schmulson, M.; Bashashati, M. Fecal Microbiota Transfer for Bowel Disorders: Efficacy or Hype? Curr. Opin. Pharmacol. 2018, 43, 72–80. [Google Scholar] [CrossRef] [PubMed]
  483. Shanahan, F.; Quigley, E.M.M. Manipulation of the Microbiota for Treatment of IBS and IBD—Challenges and Controversies. Gastroenterology 2014, 146, 1554–1563. [Google Scholar] [CrossRef] [PubMed]
  484. Lopetuso, L.R.; Ianiro, G.; Allegretti, J.R.; Bibbò, S.; Gasbarrini, A.; Scaldaferri, F.; Cammarota, G. Fecal Transplantation for Ulcerative Colitis: Current Evidence and Future Applications. Expert Opin. Biol. Ther. 2020, 20, 343–351. [Google Scholar] [CrossRef]
  485. Wang, J.-W.; Kuo, C.-H.; Kuo, F.-C.; Wang, Y.-K.; Hsu, W.-H.; Yu, F.-J.; Hu, H.-M.; Hsu, P.-I.; Wang, J.-Y.; Wu, D.-C. Fecal Microbiota Transplantation: Review and Update. J. Formos. Med. Assoc. 2019, 118, S23–S31. [Google Scholar] [CrossRef]
  486. Aroniadis, O.C.; Brandt, L.J. Fecal Microbiota Transplantation. Curr. Opin. Gastroenterol. 2013, 29, 79–84. [Google Scholar] [CrossRef]
  487. Singh, H.; Torralba, M.G.; Moncera, K.J.; DiLello, L.; Petrini, J.; Nelson, K.E.; Pieper, R. Gastro-Intestinal and Oral Microbiome Signatures Associated with Healthy Aging. Geroscience 2019, 41, 907–921. [Google Scholar] [CrossRef]
  488. Irfan, M.; Delgado, R.Z.R.; Frias-Lopez, J. The Oral Microbiome and Cancer. Front. Immunol. 2020, 11, 591088. [Google Scholar] [CrossRef]
  489. Hayes, R.B.; Ahn, J.; Fan, X.; Peters, B.A.; Ma, Y.; Yang, L.; Agalliu, I.; Burk, R.D.; Ganly, I.; Purdue, M.P.; et al. Association of Oral Microbiome With Risk for Incident Head and Neck Squamous Cell Cancer. JAMA Oncol. 2018, 4, 358. [Google Scholar] [CrossRef]
  490. Pathak, J.L.; Yan, Y.; Zhang, Q.; Wang, L.; Ge, L. The Role of Oral Microbiome in Respiratory Health and Diseases. Respir. Med. 2021, 185, 106475. [Google Scholar] [CrossRef]
  491. Sureda, A.; Daglia, M.; Argüelles Castilla, S.; Sanadgol, N.; Fazel Nabavi, S.; Khan, H.; Belwal, T.; Jeandet, P.; Marchese, A.; Pistollato, F.; et al. Oral Microbiota and Alzheimer’s Disease: Do All Roads Lead to Rome? Pharmacol. Res. 2020, 151, 104582. [Google Scholar] [CrossRef] [PubMed]
  492. Shoemark, D.K.; Allen, S.J. The Microbiome and Disease: Reviewing the Links between the Oral Microbiome, Aging, and Alzheimer’s Disease. J. Alzheimer’s Dis. 2014, 43, 725–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  493. Yang, I.; Arthur, R.A.; Zhao, L.; Clark, J.; Hu, Y.; Corwin, E.J.; Lah, J. The Oral Microbiome and Inflammation in Mild Cognitive Impairment. Exp. Gerontol. 2021, 147, 111273. [Google Scholar] [CrossRef] [PubMed]
  494. Yussof, A.; Yoon, P.; Krkljes, C.; Schweinberg, S.; Cottrell, J.; Chu, T.; Chang, S.L. A Meta-Analysis of the Effect of Binge Drinking on the Oral Microbiome and Its Relation to Alzheimer’s Disease. Sci. Rep. 2020, 10, 19872. [Google Scholar] [CrossRef] [PubMed]
  495. Cirstea, M.S.; Kliger, D.; MacLellan, A.D.; Yu, A.C.; Langlois, J.; Fan, M.; Boroomand, S.; Kharazyan, F.; Hsiung, R.G.Y.; MacVicar, B.A.; et al. The Oral and Fecal Microbiota in a Canadian Cohort of Alzheimer’s Disease. J. Alzheimer’s Dis. 2022, 87, 247–258. [Google Scholar] [CrossRef]
  496. Rozas, N.S.; Tribble, G.D.; Jeter, C.B. Oral Factors That Impact the Oral Microbiota in Parkinson’s Disease. Microorganisms 2021, 9, 1616. [Google Scholar] [CrossRef] [PubMed]
  497. Mihaila, D.; Donegan, J.; Barns, S.; LaRocca, D.; Du, Q.; Zheng, D.; Vidal, M.; Neville, C.; Uhlig, R.; Middleton, F.A. The Oral Microbiome of Early Stage Parkinson’s Disease and Its Relationship with Functional Measures of Motor and Non-Motor Function. PLoS ONE 2019, 14, e0218252. [Google Scholar] [CrossRef]
  498. Jo, S.; Kang, W.; Hwang, Y.S.; Lee, S.H.; Park, K.W.; Kim, M.S.; Lee, H.; Yoon, H.J.; Park, Y.K.; Chalita, M.; et al. Oral and Gut Dysbiosis Leads to Functional Alterations in Parkinson’s Disease. NPJ Park. Dis. 2022, 8, 87. [Google Scholar] [CrossRef] [PubMed]
  499. Fleury, V.; Zekeridou, A.; Lazarevic, V.; Gaïa, N.; Giannopoulou, C.; Genton, L.; Cancela, J.; Girard, M.; Goldstein, R.; Bally, J.F.; et al. Oral Dysbiosis and Inflammation in Parkinson’s Disease. J. Park. Dis. 2021, 11, 619–631. [Google Scholar] [CrossRef]
  500. Zapała, B.; Stefura, T.; Milewicz, T.; Wątor, J.; Piwowar, M.; Wójcik-Pędziwiatr, M.; Doręgowska, M.; Dudek, A.; Jania, Z.; Rudzińska-Bar, M. The Role of the Western Diet and Oral Microbiota in Parkinson’s Disease. Nutrients 2022, 14, 355. [Google Scholar] [CrossRef]
  501. Lamont, R.J.; Koo, H.; Hajishengallis, G. The Oral Microbiota: Dynamic Communities and Host Interactions. Nat. Rev. Microbiol. 2018, 16, 745–759. [Google Scholar] [CrossRef] [PubMed]
  502. Feng, Y.-K.; Wu, Q.-L.; Peng, Y.-W.; Liang, F.-Y.; You, H.-J.; Feng, Y.-W.; Li, G.; Li, X.-J.; Liu, S.-H.; Li, Y.-C.; et al. Oral P. Gingivalis Impairs Gut Permeability and Mediates Immune Responses Associated with Neurodegeneration in LRRK2 R1441G Mice. J. Neuroinflamm. 2020, 17, 347. [Google Scholar] [CrossRef]
  503. Pal, G.; Ramirez, V.; Engen, P.A.; Naqib, A.; Forsyth, C.B.; Green, S.J.; Mahdavinia, M.; Batra, P.S.; Tajudeen, B.A.; Keshavarzian, A. Deep Nasal Sinus Cavity Microbiota Dysbiosis in Parkinson’s Disease. NPJ Park. Dis. 2021, 7, 111. [Google Scholar] [CrossRef]
  504. Xie, J.; Tian, S.; Liu, J.; Cao, R.; Yue, P.; Cai, X.; Shang, Q.; Yang, M.; Han, L.; Zhang, D. Dual Role of the Nasal Microbiota in Neurological Diseases—An Unignorable Risk Factor or a Potential Therapy Carrier. Pharmacol. Res. 2022, 179, 106189. [Google Scholar] [CrossRef]
  505. Wagner Mackenzie, B.; Chang, K.; Zoing, M.; Jain, R.; Hoggard, M.; Biswas, K.; Douglas, R.G.; Taylor, M.W. Longitudinal Study of the Bacterial and Fungal Microbiota in the Human Sinuses Reveals Seasonal and Annual Changes in Diversity. Sci. Rep. 2019, 9, 17416. [Google Scholar] [CrossRef] [PubMed]
  506. Shen, L. Gut, Oral and Nasal Microbiota and Parkinson’s Disease. Microb. Cell. Fact 2020, 19, 50. [Google Scholar] [CrossRef] [Green Version]
  507. Boertien, J.M.; Pereira, P.A.B.; Aho, V.T.E.; Scheperjans, F. Increasing Comparability and Utility of Gut Microbiome Studies in Parkinson’s Disease: A Systematic Review. J. Park. Dis. 2019, 9, S297–S312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  508. Conlon, M.; Bird, A. The Impact of Diet and Lifestyle on Gut Microbiota and Human Health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
  509. Aslam, H.; Marx, W.; Rocks, T.; Loughman, A.; Chandrasekaran, V.; Ruusunen, A.; Dawson, S.L.; West, M.; Mullarkey, E.; Pasco, J.A.; et al. The Effects of Dairy and Dairy Derivatives on the Gut Microbiota: A Systematic Literature Review. Gut Microbes 2020, 12, 1799533. [Google Scholar] [CrossRef] [PubMed]
  510. Cortes-Ortiz, L.; Amato, K.R. Host Genetics Influence the Gut Microbiome. Science (1979) 2021, 373, 159–160. [Google Scholar] [CrossRef] [PubMed]
  511. Jain, N. The Need for Personalized Approaches to Microbiome Modulation. Front. Public Health 2020, 8, 144. [Google Scholar] [CrossRef] [PubMed]
  512. Vandeputte, D. Personalized Nutrition Through The Gut Microbiota: Current Insights And Future Perspectives. Nutr. Rev. 2020, 78, 66–74. [Google Scholar] [CrossRef]
  513. Molsberry, S.; Bjornevik, K.; Hughes, K.C.; Healy, B.; Schwarzschild, M.; Ascherio, A. Diet Pattern and Prodromal Features of Parkinson Disease. Neurology 2020, 95, e2095–e2108. [Google Scholar] [CrossRef]
Figure 1. The gut microbiota brain axis (GMBA) in Parkinson’s disease (PD). In PD, α-synuclein aggregates are retrieved both in the dopaminergic neurons of the substantia nigra, as well as in the gut, and often associate to intestinal dysbiosis. Dysregulations in the gut microbiota composition trigger chronic inflammation and blood brain barrier (BBB) disruption through increased gut permeability (the so-called leaky gut). The resulting neuroinflammation then directly or indirectly promotes cognitive decline and motor dysfunction, which are the typical PD manifestations. ↑: increase.
Figure 1. The gut microbiota brain axis (GMBA) in Parkinson’s disease (PD). In PD, α-synuclein aggregates are retrieved both in the dopaminergic neurons of the substantia nigra, as well as in the gut, and often associate to intestinal dysbiosis. Dysregulations in the gut microbiota composition trigger chronic inflammation and blood brain barrier (BBB) disruption through increased gut permeability (the so-called leaky gut). The resulting neuroinflammation then directly or indirectly promotes cognitive decline and motor dysfunction, which are the typical PD manifestations. ↑: increase.
Ijms 23 12289 g001
Figure 2. Gut microbiota (GM)-based Parkinson’s disease (PD) biomarkers. Four different traits of the intestine have been proposed as PD biomarkers: intestinal α-synuclein, gastrointestinal (GI) symptoms severity, GM-related molecules, and GM composition. Among GM-related molecules, low levels of urine urolithin, decreased plasma trimethylamine N-oxide (TMAO), reduced plasma acetic and propionic acids and low levels of circulating LPS binding protein (LBP) are associated with PD. Concerning GM composition, while decreased relative abundance of Roseburia, Barnesiella, Blautia, Fusicatenibacter, Faecalibacterium and increased Akkermansia may serve as indicators of PD progression, dysbiosis data alone or associated with dietary intake information are useful for PD diagnosis. ↑: increase; ↓: decrease.
Figure 2. Gut microbiota (GM)-based Parkinson’s disease (PD) biomarkers. Four different traits of the intestine have been proposed as PD biomarkers: intestinal α-synuclein, gastrointestinal (GI) symptoms severity, GM-related molecules, and GM composition. Among GM-related molecules, low levels of urine urolithin, decreased plasma trimethylamine N-oxide (TMAO), reduced plasma acetic and propionic acids and low levels of circulating LPS binding protein (LBP) are associated with PD. Concerning GM composition, while decreased relative abundance of Roseburia, Barnesiella, Blautia, Fusicatenibacter, Faecalibacterium and increased Akkermansia may serve as indicators of PD progression, dysbiosis data alone or associated with dietary intake information are useful for PD diagnosis. ↑: increase; ↓: decrease.
Ijms 23 12289 g002
Table 1. The effects of probiotics treatment regarding in vitro and in vivo experimental studies.
Table 1. The effects of probiotics treatment regarding in vitro and in vivo experimental studies.
ProbioticExperimental ModelTreatment DurationTreatment EffectsReference
Lactobacillus salivarius, L. plantarum, L. acidophilus, L. rhamnosus, Bifidobacterium animalis subsp. lactis, B. breveIn vitro. PBMCs from 40 PD patients24 hIncrease of anti-inflammatory cytokines (IL-4, IL-10)
Decrease of pro-inflammatory cytokines (TNF-α, IL-6, IL-17A)
Reduction of ROS production
(Magistrelli et al., 2019) [328]
In vitro. Caco-2 cells2 hProtection of epithelium from gut permeability
In vitro. Escherichia coli and Klebsiella pneumoniae inoculation48 hInhibition of pathogen bacteria proliferation
Symprove (L. acidophilus, L. plantarum, L. rhamnosus, Enterococcus faecium)In vitro. Caco-2/THP1 cells24 h
Improvement in epithelial tight-junction integrity, and in wound healing
Increase of anti-inflammatory cytokines (IL-6, IL-10)
Decrease of pro-inflammatory chemokine IL-8
(Ghyselinck et al., 2021) [329]
In vitro. Stool samples from 3 PD patients48 hModulation of GM composition (↑Firmicutes, ↓Bacteroidetes)
Production of SCFAs and lactate
L. plantarum 200655In vitro. H2O2-treated SH-SY5Y cells4 hIncrease of BDNF and TH mRNA expression
Attenuation of apoptosis (↓apoptosis-related Bax/Bcl-2 ratio, ↓caspase-3 activity)
(Cheon et al., 2021) [330]
SLAB51 (Streptococcus thermophilus, B. longum, B. breve, B. infantis, L. acidophilus, L. plantarum, L. paracasei, L. delbrueckii subsp. bulgaricus, L. brevis)In vitro. 6-OHDA-treated SH-SY5Y cells
2 hReduction in dopaminergic neuronal loss, by increasing the neuronal survival BDNF pathway (mBDNF, p-TrkB, p-ERK5, p-CREB, PI3K/Akt) and decreasing neuronal death pathway (pro-BDNF, p-JNK, p-ERK1,2, P75)
Protection against OS (↓4-HNE).
(Castelli et al., 2020) [331]
In vivo. 6-OHDA-treated mice5 weeksImprovement in motor impairment.
Reduction in neuroinflammation (↓Iba1, ↓GFAP), and in OS, by restoring Nrf2/HO-1 activity and inhibiting NF-κB.
L. rhamnosus GG, B. animalis lactis, and L. acidophilusIn vivo. MPTP- and rotenone-induced mouse model4 weeksPrevention of dopaminergic neurons loss by upregulation of BDNF and GDNF, and inhibition of MAO-B expression
Amelioration of behavioural impairments.
Raise in butyrate levels.
(Srivastav et al., 2019) [325]
L. plantarum PS128In vivo. MPTP-induced mouse model28 daysAlleviation of neuroinflammation (↓TNF-α, ↓IL-1β, ↓IL-6) and OS (↑SOD, ↑GSH, ↑CAT, ↑GPx).
Improvement in motor deficits and dopaminergic neuronal cell death.
Reduction of glial reactivity.
Increase in BDNF expression and striatal dopamine level.
(Liao et al., 2020) [332]
B. bifidum, B. longum, L. rhamnosis, L. rhamnosus GG, L. plantarum LP28, and Lactococcus lactis subsp. LactisIn vivo. MitoPark mouse model16 weeksImprovement in motor impairment, balance function, and motor coordination.
Reduction in dopaminergic neuronal loss.
(Hsieh et al., 2020) [333]
Clostridum butyricumIn vivo. MPTP-induced mouse model4 weeksReduction in motor impairment, dopaminergic neuronal loss (↑TH) and synaptic dysfunction (↑synapsin I).
Inhibition of excessive microglia activation, via reversing GM dysbiosis.
Increase in GLP-1 and GLP-1 receptor levels in the brain.
(J. Sun et al., 2021) [334]
Lactobacillus plantarum DP189In vivo. MPTP-induced mouse model14 daysModulation of OS (↑SOD, ↑GSH-Px ↓MDA, ↓ROS), inflammation (↑IL-10, ↓TNF-α, ↓IL-6 and ↓IL-1β) and GM dysbiosis (↓Proteobacteria, ↓Actinobacteria, ↑Lactobacillus, ↑Prevotella).
Reduction in α-SYN accumulation in the substantia nigra.
Activation of Nrf2/ARE and PGC-1α pathways.
Suppression of NLRP3 inflammasome.
(L. Wang et al., 2022) [335]
Lactobacillus casei Shirota40 PD patients5 weeksImprovement in stool consistency.
Reduction of bloating and abdominal pain.
(Cassani et al., 2011) [336]
L. acidophilus and B. infantis40 PD patients12 weeksReduction of bloating and abdominal pain.(Georgescu et al., 2016) [337]
L. casei, L. fermentum, L. acidophilus, B. bifidum50 PD patients12 weeksReduction of IL-1, IL-8 and TNF-α gene expression.
Increase in TGF-β and PPAR-γ.
(Borzabadi et al., 2018) [338]
L. acidophilus, L. reuteri, L. fermentum, and Bifidobacterium bifidum60 PD patients12 weeksReduction of UPDRS motor scores.
Decrease of hs-CRP, MDA, insulin levels, and insulin resistance.
Enhancement in GSH levels and insulin sensitivity.
(Tamtaji et al., 2019) [339]
Xexbio (L. acidophilus, L. casei, L. lactis, B. infantis and B. longum)48 PD patients8 weeksImprovement of constipation and gut motility.(Ibrahim et al., 2020) [340]
L. acidophilus, L. reuteri, L. gasseri, L. rhamnosus, B. bifidum, B. longum, Enterococcus faecalis, E. faecium72 PD patients4 weeksImprovement of constipation, stool consistency and quality of life.(Tan et al., 2020) [341]
Probio-M8 (B. animalis subsp. lactis Probio M-8)82 PD patients12 weeksAmelioration in sleep quality, cognitive dysfunction, and defecation.
Attenuation of anxiety, depression, and gastrointestinal symptoms.
Modulation of gut microbiome, lipid metabolism, SCFAs and neurotransmitters serum levels.
(H. Sun et al., 2022) [342]
Abbreviations: 4-HNE: 4-hydroxynonenal; Bax: Bcl-2 associated X; Bcl-2: B-cell lymphoma 2; BDNF: brain-derived neurotrophic factor; CAT: catalase; GDNF: glial cell line-derived neurotrophic factor; GFAP: glial fibrillary acid protein; GLP-1: glucagon-like peptide 1; GM: gut microbiota; GPx: glutathione peroxidase; GSH: glutathione; GSH-Px: plasma glutathione peroxidase; hs-CRP: high-sensitivity C-reactive protein; Iba1: ionized calcium-binding adapter molecule 1; IL-1: interleukin 1; IL-10: interleukin 10; IL-17A: interleukin 17A; IL-1β: interleukin 1beta; IL-4: interleukin 4; IL-6: interleukin 6; IL-8: interleukin 8; MAO-B: monoamine oxidase-B; mBDNF: mature brain-derived neurotrophic factor; MDA: malondialdehyde; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3: NLR family pyrin domain containing 3; Nrf2/ARE: nuclear factor erythroid 2-related factor 2/antioxidant response element; Nrf2/HO-1: Nrf2/hemoxygenase-1; OS: oxidative stress; PBMCs: peripheral blood mononuclear cells; p-CREB: phosphorylated cAMP response element-binding protein; PD: Parkinson’s disease; p-ERK1,2: phosphorylated extracellular signal-regulated kinase; p-ERK5: phosphorylated extracellular-signal-regulated kinase 5; PGC-1α: peroxisome proliferator-activated receptor-gamma coactivator-1alpha; PI3K/Akt: phosphoinositide 3-kinase/phosphorylated protein kinase B; p-JNK: phosphorylated c-Jun N-terminal kinase; PPAR-γ: peroxisome proliferator-activated receptor gamma; pro-BDNF: pro-brain-derived neurotrophic factor; p-TrkB: phosphorylated tyrosine receptor kinase B; ROS: reactive oxygen species; SCFAs: short chain fatty acids; SOD: superoxide dismutase; TGF-β: transforming growth factor beta; TH: tyrosine hydroxylase; TNF-α: tumor necrosis factor-alpha; UPDRS: unified Parkinson’s Disease rating scale; α-SYN: alpha-synuclein; ↑: increase; ↓: decrease.
Table 2. The effects of prebiotics treatment regarding in vivo experimental studies.
Table 2. The effects of prebiotics treatment regarding in vivo experimental studies.
PrebioticExperimental ModelTreatment DurationTreatment EffectsReference
GOS, lcFOS, scFOS, nutrioseRotenone-induced mice model10 weeksImprovement of motor symptoms, gastrointestinal dysfunction, and inflammation (↓GFAP, ↓T-cells infiltration).
Restoration of DAT expression.
Reduction of α-synuclein levels.
(Perez-Pardo et al., 2017) [372]
Polymannuronic acidMPTP-induced model mice5 weeksAbolition of the apoptotic process (↓Bax, ↓Bax/Bcl-2 ratio).
Prevention of dopaminergic neuronal loss (↑TH gene and protein expression in the striatum).
Increase of faecal acetate, butyrate, and total SCFAs levels.
Inhibition of striatal inflammation (↓TNF-α mRNA levels).
(Liu et al., 2022) [373]
Prebiotic residual medium obtained from L. salivarius subsp. salicinium AP-32 culture medium6-OHDA-induced rat model8 weeksReduction of dopaminergic neuronal loss, motor dysfunctions, and muscle atrophy.
Increase of GPx and faecal SCFAs (propionate, butyrate).
Restoration of mitochondrial function and energy metabolism.
(Nurrahma et al., 2021) [376]
Prebiotic residual medium obtained from L. salivarius subsp. salicinium AP-32 culture medium6-OHDA-induced rat model8 weeksAmelioration of motor symptoms.
Reduction of inflammation (↓TNF-α) and OS (↓ROS, ↑SOD, ↑GPx).
Increase of SCFAs production (propionate, butyrate).
Modulation of GM composition (↑Ruminococcaceae, ↑Bifidobacterium, ↑Faecalibacterium, ↓Propionibacterium, ↓Clostridium, ↓Cylindriodes, ↓Ruminantium).
(Tsao et al., 2021) [346]
Dietetic fiber supplements (wheat, pectin, dimethylpolyoxyhexane-900)19 PD patients8 weeksImprovement in constipation and in motor function.
Increase of total plasma levodopa levels.
(Astarloa et al., 1992) [374]
Psyllium7 PD patients8 weeksIncrease in stool frequency and weight.(Ashraf et al., 1997) [375]
Resistant starch57 PD patients8 weeksImprovement of non-motor symptoms.
Reduction of calprotectin levels.
Increase in butyrate concentration.
(Becker et al., 2021) [316]
Abbreviations: 6-OHDA: 6-hydroxydopamine; Bax: Bcl-2 associated X; Bcl-2: B-cell lymphoma 2; DAT: dopamine transporter; GFAP: glial fibrillary acid protein; GM: gut microbiota; GOS: galactooligosaccharides; GPx: glutathione peroxidase; lcFOS: long-chain fructooligosaccharide; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; OS: oxidative stress; PD: Parkinson’s disease; ROS: reactive oxygen species; SCFAs: short chain fatty acids; scFOS: short-chain fructooligosaccharides; SOD: superoxide dismutase; TH: tyrosine hydroxylase; TNF-α: tumor necrosis factor-alpha; ↑: increase; ↓: decrease.
Table 3. The effects of dietary interventions in PD clinical trials.
Table 3. The effects of dietary interventions in PD clinical trials.
ReferenceType of StudyType of Dietary InterventionAimOutcomes
Metcalfe-Roach et al., 2021 [383]CrSMIND or MediMIND/Medi vs. PD onsetBoth diets delay PD onset; MIND slightly superior in the female subgroup.
Paknahad et al., 2020 [384]CTMediMedi vs. cognitive functionImprovement in executive function, language, attention, concentration, active memory and in the total score of cognitive assessment.
Rusch et al., 2021 [385]CTMediMedi vs. GI functionCorrelation with weight loss, improved constipation, and modified gut microbiota in PD patients.
Cassani et al., 2017 [386] MediMedi vs. PD progressionNo significant correlation.
Maraki et al., 2019 [387]CSMediMedi vs. PD onsetCorrelation with lower probability of prodromal PD in older people.
Zamzam Paknahad et al., 2022 [388]CTMediMedi vs. total antioxidant capacity (TAC) and PD severityImprovements in TAC and PD severity.
Alcalay et al., 2012 [389]CCSMediMedi vs. PD statusMedi adherence is associated with PD age at onset.
Strikwerda et al., 2021 [390]CSMedi, Dutch dietsMedi, Dutch diets vs. PD riskProtective effect.
Yin et al., 2021 [391]CSMediMedi vs. PD riskProtective effect.
Agarwal et al., 2018 [392]LSMINDMIND vs. PD development and progressionDecreased risk and slower progression of PD in older adults.
Lawrie et al., 2022 [393]CrSMINDMIND vs. PD severityDecreased fatigue and depression.
Koyuncu et al., 2021 [394] KDKD vs. PD patients voice qualityVHI * score improvement
VanItallie et al., 2005 [395]Feasibility studyKDKD vs. PD progressionUPDRS scores improvement.
Phillips et al., 2018 [396]CTKD vs a low-fat, high-carbohydrate dietKD vs. PD progressionMotor and nonmotor symptoms improvement.
Tidman et al., 2022 [397]CTKDKD vs. PD progressionUPDRS scores improvement.
Abbreviations: CCS: case-control study; CrS: cross-sectional study; CT: controlled trial; KD: ketogenic diet; LS: longitudinal study; Medi: Mediterranean diet; MIND: Mediterranean-DASH diet intervention for neurodegenerative delay; PUFAs: polyunsaturated fatty acids; UPDRS: Unified Parkinson’s Disease Rating Scale; VHI score: voice handicap index; * VHI is patient-rated scale developed to assess the level of disability experienced by patients affected by various voice disorders.
Table 4. Preclinical and clinical studies on the use of fecal microbiota transplantation for Parkinson’s disease.
Table 4. Preclinical and clinical studies on the use of fecal microbiota transplantation for Parkinson’s disease.
Ref.Study CohortStudy GroupsDonorRecipientExperimental ProcedureResultsAdverse Events
Zhao et al., 2021 [470]MiceControls (n = 15), rotenone (n = 15) and rotenone + FMT (n = 15)Control miceRotenone-induced PD miceOral gavage (100 μL bacterial suspension) daily for 2 weeks↓ Dysbiosis
↓ Motor symptoms
↑ Intestinal barrier and BBB integrity
↓ Systemic inflammation
↓ Neuroinflammation (SN)
↓ LPS (serum, colon and SN)
↓ TLR4/NF-κB pathway (colon and SN)
N.A.
Sun et al., 2018 [469]MiceControls (n = 15), MPTP + PBS (n = 15) and MPTP + FMT (n = 15)Control miceMPTP-induced PD miceGavage (200 μL bacterial suspension containing 108 CFU/mL) daily for 7 days↓ Dysbiosis
↓ Fecal SCFAs
↓ Motor symptoms
↑ DA and 5-HT (striatum)
↓ Microglia and astrocyte activation (SN)
↓ TLR4/TNF-α pathway (gut and brain)
N.A.
Zhong et al., 2021 [471]MiceControls + PBS (n = 10), controls + FMT (n = 10), MPTP + PBS (n = 10), MPTP + FMT (n = 10)Control miceControls or MPTP-induced PD miceGavage (200 μL bacterial suspension containing 108 CFU/mL) daily for 7 days↓ Motor symptoms
↓ Fecal SCFAs
↓ α-syn (SN)
↓ Microglia activation (SN)
↓ TLR4/NF-κB pathway (striatum and SN)
N.A.
Zhang et al., 2021 [472]MiceControls (n = 3), MPTP (n = 3) and MPTP + FMT (n = 3)Control miceMPTP-induced PD miceTransplantation with 200 μL bacterial suspension (containing 108 CFU/mL) daily for 2 weeks↓ Neuroinflammation (SN)
↓ Motor symptoms
Blautia
Anaerostipes, ASF356, Ruminococcus and Bifidobacterium
↓ Microglia and astrocyte activation (SN)
↓ IL-1β, iNOS, GSK3β and p-PTEN (SN)
N.A.
Zhou et al., 2019 [473]MiceMice pre-treated with MPTP and antibiotics, divided in PD-PBS (n = 8), PD-NA (n = 8), PD-NF (n = 8) and PD-NF/HK (n = 8)Control mice or control mice undergoing FMTMPTP-induced PD mice pre-treated with antibioticsGastric gavage (200 μL bacterial suspension containing 108 CFU/mL) daily for 7 days↑ DA and 5-HT (striatum) in PD-NF mice
↑ Neuroprotection in PD-NF mice
N.A.
Sampson et al., 2016 [40]MiceGF + FMT from SPF control mice, GF + FMT from PD patients, GF + FMT from healthy patientsHuman PD patients (n = 6), human healthy controls (n = 6) or SPF control mice (n = 3)α-syn-overexpressing miceOral gavageIn GF + PD-FMT mice:
↑ Physical impairment
Proteus, Bilophila and Roseburia
Lachnospiraceae, Rikenellaceae, Peptostreptococcaceae and Butyricicoccus
↓ Acetate
↑ Proprionate and butyrate
N.A.
Huang et al., 2019 [474]Human (case report)PD patient presenting tremor for 7 years and constipation (>3 years)26 y.o. healthy male71 y.o. male PD patientsColonoscopy (200 mL of fecal microbiota suspension) daily for 3 days↓ Tremor (no tremor for 2 months)
↓ Constipation
↑ α-diversity
↓ UPDRS score 1 week after FMT
No
Kuai et al., 2021 [475]Humans (prospective single study)PD patientsFrozen fecal microbiota from the China fmtBankPD patients (n = 11)Intra-intestine transplantation of 40–50 mL of frozen fecal microbiota resuspended in 200 mL saline solutionBlautia and Prevotella
Bacteroidetes
↓ H-Y, UPDRS and NMSS scores
↓ Wexner constipation and PAC-QOL scores
No
Xue et al., 2020 [476]HumansPD patients + FMT (via colonoscopy, n = 10; nasointestinally, n = 5)5 Healthy donors (mean 22 y.o., 3 males and 2 females)PD patientsColonoscopy or nasointestinal administration↓ PSQI, HAMA, PDQ-39, HAMD, UPDRS-III and NMSQ5 cases: diarrhea (n = 2), abdominal pain (n = 2) and flatulence (n = 1)
Segal et al., 2021 [477]Humans (uncontrolled case series)6 PD patients with symptoms for 5 years (mean).2 healthy donors (males, 38 and 50 y.o.)6 PD patients with constipation (mean 52 y.o.), 3 males and 3 femalesColonoscopy (300 mL of fecal suspension)↓ Motor and non-motor symptoms
↓ Constipation
1 case requiring hospitalization for observation
Abbreviations: α-syn: α-synuclein; BBB: blood-brain barrier; CFU: colony forming units; DA: dopamine; FMD: fasting mimicking diet; FMT: fecal microbiota transplantation; GF: germ-free;GSK3β: glycogen synthase kinase-3 beta; H-Y: Hoehn and Yahr scale; HAMA: Hamilton anxiety scale; HAMD: Hamilton depression rating scale; 5-HT: serotonin;IL-1β: interleukin 1 beta; iNOS: inducible nitric oxide synthase; LPS: lipopolysaccharide; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; NMSQ: non-motor symptoms questionnaire; NMSS: non-motor symptoms scale; p-PTEN: phosphorylated PTEN; PAC-QOL: patient assessment of constipation quality of life questionnaire; PBS: phosphate buffered saline; PD-NA: MPTP-induced PD mice treated receiving FMT from control mice; PD-NF: MPTP-induced PD mice treated receiving FMT from control mice undergoing FMD; PD-NF/HK: MPTP-induced PD mice treated receiving heat inactivated FMT from control mice undergoing FMD; PD-PBS: MPTP-induced PD mice treated receiving PBS; PD: Parkinson’s disease; PDQ-39: Parkinson’s Disease Questionnaire; PSQI: Pittsburgh sleep quality index; SCFAs: short chain fatty acids; SN: substantia nigra; SPF: specific pathogen free; TLR4: toll like receptor 4; TNF-α: tumor necrosis factor α; UPDRS: unified Parkinson’s disease rating scale; y.o.: years old; ↓: decrease; ↑: increase.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Varesi, A.; Campagnoli, L.I.M.; Fahmideh, F.; Pierella, E.; Romeo, M.; Ricevuti, G.; Nicoletta, M.; Chirumbolo, S.; Pascale, A. The Interplay between Gut Microbiota and Parkinson’s Disease: Implications on Diagnosis and Treatment. Int. J. Mol. Sci. 2022, 23, 12289. https://doi.org/10.3390/ijms232012289

AMA Style

Varesi A, Campagnoli LIM, Fahmideh F, Pierella E, Romeo M, Ricevuti G, Nicoletta M, Chirumbolo S, Pascale A. The Interplay between Gut Microbiota and Parkinson’s Disease: Implications on Diagnosis and Treatment. International Journal of Molecular Sciences. 2022; 23(20):12289. https://doi.org/10.3390/ijms232012289

Chicago/Turabian Style

Varesi, Angelica, Lucrezia Irene Maria Campagnoli, Foroogh Fahmideh, Elisa Pierella, Marcello Romeo, Giovanni Ricevuti, Marchesi Nicoletta, Salvatore Chirumbolo, and Alessia Pascale. 2022. "The Interplay between Gut Microbiota and Parkinson’s Disease: Implications on Diagnosis and Treatment" International Journal of Molecular Sciences 23, no. 20: 12289. https://doi.org/10.3390/ijms232012289

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