Next Article in Journal
Omega-3 Fatty Acid Supplementation on Post-Exercise Inflammation, Muscle Damage, Oxidative Response, and Sports Performance in Physically Healthy Adults—A Systematic Review of Randomized Controlled Trials
Previous Article in Journal
Food Resource Management and Healthy Eating Focus Associates with Diet Quality and Health Behaviors in Low-Income Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut–Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson’s Disease

by
Elizabeth Riegelman
,
Kathy S. Xue
,
Jia-Sheng Wang
and
Lili Tang
*
Department of Environmental Health Science, University of Georgia, Athens, GA 30602, USA
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(13), 2041; https://doi.org/10.3390/nu16132041
Submission received: 11 June 2024 / Revised: 20 June 2024 / Accepted: 25 June 2024 / Published: 27 June 2024
(This article belongs to the Section Prebiotics and Probiotics)

Abstract

:
With the recognition of the importance of the gut–brain axis in Parkinson’s disease (PD) etiology, there is increased interest in developing therapeutic strategies that target α-synuclein, the hallmark abhorrent protein of PD pathogenesis, which may originate in the gut. Research has demonstrated that inhibiting the aggregation, oligomerization, and fibrillation of α-synuclein are key strategies for disease modification. Polyphenols, which are rich in fruits and vegetables, are drawing attention for their potential role in this context. In this paper, we reviewed how polyphenols influence the composition and functional capabilities of the gut microbiota and how the resulting microbial metabolites of polyphenols may potentially enhance the modulation of α-synuclein aggregation. Understanding the interaction between polyphenols and gut microbiota and identifying which specific microbes may enhance the efficacy of polyphenols is crucial for developing therapeutic strategies and precision nutrition based on the microbiome.

1. Introduction

Parkinson’s disease (PD) is the second most common neurodegenerative disease, after Alzheimer’s, affecting approximately 2–3% of the population more than 60 years of age and up to 5% of the population over 85 years of age [1,2]. It is estimated that there are 1.5 million people in the United States living with PD, and 60,000 Americans are diagnosed with this disease annually [3]. PD is a progressive neurological disorder that primarily affects motor function, leading to symptoms such as tremors, stiffness, slowness of movement, and impaired balance and coordination [4,5]. One of the hallmark pathologies of PD is the accumulation of alpha-synuclein (α-synuclein), a presynaptic neuronal protein that aggregates into insoluble fibrils, forming Lewy bodies (LBs) and Lewy neurites in the brain [6]. Although the exact function of α-synuclein remains partially understood, it has been linked to a range of neuronal functions, including neurotransmitter release, the modulation of a variety of enzymes and transporters, the dynamics of presynaptic vesicles, and the support of neuronal plasticity [7]. Its abnormal accumulation contributes to neuronal death and the subsequent manifestation of PD symptoms [8].
The pathogenesis of PD has not yet been fully elucidated. While the majority of earlier PD studies focused solely on brain pathologies, the gastrointestinal (GI) system is now recognized as a pivotal participant in the pathogenesis of PD [9,10,11,12,13]. Recent research has increasingly focused on the gut–brain axis, a complex bidirectional communication system that links the enteric nervous system (ENS) of the GI tract with the central nervous system (CNS). This axis not only plays a critical role in maintaining GI homeostasis but also appears to be involved in the pathogenesis of several neurological disorders, including PD. The emerging evidence suggests that α-synuclein pathology may originate in the gut and then spread to the brain via the vagus nerve, a concept supported by observations of GI abnormalities in PD patients years before the onset of motor symptoms. Moreover, alterations in the gut microbiota composition have been observed in PD patients, indicating a potential role of gut dysbiosis in the disease’s progression.
Currently, there is no existing cure for PD, and drugs that are presently used in clinics only provide symptomatic treatment rather than preventing or slowing the pathogenic progression of neurodegeneration [14]. In addition, some of these drugs present many side effects in patients [15]. Therefore, there is an urgent need to develop novel therapeutic agents with lower side effects and a broader spectrum of targets to not only treat the symptoms but also potentially prevent or slow the pathogenic progression of PD. Given the interaction between the gut microbiota and the CNS, dietary interventions that modulate the gut microbiome have garnered interest as a potential therapeutic strategy for PD. Polyphenols, a diverse group of phytochemicals found in fruits, vegetables, tea, wine, and certain herbs, have garnered attention in this context. These compounds are known for their antioxidant and anti-inflammatory properties and have been shown to influence the composition and function of the gut microbiota. Importantly, polyphenols may also inhibit the aggregation of α-synuclein, offering a dual mechanism by which they could mitigate PD pathology. By modulating the gut microbial community, dietary polyphenols may reduce intestinal inflammation, enhance gut barrier function, and produce metabolites that could potentially inhibit the aggregation of α-synuclein. Therefore, understanding the mechanisms underlying the interactions between dietary polyphenols, the gut–brain axis, and α-synuclein pathology could open new avenues for preventing or slowing the progression of PD, highlighting the significance of diet and gut microbiota in the pathogenesis of neurodegenerative diseases. This review aims to integrate the current knowledge and recent discoveries to shed light on the complex interactions between dietary polyphenols, gut microbiota, and α-synuclein aggregation, offering insights into potential novel therapeutic strategies for PD that leverage the gut–brain axis.

2. α-Synuclein: A Key Player in Parkinson’s Disease Pathology

α-synuclein is a protein comprising 140 amino acids encoded by the SNCA gene, which normally exists in naturally occurring monomers. Under pathological conditions, natively unfolded monomers can undergo self-aggregation, forming pathological oligomers that further mature into fibrils [16]. The accumulation of α-synuclein aggregates (particularly into oligomers and fibrils) disrupts normal cellular processes, causes mitochondrial dysfunction, triggers inflammation, and leads to neuronal death. Therefore, treatments that target α-synuclein oligomers and/or fibrils may reduce neurodegeneration, which is a promising therapeutic strategy for PD [17].

2.1. α-Synuclein Structure and Physiological Function

α-synuclein is a relatively small protein within the synuclein family, which also includes β-synuclein and γ-synuclein. With a molecular weight of approximately 14 kDa, α-synuclein is predominantly located at the presynaptic terminals in the CNS and accounts for about 1% of all cytosolic proteins in the brain [7,18].
A schematic figure about the structure of α-synuclein is shown in Figure 1. The protein can be divided into three distinct domains: The N-terminal amphipathic domain, which contains the evolutionary conserved KTEGV motifs and the main mutations associated with familial PD, such as A53T and A30p [19]. This region is responsible for membrane binding as well. Additionally, there is a central hydrophobic segment known as the non-amyloid component (NAC), which confers the potential for a β-pleated sheet, and a highly negatively charged carboxyl tail at the C-terminal end, which contains a Ca2+-binding site and the main phosphorylation site at Ser129, modulating α-synuclein aggregation. The hydrophobic NAC region, in particular, is critical for the protein’s propensity to adopt β-sheet configurations that can self-assemble into fibrillar aggregates, forming the core of LBs, a pathological hallmark found in PD brains [16]. Once α-synuclein fibrils are established in a neuron, they can act as a guide for the aggregation of endogenous α-synuclein protein [20], further initiating PD progression.
Although the main function of α-synuclein in both the central and peripheral nervous systems remains unclear, it has been linked to a range of neuronal functions, including neurotransmitter release, the modulation of a variety of enzymes and transporters, the dynamics of presynaptic vesicles, and the support of neuronal plasticity [7]. Additionally, α-synuclein is involved in the formation of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex at presynaptic nerve terminals. This latter function is thought to be mediated by a chaperone activity that has not yet been identified, highlighting α-synuclein’s potential role in ensuring efficient neurotransmission and synaptic function [16,21].

2.2. Role of α-Synuclein in PD

α-synuclein plays a significant role in the pathogenesis of both familial and sporadic forms of PD [17], as well as in other synucleinopathies [22]. Under normal conditions, native α-synuclein exists in a dynamic equilibrium between unfolded monomers and α-helically folded tetramers with a low propensity for aggregation [23]. The aggregation process of α-synuclein involves a conformational change whereby it adopts a β-sheet-rich structure that facilitates its aggregation into oligomers, protofibrils, and insoluble fibrils that finally accumulate in Lewy bodies within neurons [24]. Almost all pathologically aggregated α-synuclein is phosphorylated at the Ser129 site [25]. These aggregates disrupt various cellular processes, leading to neuronal dysfunction and, ultimately, cell death [26,27] (Figure 2).
One of the critical ways that misfolded α-synuclein contributes to neuronal dysfunction is through the disruption of mitochondrial function [28,29,30]. Mitochondria are essential for energy production, and their dysfunction leads to reduced cellular energy levels, increased oxidative stress, and the activation of apoptotic pathways [31,32]. Misfolded α-synuclein aggregates can impair mitochondrial dynamics, including fission and fusion processes, and interfere with mitochondrial transport along axons, further exacerbating neuronal dysfunction [33].
Another significant impact of misfolded α-synuclein is on synaptic function [34]. Synapses are critical for neuronal communication; therefore, any disruption in their function can lead to significant impairments in neuronal signaling [35]. Misfolded α-synuclein can impair synaptic vesicle release and reuptake, leading to reduced neurotransmitter availability and synaptic transmission efficiency. This disruption contributes to the motor and cognitive symptoms observed in PD and related disorders [36,37].

2.3. α-Synuclein Propagation

Studies have suggested that α-synuclein can be transmitted between neurons [38] and can seed the formation of toxic aggregates in recipient neurons in a prion-like manner [39]. α-synuclein demonstrates several prion-like characteristics, notably the capacity of its aggregated forms to self-propagate by promoting the aggregation of normal α-synuclein, a process known as ‘seeding’. This mechanism can increase the aggregate burden within a cell. Similar to prions, misfolded α-synuclein can act as a template, inducing the misfolding of native α-synuclein in neighboring neurons. This templated misfolding can lead to the cell-to-cell transmission of pathological α-synuclein aggregates, contributing to the progressive nature of PD. For example, fetal dopamine cells transplanted into the striatum of patients with PD were found to develop Lewy pathology when examined neuropathologically 1–2 decades later [40]. Similar to the human transplants, α-synuclein has been observed in cell culture and rodent transplantation experiments, where it was transferred from one cell to another [38,41,42].

3. Gut–Brain Axis: A New Frontier in Parkinson’s Disease Research

The gut–brain axis represents a complex communication network linking the GI tract and the CNS, fundamentally impacting health and disease [43]. Research has highlighted that gut microbiota might play a significant role in the onset and progression of PD [44]. Various studies indicate that alterations in the gut microbiome have been observed in PD patients, with changes in the abundance of certain bacterial species compared to healthy controls. Additionally, α-synuclein also accumulates in the ENS [45,46,47]. This accumulation can occur years before the typical motor symptoms of PD appear, suggesting that the gut might be an early site of disease pathology. Understanding the gut–brain axis in PD could open new avenues for early diagnosis and targeted treatments that modulate the gut microbiota or its metabolic outputs, potentially slowing disease progression or alleviating symptoms.

3.1. Gut–Brain Axis

The concept of the gut–brain axis describes bidirectional communication between the CNS and ENS of the GI tract, which is linked by neurons of the sympathetic and parasympathetic nervous system [48,49]. Through this bidirectional communication network, signals from the brain can influence the motor, sensory, and secretory modalities of the GI tract and, conversely, visceral messages from the GI tract can influence brain function [50]. At present, it is suggested that gut microbes may serve as significant contributors to the bidirectional communication that takes place along the gut–brain axis. The microbiome community carries out important metabolic and physiological functions for the host and contributes to overall health and homeostasis [51,52,53]. Consequently, the gut microbiota has emerged as a potential diagnostic and therapeutic target in disorders as diverse as PD [54,55,56,57].

3.2. Dysbiosis of Microbiome in PD

Dysbiosis of the gut microbiome in PD patients is currently being investigated to determine which microbiota actively produce metabolites that are implemented in microglia activation, α-synuclein aggregation, and inflammation in the gut. Gut microbes play a crucial role in the gut–brain axis, which interacts with the ENS, enterocytes, and immune system to influence host health [58]. Evidence suggests that PD pathogenesis may be influenced by the interplay between the imbalance of gut microbes and altered bacterial metabolites [59,60,61]. Numerous findings in both observational PD patient studies and experimental animal studies have revealed that gut bacteria aid in the regulation of anti-inflammatory and pro-inflammatory profiles, suggesting that alterations within an individual’s gut microbiome can influence the risk of developing PD [44,57,62,63,64,65,66,67,68,69,70]. Newly diagnosed PD patients often exhibit intestinal hyperpermeability, endotoxemia, and microbial dysbiosis, which bolsters the hypothesis that gut-derived inflammation promotes neuroinflammation and neurodegenerative changes in PD pathogenesis [65,68,70,71]. Clinical studies have shown an increase in intestinal permeability in PD, where bacterial endotoxins in the form of lipopolysaccharides are associated with increased α-synuclein accumulation within the GI tract [72,73]. Some PD patients have shown elevated lipopolysaccharide serum levels, which could correlate to increased intestinal permeability [74,75,76]. Further, the guts of PD patients are often colonized by lipopolysaccharide-producing bacteria such as Helicobacter pylori, which induces chronic inflammation and degradation of the gut mucosal lining [9,75,77,78].
Individuals with high-risk factors for PD or the diagnosis of PD have been documented to have significantly different compositions of gut microbes compared to those of healthy controls [13,79,80,81]. The relative abundances of anti-inflammatory bacteria such as Blautia, Coprococcus, Roseburia, Fusicatenibacter, Faecalibacterium, and Lachnospira are reduced in PD patients, while, in contrast, Lactobacillus, Bifidobacterium, and Akkermansia phyla are higher in PD patients [65,76,82,83,84,85,86]. Numerous studies have documented the alterations in the composition of the gut microbiome in PD, as seen in Table 1 [44,68,82,83,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106].
Lower levels of fecal SCFAs have been observed in PD patients compared to healthy controls [82,87,107,108], and the evidence suggests that SCFA-producing bacteria may modify the microbiome’s genetic potential to produce the enzymes needed for SCFA formation. A recent meta-analysis determined that PD patients had reduced levels of bacteria in the Lachnospiraceae and Ruminococcaceae families [65]. Similarly, another meta-analysis reported that PD patients had decreased levels of the genera Roseburia and Faecalibacterium, both belonging to the Firmicutes Phyla [83]. These bacteria are known SCFA producers, and reductions in their levels have been documented in other diseases with an inflammatory component. It is important to note, however, that not all studies have obtained the same results. Although it is not clear whether alterations in the microbiome are the origin of PD pathogenesis, it is evident that disease pathology influences the microbiota over time and vice versa.

3.3. Gut–Brain Axis and α-Synuclein

α-synuclein is a protein primarily found in the brain; however, recent evidence suggests that it also occurs in other peripheral tissues such as the GI tract [109,110,111]. Biopsies of GI tissues from PD patients have shown α-synuclein accumulation in the lower parts of the esophagus, stomach, duodenum, colon, and rectum [112,113,114]. Finding α-synuclein outside the CNS supports the hypothesis that the presence of α-synuclein in both the brain and the gut may result from a common pathological aggregation pathway involving the vagus nerve. This evidence indicates that the initial process of α-synuclein misfolding and pathology might start in the GI tract before spreading to the brain [115,116]. This observation supports Braak’s hypothesis, which proposes that α-synuclein aggregates originate in the gut and then propagate through the gut–brain axis, triggering the onset of PD [117,118]. Further, the retrograde spread of α-synuclein towards the brain suggests that GI-derived α-synuclein could ascend the ENS and propagate through interconnected neurons toward the midbrain of the CNS via the vagus nerve, ultimately reaching the Substantia nigra pars compacta (SNpc) and forming fibril structures that cause motor dysfunction [47,116,117,119,120].
Research suggests that certain microbes may influence the misfolding and abnormal formation of α-synuclein through extracellular mechanisms [121,122,123,124]; an example of this may be Helicobacter pylori, a gram-negative spiral bacterium known as the causative agent of gastric ulcers, present in 50% of the world population [125]. H. pylori contains cholesterol-α-glucosyltransferase, which catalyzes the conversion of membrane cholesterol to cholesteryl glucosides. The glycosylated derivatives can exert a neurotoxic effect on dopamine neurons and promote the aggregation of α-synuclein in the vagus nerve from the level of the stomach [126,127]. Another gram-negative bacteria species, i.e., Escherichia coli, has been documented to be implicated in α-synuclein misfolding [128]. E. coli produces an amyloid fiber protein called curli, whose purpose is to promote cell community behavior through the formation of biofilms in the extracellular matrix. The administration of E. coli in animal models has demonstrated an increase in α-synuclein fibril reactivity and accumulation of insoluble α-synuclein in the substantia nigra portion of the midbrain compared to controls, indicating that exposure to microbial amyloids in the GI may accelerate α-synuclein aggregation in the gut and brain [129,130].

3.3.1. Braak’s Hypothesis

Braak’s hypothesis is a significant theory in the field of neurology, particularly concerning the pathology of PD. Proposed by Braak and his colleagues in the early 2000s, this hypothesis suggests that PD may start outside the brain, specifically in the gut or the nasal cavity, and then spread to the brain via the nervous system. According to Braak’s hypothesis, an unknown pathogen (either a virus or bacterium) in the gut could trigger the onset of sporadic PD [117]. Along with this hypothesis, they presented a staging system for PD that is based on a specific pattern of the spread of α-synuclein [131]. Subsequent to these publications, the broader dual-hit hypothesis was proposed, which posits that sporadic PD begins simultaneously in two locations: the neurons of the nasal cavity and the neurons in the gut [127,132]. From these places, the pathology is hypothesized to spread according to a specific pattern, i.e., via the olfactory tract and the vagus nerve, respectively, toward and within the CNS. There is experimental and clinical evidence supporting Braak’s hypothesis [133,134,135,136,137,138,139,140,141]. Additionally, α-synuclein aggregations have been found in the GI tract of animal models of early and advanced PD [142,143,144,145].
Braak’s hypothesis has had a substantial impact on research directions, focusing attention on the potential role of the gut–brain axis and the olfactory system in the early detection and understanding of PD [146,147,148]. It also supports the observation that non-motor symptoms, such as the loss of smell or GI issues, can precede the motor symptoms by several years.

3.3.2. α-Synuclein from the Gut to the Brain

The transmission of α-synuclein from the ENS to the CNS is a critical research area for understanding the pathogenesis of neurodegenerative diseases, particularly PD. This area has gained significant attention as it may explain the initiation and progression of PD [12,149,150].
Direct evidence of gut–brain α-synuclein transmission in rodents was provided by Holmqvist et al. [47] who demonstrated that α-synuclein fibrils derived from PD patients could migrate from the GI tract to the brain via the vagus nerve in rats. Challis et al. [151] complemented these findings by inoculating the duodenal walls of aged mice with α-synuclein pre-formed fibrils (PFFs) and observed the progression of α-synuclein pathology to the brain. Similarly, Kim et al. provided additional evidence by injecting mouse α-synuclein PFFs into the pylorus and duodenum, noting the subsequent detection of phosphorylated α-synuclein in the CNS, beginning in the dorsal motor nucleus of the vagus nerve and progressing to regions like the locus coeruleus, amygdala, substantia nigra, and prefrontal cortex [119]. This progression closely reflects the Braak staging scheme for PD. Crucially, the study showed that these effects were negated if α-synuclein PFFs were injected into the intestinal wall following truncal vagotomy, which involves the severing of the vagus nerve. In this experimental setup, phosphorylated α-synuclein was still detectable in the upper duodenum 7 months after injection, but there was no spread to the substantia nigra [119]. These findings robustly support the notion that gut-derived α-synuclein is capable of propagating through the vagus nerve in a prion-like manner to induce CNS disease.

4. Polyphenols: Beyond Antioxidants

4.1. Types and Dietary Sources

Polyphenols are highly specific secondary metabolites of plants that have a variety of biological roles, from resistance to infection by pathogenic microorganisms to defense against ultraviolet radiation [152]. Generally speaking, “polyphenol” refers to compounds derived entirely from the shikimate/phenylpropanoid pathway and/or the polyketide pathway, characterized by the presence of more than one phenolic unit and devoid of nitrogen-based functions [153]. To date, thousands of polyphenols have been identified from natural dietary plants. In general, polyphenols are mainly classified as phenolic acids, flavonoids, lignans, coumarins, and stilbenes, as outlined in Figure 3 [154]. The presence of polyphenol compounds in food greatly depends on environmental factors and food processing, production, and storage [152].

4.1.1. Phenolic Acids

Phenolic acids are phytochemicals found in the majority of plant tissues and comprise a phenolic ring with an attached organic carboxylic acid. The secondary plant metabolites are aromatic acids that can be further classified into two broad groups, including hydroxycinnamic acids (HCAs) and hydroxybenzoic acids (HBAs), the latter of which are less common. HCAs are most concentrated in the outer ripened parts of fruits and are present in a variety of fruits, vegetables, and seeds [155,156]. The most abundant HCAs include the free forms of para-coumaric acid, caffeine, ferulic acid, and sinapic acid, whereas the bound forms include glycosylated derivatives of quinic acid, shikimic acid, and tartaric acid [157]. The most common HBAs include gallic acid, ellagic acid, syringic acid, and salicylic acid [158]. A wide variety of fruits and vegetables contain phenolic acids, including apples that contain chlorogenic acid and blueberries that contain para-coumaric, caffeic, and ferulic acids [159]. Phenolic acids are potent antioxidants due to their high potential to serve as hydrogen donors and oxygen quenchers and reduce metal-chelating agents [156,160,161]. Furthermore, HCAs and HBAs have been found to exhibit protective effects on neural, cardiac, and hepatic tissues, making this group of polyphenols an interesting target for research in the field of therapeutic medicine.

4.1.2. Coumarins

Coumarins belong to the benzopyrone family and comprise a six-membered benzene ring attached to an α-pyrone ring [162]. Coumarins are present in several plants but are highest in concentration in the tonka bean, vanilla grass, sweet clover, and cinnamon [163]. These small molecule candidates have been considered for drug development due to their high solubility, low molecular weight, low toxicity, and high bioavailability [163,164]. Coumarins act as secondary plant metabolites to protect against herbivores and microorganisms through interactions with plant growth hormones and respiration; however, regarding biochemistry and physiology, coumarins are known to exhibit antioxidant effects and act as enzyme inhibitors [165]. Coumarins can be further classified into four groups: simple coumarins, furanocoumarins, pyranocoumarins, and pyrone-substituted coumarins, whereas both the free and glycosidic forms of coumarin can be found in plants [166]. While coumarins pose as promising therapeutic agents for human health, there are certain derivatives of coumarins, such as coumarin chromen-2-one, which are potentially poisonous for human consumption [167]. Still, the majority of coumarins have been recognized as harmless compounds and are considered to be candidates for medicinal drugs with diverse pharmacological activities, high bioavailability, and generally low toxicity [165].

4.1.3. Stilbenes

Stilbenes accumulate in the vine tissues of plants under stressful biotic and abiotic conditions [168]. Over 400 natural stilbenes have been described [169], and their potential as antioxidants and chemopreventive agents is of interest to the scientific community. A major stilbene of interest, namely, resveratrol, has been researched for its potential cardioprotective effects. Resveratrol has been identified in at least 185 different plant species and is associated with antioxidant activity and its positive effect on lifespan and age-related diseases [170,171]. The regular consumption of stilbenes has been demonstrated to alleviate intracellular oxidative stress, reduce chronic inflammation, and suppress adipogenesis and lipogenesis [172].

4.1.4. Lignans

Lignans consist of two phenylpropanoid units coupled by a β-β′-bond and are found in various seeds, grains, vegetables, and fruits [173]. Lignan bioavailability depends heavily on the diet due to its relatively low concentrations [174]. As a polyphenol class, lignans have received growing attention given their potentially beneficial bioactive properties due to their steroid-analogous chemical structure. Technically considered phytoestrogens, lignans have been documented to exhibit anti-estrogenic, antioxidant, and anti-carcinogenic effects. The lignans most common in the human diet include lariciresinol, matairesinol, pinoresinol, and secoisolariciresional which are most concentrated in sesame and flax seeds [173,175,176]. While the database on the content of lignans in foods is growing, more evidence needs to be added on the number of lignan dietary sources and the physiological role that the diverse compounds play in vivo.

4.1.5. Flavonoids

Flavonoids are the largest class of dietary polyphenols and, hence, contribute greatly to our understanding of the broad spectrum of their health-promoting effects. There are over 6000 flavonoids that contribute to the pigments and protection of different fruits, vegetables, and medicinal plants [152]. All flavonoids contain at least 15 carbons with two benzene A and B rings and are further classified based on variations within the heterocyclic C ring [158]. The main groups of flavonoids are flavones, flavonols, catechins, isoflavones, flavanones, and anthocyanins. A broad spectrum of health-promoting effects has been attributed to the flavonoid group largely due to their ability to modulate key cellular enzyme functions and act as antioxidant, anti-inflammatory, anti-mutagenic, and anti-carcinogenic compounds [177]. Research has shown that flavones and catechins have the highest biological significance as antioxidants for protection against reactive oxygen species (ROS). Further, flavonoids are capable of inhibiting enzymes such as phosphodiesterase, lipoxygenase, Ca2+ ATPase, and COX, which are implicated in neurodegenerative diseases [177]. Researchers have also recognized flavonoids for their antimicrobial [178,179], antifungal [180], and antiviral [181,182] activities in vivo.

4.2. Gut Microbiota and Polyphenols

Dietary polyphenols can interfere directly with the enhancement or impairment of nutrient absorption. Given the antioxidant capabilities of many polyphenols, macro- and micronutrient oxidation may be prevented during metabolism, which could protect the quality of the nutrients. Carbohydrate absorption and glycemia may be decreased by polyphenols through their amylase-antagonizing activities [183,184], providing carbohydrates for bacteria such as Bacteroides, Bifidobacterium, Clostridium, Eubacterium, Lactobacillus, and Ruminococcus [185]. In this sense, polyphenols can influence bacterial diversity, gut peptide synthesis, energy and nutrient absorption, and insulin sensitivity [158]. However, polyphenol’s antioxidant capabilities may be a double-edged sword as polyphenols can interfere with mineral absorption. Gallic acid, chlorogenic acid, and polyphenolic polymerization products have been demonstrated to inhibit iron absorption [186,187,188,189]. Tannins and gallic acid have been documented to have an affinity for binding to zinc, further impairing zinc absorption [190,191]. Regardless of the caveats that accompany dietary polyphenols, research has elucidated the beneficial potentials and relationships between the gut microbiome and commonly consumed polyphenol substances.

4.2.1. Curcumin

Curcumin, commonly known as turmeric, is a popular HCA that is known for its variety of pharmacological and restorative effects, including therapeutic approaches for chronic diseases, cardiovascular disease, depression, skin diseases, obesity, diabetes, and multiple types of cancer [192,193,194,195]. Studies that have observed the pharmacokinetics and bioavailability of curcumin have documented how poorly absorbed and rapidly metabolized the compound is, resulting in speculation about its clinical use [196]. Nevertheless, peripheral influences of curcumin may play a more important role in the CNS [197]. Curcumin may prevent the formation of ROS and glial cell activation, further hindering α-synuclein aggregation and neuronal cell apoptosis [197,198,199,200]. It has been proposed that curcumin can modulate gut signaling pathways and potentially exert effects on the microbiome gut–brain axis. Oral administration of curcumin has been demonstrated to promote beneficial gut bacteria growth, including Bifidobacteria and Lactobacilli strains, while reducing the abundance of pathogenic bacteria strains, including Prevotellaceae, Coriobacterales, Enterobacteria, and Rikenellaceae [201,202,203]. The primary metabolite of curcumin, i.e., tetrahydrocurcumin, has been documented to restore gut microbiome dysbiosis by lowering the relative abundance of Actinobacteria and Proteobacteria and by modifying the ratio of Firmicutes to Bacteroidetes [204]. An in vivo study showed that curcumin intervention improved motor deficits, glial cell activation, and the aggregation of α-synuclein in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) PD mouse model. The microbiome data showed elevated levels of Lactobacillaceae, Lachnospiraceae, Muribaculaceae, and Eggerthellaceae in the treatment groups compared to controls [197]. These studies suggest that oral administration of curcumin may promote the growth and proliferation of beneficial gut microbes.

4.2.2. Anthocyanins

Anthocyanins are water-soluble pigments in the polyphenol family, found in flowers and fruits that have red, blue, or purple hues as well as in nuts and certain vegetables. Anthocyanins are a group of flavonoids with a wide range of uses, such as medicinal herbs, including antidiabetic, anticancer, anti-inflammatory, antimicrobial, and anti-obesity effects [205]. The most common types of anthocyanins include cyanidin, delphinidin, pelargonidin, peonidin, petunidin, and malvidin, which are all potent nutraceutical or pharmaceutical components. These compounds have low bioavailability due to their rapid absorption in the stomach and small intestine, causing low absorption into the blood and circulatory system and high excretion rates and reducing the efficiency of anthocyanins as free-radical scavengers. An in vitro incubation study in 2012 assessed anthocyanins’ effects on the growth of Bifidobacterium spp., Lactobacillus spp., and Bacteroides spp. and found that the mixture of malvidin-3-glucoside, delphinidin-3-glucoside, peonidin-3-glucoside, petunidin-3-glucoside, and cyanidin-3-glucoside had a synergistic effect on the growth of beneficial bacteria [206]. One of the microbiota metabolites of anthocyanin, namely, gallic acid, was observed to have an inhibitory effect on a group of potentially harmful bacteria, including Clostridium histolyticum, without affecting the beneficial bacteria. Furthermore, gallic acid produced from the administration of the anthocyanin mixture significantly reduced the Bacteroides spp. group [206]. A 2012 intervention study aimed to evaluate the effect of the moderate intake of anthocyanins in red wine on certain gut microbial groups on host health benefits and found that red wine polyphenols could have a significant effect on the growth of select gut microbes, including Proteobacteria, Fusobacteria, Firmicutes, and Bacteroidetes [207].

4.2.3. Tea Polyphenols

The evergreen shrub Camellia sinesis can be consumed as unfermented green tea, semifermented oolong tea, and fermented (oxidized) black tea [208,209]. Tea leaves contain a variety of components, including polyphenols, terpenoids, purine alkaloids, the amino acid L-theanine, carbohydrates, caffeine, theaflavins, and a mixture of natural flavonoids called tea catechins (TCs). The health benefits of tea have been attributed in part to their antioxidant and anti-inflammatory properties; however, some benefits have been linked to the relationship between tea bioactive compounds and the gut microbiota [210]. Tea polyphenols have a low bioavailability; additionally, it has been estimated that 90–95% of the dietary polyphenol compounds travel to the large intestine where they are in direct contact with the gut microbiota [211]. Both green and black teas are rich in flavonoids, comprising up to 30% of their dried volume [212].

Theaflavins

Theaflavin (3,4,5-trihydroxybenzocyclohepten-6-one) is a polyphenol and biflavonoid responsible for the red pigment in black and oolong tea formed by oxidation during fermentation [213,214]. Theaflavins (TFs) are further divided into theaflavin (TF1), theaflavin-3-gallated (TF2A), theaflavin-3′-gallate (TF2B), and theaflavin-3,3′-digallate (TF3) [215]. TFs consist of a benzotropolone skeleton that is formed by the oxidation of epicatechin (EC) and epigallocatechin-3-gallate (EGCG) in the presence of polyphenol oxidase and peroxidase enzymes [216]. TFs contribute to the health-promoting effects of the gut microbiota due to the bacteria-mediated metabolism that occurs in the lower GI tract. TFs that reach the large intestine are modified by ring-cleavage, reduction, hydrolysis, decarboxylation, and dihydroxylation reactions [217,218]. An in vitro fecal fermentation study demonstrated that TF administration promoted the growth of Bacteroides, Lachnoclostridium, Faecalibacterium, Parabacteroides, and Bifidobacterium, whereas Prevotella and Fusobacterium growth was inhibited [219]. TF3 was shown to inhibit the growth of gram-negative bacteria, including Klebsiella aerogenes, Escherichia coli, Pseudomonas aeruginosa, and Proteus mirabilis, and gram-positive bacteria, including Staphylococcus aureus, Streptococcus pyogenes, and Mycobacterium smegmatis [220].
The exerted health benefits may be due to the regulation of various cellular signaling pathways, including signaling to mitigate cellular inflammatory responses and blocking of the mitogen-activated protein kinase [221]. TFs have several therapeutic properties, including a protective effect on neuronal cell damage, promotion of immune response, and protection against certain disease initiation via the induction of apoptosis, cell cycle arrest, and suppression of inflammation [222,223]. TFs have been documented to act as strong free-radical scavengers, inhibit oxygen radical-mediated lipid peroxidation, and induce activation in different antioxidant enzymes [215,224]. TFs have been demonstrated in vivo to have the ability to penetrate the blood–brain barrier (BBB) and offer neuroprotection via radical scavenging, antioxidative, antiapoptotic, and cell-regulating pathways [222,225,226]. When TFs are hydrolyzed by Bifidobacteria and Lactobacilli species, the corresponding metabolites TF1, TF2A, TF2B, gallic acid, and pyrogallol become available metabolites in circulation [218,227]. Additionally, two microbial metabolites of TFs, i.e., 3-4′-hydroxyphenylpropionic acid and gallic acid, can increase tyrosine hydroxylase (TH) and dopamine transporter (DAT) immune responses. TFs should be considered promising prebiotic components due to their capacity to increase the abundance of beneficial bacteria and decrease the abundance of potentially harmful bacteria.

Green Tea Catechins

There are four major TC compounds found in green tea leaves, identified as (-)-epicatechin (EC), (-)-epigallocatechin (EGC), (-)-epicatechin gallate (ECG), and (-)-epigallocatechin gallate (EGCG) [228], whereas EGCG is the most abundant and the most active component among catechins. TCs offer a spectrum of health benefits to animals and humans, including protection from cardiovascular diseases [229], cancers [230], and neurodegenerative diseases [231,232]. Several cell-based and animal experiments have provided evidence demonstrating that tea polyphenols and TCs have beneficial effects against PD [66,209,233,234,235]. The mode of action of TCs is mainly through the prevention of aggregate formation and dopamine loss, alleviating mitochondrial dysfunction, antioxidative stress, and anti-neuroinflammation, as well as the activation of neurotrophic factor and signaling pathways [236]. Metabolites of TCs are readily absorbed into circulation and may lead to the enhanced bioavailability and bioactivity of TCs [237,238,239]. TCs have been documented to modify gut-microbiota-dependent metabolisms of bile constituents and micronutrients [240], including the TCA cycle, bile acid metabolism, and metabolism of purine, pyrimidine, and amino acids [241]. EGCG has been reported to cross the BBB after ingestion, leading to increased activity of antioxidants, iron chelation, and antimutagenic effects [232]. Additionally, aromatic rings and hydroxyl groups produced by TC metabolism may provide neuroprotection by reducing lipid peroxidation and antioxidant activity [242].
Overall, the evidence has demonstrated that TCs can significantly impact the biodiversity of host gut microbiota and overall health by stimulating or hindering the growth of certain microbial species [210,243]. Previous studies [234] have shown that long-term supplementation with TCs significantly affected the compositions of the gut microbiome in time-dependent and dose-dependent patterns in Sprague–Dawley (SD) rats. Bacteroidetes and Oscillospira, both beneficial microbial families, were significantly enriched, while Peptostreptococcaceae, a gram-positive bacterium that is overrepresented in the gut of colon cancer patients, was significantly diminished. TCs have shown inhibitory effects on certain bacteria, such as Bacillus cereus, Campylobacter jejuni, Clostridium perfringens, E. coli, H. pylori, Legionella pneumophila, and Mycobacterium species [244]. Gram-negative bacteria are surrounded by a negatively charged lipopolysaccharide membrane that repels catechins [245]; however, some evidence shows that TC supplementation can inhibit the growth of harmful gram-negative bacteria. A 2013 study [246] found that in vitro EGCG, gallocatechin gallate (GCG), and EGCG 3”-methyl could significantly increase the abundance of beneficial bacteria, including Bifidobacterium spp., Lactobacillus, and Enterococcus, successively increasing the production of SCFAs. It was also observed that catechins were able to reduce the growth of Bacteroides, Prevotella, Clostridium hystoliticum, Eubacterium, and Clostridium species [246].

4.3. Polyphenols and α-Synuclein

Research has demonstrated that α-synuclein is a crucial therapeutic target for PD, and inhibiting its aggregation, oligomerization, and fibrillation are key strategies for disease modification [247,248]. Numerous studies have identified a variety of polyphenolic compounds capable of inhibiting fibrils and oligomer formation, as well as stabilizing or disaggregating the α-synuclein oligomers [249,250,251,252]. This makes them promising therapeutic candidates for PD and related synucleinopathies. EGCG, quercetin, polyphenolic acids, curcumin, and their derivatives are some of the most well-known and effective polyphenolic inhibitors of α-synuclein.

4.3.1. EGCG

EGCG is a well-studied inhibitor of α-synuclein fibrillization, with a notable inhibition concentration (IC50) of 9.8 μM [253,254,255,256]. First reported by Ehrnhoefer in 2008 [253], EGCG inhibits α-synuclein by redirecting its aggregation into stable, spherical, and off-pathway oligomers and transforming pre-formed fibrils into unstructured aggregates. Apart from redirecting the aggregated α-synuclein to off-pathway oligomers, EGCG was reported to be able to transform already-formed fibrils into unstructured aggregates without releasing monomers [257]. The binding of EGCG with α-synuclein is non-specific, and it exhibits anti-amyloidogenic properties against multiple proteins, including Aβ and huntingtin [258,259,260]. Further studies suggest that EGCG crosslinks α-synuclein into compact structures that cannot bind to the normal fibrils [261]. Importantly, EGCG was reported to bind to the same binding sites on α-synuclein fibrils as Thioflavin T(ThT) and substitute it, leading to misinterpretation of the aggregation readout [262]. Its efficacy, including in cells overexpressing α-synuclein or its A53T mutant [256], suggests that EGCG-induced aggregates are less likely to damage cell membranes compared to aggregates formed in its absence [263]. However, the full scope of EGCG’s activity remains under investigation and is not yet fully understood [264].

4.3.2. Curcumin

Curcumin has been recognized for its potential to slow the progression of neurodegenerative diseases such as PD [265]. Over the past few decades, the effects of curcumin on α-synuclein aggregation have been widely studied [266,267,268,269,270,271,272,273]. It has been reported that curcumin can inhibit α-synuclein fibrillization in vitro and disassemble already-formed fibrils [266,267,270]. Additionally, several modified analogs of curcumin with improved stability have also been proven effective in inhibiting α-synuclein amyloid aggregation and depolymerizing α-synuclein fibrils [269,274]. Interestingly, some studies suggested that curcumin may bind to oligomers and fibrils, thereby accelerating α-synuclein fibrillation, while producing less toxic α-synuclein aggregates [266,275]. Curcumin has also been shown to prevent hexokinase I (HKI) release and reactive oxygen species (ROS) enhancement triggered by α-synuclein fibrils in mitochondria [276,277]. Animal testing in an α-synuclein transgenic PD mouse model has demonstrated that the curcumin-rich diet showed improvements in mice motor behavior, although no changes in aggregate levels were detected [278].
Although in vitro data have shown that many polyphenols successfully modify α-synuclein, they still require systematic pharmacokinetic evaluations through in vivo studies. Additionally, many polyphenolic compounds face challenges in crossing the BBB due to their non-lipophilic nature, potentially preventing them from reaching the concentrations necessary to exert effects in the brain [279,280]. Several factors, such as stability, solubility in an acidic environment at a gastric pH, absorption pattern, gut microbiota, enterohepatic circulation, first-pass metabolism, and metabolic fate concerning phase I or phase II metabolism, play a key role in achieving the ideal bioavailability of the phytochemicals in the brain [281,282].

5. Interplay between Polyphenols, Gut Microbiota, and α-Synuclein

With increasing recognition of the importance of the gut–brain axis in PD etiology, there is increasing interest in developing interventional strategies that target and attenuate α-synuclein aggregation and dispersion. As discussed in the above section, polyphenols are capable of regulating the gut microbiome composition and the metabolic pathways, mostly through the inhibition of pathogenic bacteria and the stimulation of beneficial bacteria [283,284,285]; in turn, polyphenols are extensively metabolized by the gut bacteria, resulting in the generation of bioactive secondary metabolites, which enhance bioavailability [286]. These gut bacterial metabolites inhibit fibrils and oligomer formation and stabilize or disaggregate the α-synuclein oligomers. More recent research suggested that the microbial metabolites of polyphenols can inhibit the spread of α-synuclein in a cell-based system [249,287] (Figure 4). However, the mechanisms of such an interaction between polyphenols, gut microbiota, and α-synuclein are largely unknown.

5.1. Bioactivity and Efficacy of Polyphenols Are Affected by Gut Microbiota

Polyphenols that are most commonly found in dietary components are glycosides [288], which are not completely absorbed in the upper GI tract; only aglycones and some glucosides can be absorbed in the intestinal mucosa [289]. Recent evidence shows that the bioactivity and efficacy of polyphenols are significantly influenced by the gut microbiota [286,290,291,292]. The microbes in the gut can transform polyphenols into various metabolites that may have different bioactive properties from the original compounds [293,294]. Additionally, the composition of an individual’s gut microbiota, which varies greatly among individuals, can determine the degree to which polyphenols are metabolized and utilized [295]. For example, interpersonal heterogeneity in gut microbiota may lead to interpersonal variabilities for their efficacy in metabolizing dietary flavanols into certain biologically available phenolic acid metabolites that interfere with α-synuclein misfolding [252]. Preclinical investigations have demonstrated that dietary supplementation with select bioactive polyphenol-rich dietary preparations, such as a grape seed polyphenol extract (GSPE) and a standardized Bioactive Dietary Polyphenol Preparation (BDPP, comprised of a select Concord grape juice, GSPE, and resveratrol) is mechanistically effective in modulating diverse neuropathologic phenotypes [296]. Furthermore, treatment with a flavanol-rich preparation (FRP) in gnotobiotic mice yielded brain-bioavailable polyphenol metabolites [297]. A recent review highlighted that polyphenols’ neuroprotective effects can be direct, after the metabolites of polyphenols cross the BBB, or indirect, with influences on gut microbial communities [298]. Clinical trials have demonstrated that polyphenol metabolites are positively associated with cerebral blood flow and oxygenation, resulting in direct neuroprotective effects [299,300]. Meanwhile, the gut microbiome can produce active metabolites of polyphenols, which indirectly enhance their neuroprotective capacity, an example of which is curcumin being transformed into demethylcurcumin and bisdemethoxycurcumin [298,301]. Therefore, understanding the interaction between polyphenols and gut microbiota, and identifying the specific microbes that enhance the efficacy of polyphenols is crucial for developing therapeutic strategies and precision nutrition based on the microbiome.

5.2. Anti-α-Synuclein Microbial Polyphenol Metabolites

As α-synuclein is a crucial therapeutic target for PD, inhibiting its aggregation, oligomerization, and fibrillation are key strategies for disease modification [302]. Polyphenols are among the emerging therapeutic options for combatting abhorrent α-synuclein as they exist in a wide variety of chemical compounds and have the ability to produce secondary metabolites after undergoing microbial transformation by gut microbiota. These metabolites are known for their varied pharmacological properties, which have been extensively documented in scientific reviews [294,295,303,304].
Ono and colleagues [252] established two experimental groups of humanized gnotobiotic mice with diverse gut bacteria compositions and administered a polyphenol-rich preparation orally. They detected 15 polyphenol-derived microbial metabolites, which are generated by the gut microbiota fermentation of grape seed polyphenol extract, in the cecal compartment, including caffeic acid, ferulic acid (FA), gallic acid (GA), and vanillic acid at μM to sub-μM concentrations. Notably, three metabolites—3,4-dihydroxybenzoic acid (3,4-diHBA), 3-hydroxybenzoic acid (3-HBA), and 3-(3′-hydroxyphenyl) propionic acid (3-HPPA)—were found to accumulate in the brain. The in vitro study confirmed that 3-HBA, 3,4-diHBA, and 3-HPPA inhibit α-synuclein aggregation, including the formation of low-order oligomers such as dimers and trimers. The findings align with reports from another study that showed that 3-HBA and 3-HPPA prevent the misfolding and assembly of Aβ peptides into neurotoxic aggregates, such as Aβ oligomers [305]. Furthermore, using the A53T mutant Drosophila model of PD, these metabolites effectively improved motor dysfunction, indicating a mitigating effect on mutant α-synuclein-mediated neurotoxicity [306]. Very recently, 3-HPPA, 3,4-diHBA, 3-HBA, and 4-HBA were found to significantly attenuate intracellular α-synuclein seeding aggregation in a cell-based system, and the findings were confirmed using insoluble α-synuclein aggregates extracted from post-mortem Multiple System Atrophy (MSA) and PD brain specimens [249].

6. Conclusions and Perspectives

Dietary polyphenols are promising protective agents for PD prevention because of their abundance and relatively low toxicity [307]. Previous studies have shown that polyphenols can combat PD through multiple mechanisms, including reducing neuronal apoptosis, attenuating oxidative stress, and downregulating neuroinflammation. Recent evidence has also demonstrated that polyphenols inhibit the formation and spread of α-synuclein aggregates, a hallmark of PD pathology, which may originate from the gut. Furthermore, polyphenols are capable of regulating the gut microbiota composition and its metabolic pathways, mostly through the inhibition of pathogenic bacteria and the stimulation of beneficial bacteria [283]; in turn, polyphenols are extensively metabolized by gut bacteria, resulting in the generation of bioactive secondary metabolites that enhance their bioavailability.
While polyphenols showed benefits in animal models of PD, evidence from epidemiological studies primarily demonstrated the association between the dietary intake of polyphenols and a reduced risk of developing PD. However, data from prospective randomized controlled trials in patients with pre-existing PD are limited. Therefore, future clinical studies are necessary to evaluate the effectiveness of dietary polyphenols in slowing the progression of PD. Although the existing evidence points to the potential of polyphenols to favorably modulate the gut–brain axis, it is evident that more focused research is needed to fully understand the mechanisms between polyphenols, the gut microbiota, and α-synuclein in order to establish the therapeutic viability of polyphenols in clinical settings.

Author Contributions

E.R. conceived and wrote the paper. K.S.X., J.-S.W. and L.T. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funding from the USDA NIFA (No. 2022-67017-36237).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tysnes, O.B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef] [PubMed]
  2. Cerri, S.; Mus, L.; Blandini, F. Parkinson’s Disease in Women and Men: What’s the Difference? J. Parkinson’s Dis. 2019, 9, 501–515. [Google Scholar] [CrossRef] [PubMed]
  3. Marras, C.; Beck, J.C.; Bower, J.H.; Roberts, E.; Ritz, B.; Ross, G.W.; Abbott, R.D.; Savica, R.; Van Den Eeden, S.K.; Willis, A.W.; et al. Prevalence of Parkinson’s disease across North America. NPJ Parkinson’s Dis. 2018, 4, 21. [Google Scholar] [CrossRef] [PubMed]
  4. Armstrong, M.J.; Okun, M.S. Diagnosis and Treatment of Parkinson Disease: A Review. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef] [PubMed]
  5. Fahn, S. Description of Parkinson’s disease as a clinical syndrome. Ann. N. Y. Acad. Sci. 2003, 991, 1–14. [Google Scholar] [CrossRef] [PubMed]
  6. Forno, L.S. The Lewy body in Parkinson’s disease. Adv. Neurol. 1987, 45, 35–43. [Google Scholar] [PubMed]
  7. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001, 2, 492–501. [Google Scholar] [CrossRef] [PubMed]
  8. Bras, I.C.; Xylaki, M.; Outeiro, T.F. Mechanisms of alpha-synuclein toxicity: An update and outlook. Prog. Brain Res. 2020, 252, 91–129. [Google Scholar]
  9. Omotosho, A.O.; Tajudeen, Y.A.; Oladipo, H.J.; Yusuff, S.I.; AbdulKadir, M.; Muili, A.O.; Egbewande, O.M.; Yusuf, R.O.; Faniran, Z.O.; Afolabi, A.O.; et al. Parkinson’s disease: Are gut microbes involved? Brain Behav. 2023, 13, e3130. [Google Scholar] [CrossRef]
  10. Li, Q.; Meng, L.-B.; Chen, L.-J.; Shi, X.; Tu, L.; Zhou, Q.; Yu, J.-L.; Liao, X.; Zeng, Y.; Yuan, Q.-Y. The role of the microbiota-gut-brain axis and intestinal microbiome dysregulation in Parkinson’s disease. Front. Neurol. 2023, 14, 1185375. [Google Scholar] [CrossRef]
  11. Pant, A.; Bisht, K.S.; Aggarwal, S.; Maiti, T.K. Human gut microbiota and Parkinson’s disease. Prog. Mol. Biol. Transl. Sci. 2022, 192, 281–307. [Google Scholar] [PubMed]
  12. 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]
  13. Menozzi, E.; Macnaughtan, J.; Schapira, A.H.V. The gut-brain axis and Parkinson disease: Clinical and pathogenetic relevance. Ann. Med. 2021, 53, 611–625. [Google Scholar] [CrossRef]
  14. Elsworth, J.D. Parkinson’s disease treatment: Past, present, and future. J. Neural Transm. 2020, 127, 785–791. [Google Scholar] [CrossRef] [PubMed]
  15. Solla, P.; Cannas, A.; Congia, S.; Floris, G.; Aste, R.; Tacconi, P.; Marrosu, M.G. Levodopa/carbidopa/entacapone-induced acute Pisa syndrome in a Parkinson’s disease patient. J. Neurol. Sci. 2008, 275, 154–156. [Google Scholar] [CrossRef] [PubMed]
  16. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef] [PubMed]
  17. Dehay, B.; Bourdenx, M.; Gorry, P.; Przedborski, S.; Vila, M.; Hunot, S.; Singleton, A.; Olanow, C.W.; Merchant, K.M.; Bezard, E.; et al. Targeting α-synuclein for treatment of Parkinson’s disease: Mechanistic and therapeutic considerations. Lancet Neurol. 2015, 14, 855–866. [Google Scholar] [CrossRef] [PubMed]
  18. Norris, E.H.; Giasson, B.I.; Lee, V.M. Alpha-synuclein: Normal function and role in neurodegenerative diseases. Curr. Top. Dev. Biol. 2004, 60, 17–54. [Google Scholar] [PubMed]
  19. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. α-Synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841. [Google Scholar] [CrossRef]
  20. Karpowicz, R.J.; Trojanowski, J.Q.; Lee, V.M.-Y. Transmission of alpha-synuclein seeds in neurodegenerative disease: Recent developments. Lab. Investig. 2019, 99, 971–981. [Google Scholar] [CrossRef]
  21. Burré, J.; Sharma, M.; Tsetsenis, T.; Buchman, V.; Etherton, M.R.; Südhof, T.C. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010, 329, 1663–1667. [Google Scholar] [CrossRef] [PubMed]
  22. Walker, L.C.; Jucker, M. Neurodegenerative diseases: Expanding the prion concept. Annu. Rev. Neurosci. 2015, 38, 87–103. [Google Scholar]
  23. Lashuel, H.A.; Overk, C.R.; Oueslati, A.; Masliah, E. The many faces of α-synuclein: From structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 2013, 14, 38–48. [Google Scholar] [CrossRef] [PubMed]
  24. Mehra, S.; Sahay, S.; Maji, S.K. α-Synuclein misfolding and aggregation: Implications in Parkinson’s disease pathogenesis. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2019, 1867, 890–908. [Google Scholar] [CrossRef] [PubMed]
  25. Anderson, J.P.; Walker, D.E.; Goldstein, J.M.; de Laat, R.; Banducci, K.; Caccavello, R.J.; Barbour, R.; Huang, J.; Kling, K.; Lee, M.; et al. Phosphorylation of Ser-129 is the dominant pathological modification of α-synuclein in familial and sporadic Lewy body disease. J. Biol. Chem. 2006, 281, 29739–29752. [Google Scholar] [CrossRef] [PubMed]
  26. Yasuda, T.; Nakata, Y.; Mochizuki, H. Alpha-Synuclein and neuronal cell death. Mol. Neurobiol. 2013, 47, 466–483. [Google Scholar] [CrossRef]
  27. Benskey, M.J.; Perez, R.G.; Manfredsson, F.P. The contribution of alpha synuclein to neuronal survival and function—Implications for Parkinson’s disease. J. Neurochem. 2016, 137, 331–359. [Google Scholar] [CrossRef]
  28. Pozo Devoto, V.M.; Falzone, T.L. Mitochondrial dynamics in Parkinson’s disease: A role for alpha-synuclein? Dis. Model. Mech. 2017, 10, 1075–1087. [Google Scholar] [CrossRef]
  29. Lee, S.J. Alpha-synuclein aggregation: A link between mitochondrial defects and Parkinson’s disease? Antioxid. Redox Signal. 2003, 5, 337–348. [Google Scholar] [CrossRef]
  30. Mullin, S.; Schapira, A. Alpha-Synuclein and mitochondrial dysfunction in Parkinson’s disease. Mol. Neurobiol. 2013, 47, 587–597. [Google Scholar] [CrossRef]
  31. Ozawa, T.; Tanaka, M.; Suzuki, H.; Nishikimi, M. Structure and function of mitochondria: Their organization and disorders. Brain Dev. 1987, 9, 76–81. [Google Scholar] [CrossRef] [PubMed]
  32. Sherratt, H.S. Mitochondria: Structure and function. Rev. Neurol. 1991, 147, 417–430. [Google Scholar] [PubMed]
  33. Sohrabi, T.; Mirzaei-Behbahani, B.; Zadali, R.; Pirhaghi, M.; Morozova-Roche, L.A.; Meratan, A.A. Common Mechanisms Underlying alpha-Synuclein-Induced Mitochondrial Dysfunction in Parkinson’s Disease. J. Mol. Biol. 2023, 435, 167992. [Google Scholar] [CrossRef] [PubMed]
  34. Bellani, S.; Sousa, V.L.; Ronzitti, G.; Valtorta, F.; Meldolesi, J.; Chieregatti, E. The regulation of synaptic function by alpha-synuclein. Commun. Integr. Biol. 2010, 3, 106–109. [Google Scholar] [CrossRef] [PubMed]
  35. Burre, J. The Synaptic Function of alpha-Synuclein. J. Parkinson’s Dis. 2015, 5, 699–713. [Google Scholar] [CrossRef] [PubMed]
  36. Parra-Rivas, L.A.; Madhivanan, K.; Aulston, B.D.; Wang, L.; Prakashchand, D.D.; Boyer, N.P.; Saia-Cereda, V.M.; Branes-Guerrero, K.; Pizzo, D.P.; Bagchi, P.; et al. Serine-129 phosphorylation of alpha-synuclein is an activity-dependent trigger for physiologic protein-protein interactions and synaptic function. Neuron 2023, 111, 4006–4023.e10. [Google Scholar] [CrossRef] [PubMed]
  37. Sharma, M.; Burre, J. Alpha-Synuclein in synaptic function and dysfunction. Trends Neurosci. 2023, 46, 153–166. [Google Scholar] [CrossRef] [PubMed]
  38. Desplats, P.; Lee, H.-J.; Bae, E.-J.; Patrick, C.; Rockenstein, E.; Crews, L.; Spencer, B.; Masliah, E.; Lee, S.-J. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of α-synuclein. Proc. Natl. Acad. Sci. USA 2009, 106, 13010–13015. [Google Scholar] [CrossRef] [PubMed]
  39. Mougenot, A.-L.; Nicot, S.; Bencsik, A.; Morignat, E.; Verchère, J.; Lakhdar, L.; Legastelois, S.; Baron, T. Prion-like acceleration of a synucleinopathy in a transgenic mouse model. Neurobiol. Aging 2012, 33, 2225–2228. [Google Scholar] [CrossRef]
  40. Kordower, J.H.; Chu, Y.; Hauser, R.A.; Olanow, C.; Freeman, T.B. Transplanted dopaminergic neurons develop PD pathologic changes: A second case report. Mov. Disord. 2008, 23, 2303–2306. [Google Scholar]
  41. Conway, K.A.; Harper, J.D.; Lansbury, P.T. Accelerated in vitro fibril formation by a mutant α-synuclein linked to early-onset Parkinson disease. Nat. Med. 1998, 4, 1318–1320. [Google Scholar] [CrossRef] [PubMed]
  42. Hansen, C.; Angot, E.; Bergström, A.-L.; Steiner, J.A.; Pieri, L.; Paul, G.; Outeiro, T.F.; Melki, R.; Kallunki, P.; Fog, K.; et al. α-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J. Clin. Investig. 2011, 121, 715–725. [Google Scholar] [CrossRef] [PubMed]
  43. Ullah, H.; Arbab, S.; Tian, Y.; Liu, C.-Q.; Chen, Y.; Qijie, L.; Khan, M.I.U.; Hassan, I.U.; Li, K. The gut microbiota-brain axis in neurological disorder. Front. Neurosci. 2023, 17, 1225875. [Google Scholar] [CrossRef] [PubMed]
  44. 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] [PubMed]
  45. Yang, Z.-X.; Zhang, Y.; Wang, Q.; Zhang, L.; Liu, Y.-F.; Zhang, Y.; Ren, Y.; Zhou, C.; Gao, H.-W.; Zhang, N.-X.; et al. Addition of α-synuclein aggregates to the intestinal environment recapitulates Parkinsonian symptoms in model systems. Acta Pharmacol. Sin. 2024, 45, 36–51. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, K.; Zhang, C.; Zhang, B.; Li, G.; Shi, G.; Cai, Q.; Huang, M. Gut dysfunction may be the source of pathological aggregation of alpha-synuclein in the central nervous system through Paraquat exposure in mice. Ecotoxicol. Environ. Saf. 2022, 246, 114152. [Google Scholar] [CrossRef] [PubMed]
  47. Holmqvist, S.; Chutna, O.; Bousset, L.; Aldrin-Kirk, P.; Li, W.; Björklund, T.; Wang, Z.-Y.; Roybon, L.; Melki, R.; Li, J.-Y. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 2014, 128, 805–820. [Google Scholar] [CrossRef] [PubMed]
  48. Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef] [PubMed]
  49. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  50. O’Mahony, S.M.; Hyland, N.P.; Dinan, T.G.; Cryan, J.F. Maternal separation as a model of brain–gut axis dysfunction. Psychopharmacology 2011, 214, 71–88. [Google Scholar] [CrossRef]
  51. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef] [PubMed]
  52. 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] [PubMed]
  53. Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.-F. Impact of microbiota on central nervous system and neurological diseases: The gut-brain axis. J. Neuroinflammation 2019, 16, 53. [Google Scholar] [CrossRef] [PubMed]
  54. Yarandi, S.S.; Peterson, D.A.; Treisman, G.J.; Moran, T.H.; Pasricha, P.J. Modulatory effects of gut microbiota on the central nervous system: How gut could play a role in neuropsychiatric health and diseases. J. Neurogastroenterol. Motil. 2016, 22, 201. [Google Scholar] [CrossRef] [PubMed]
  55. Raj, K.; Singh, S.; Chib, S.; Mallan, S. Microbiota-Brain-Gut-Axis Relevance to Parkinson’s Disease: Potential Therapeutic Effects of Probiotics. Curr. Pharm. Des. 2022, 28, 3049–3067. [Google Scholar] [CrossRef] [PubMed]
  56. Loh, J.S.; Mak, W.Q.; Tan, L.K.S.; Ng, C.X.; Chan, H.H.; Yeow, S.H.; Foo, J.B.; Ong, Y.S.; How, C.W.; Khaw, K.Y. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct. Target Ther. 2024, 9, 37. [Google Scholar] [PubMed]
  57. Chen, S.-J.; Lin, C.-H. Gut microenvironmental changes as a potential trigger in Parkinson’s disease through the gut-brain axis. J. Biomed. Sci. 2022, 29, 54. [Google Scholar] [CrossRef] [PubMed]
  58. Dowling, L.R.; Strazzari, M.R.; Keely, S.; Kaiko, G.E. Enteric nervous system and intestinal epithelial regulation of the gut-brain axis. J. Allergy Clin. Immunol. 2022, 150, 513–522. [Google Scholar] [CrossRef] [PubMed]
  59. dos Santos, J.C.C.; Oliveira, L.F.; Noleto, F.M.; Gusmão, C.T.P.; Brito, G.A.d.C.; Viana, G.S.d.B. Gut-microbiome-brain axis: The crosstalk between the vagus nerve, alpha-synuclein and the brain in Parkinson’s disease. Neural Regen. Res. 2023, 18, 2611–2614. [Google Scholar] [CrossRef]
  60. 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]
  61. Elfil, M.; Kamel, S.; Kandil, M.; Koo, B.B.; Schaefer, S.M. Implications of the Gut Microbiome in Parkinson’s Disease. Mov. Disord. 2020, 35, 921–933. [Google Scholar] [CrossRef]
  62. Nie, S.; Ge, Y. The link between the gut microbiome, inflammation, and Parkinson’s disease. Appl. Microbiol. Biotechnol. 2023, 107, 6737–6749. [Google Scholar] [CrossRef]
  63. Nie, S.; Wang, J.; Deng, Y.; Ye, Z.; Ge, Y. Inflammatory microbes and genes as potential biomarkers of Parkinson’s disease. NPJ Biofilms Microbiomes 2022, 8, 101. [Google Scholar] [CrossRef] [PubMed]
  64. Zhu, M.; Liu, X.; Ye, Y.; Yan, X.; Cheng, Y.; Zhao, L.; Chen, F.; Ling, Z. Gut Microbiota: A Novel Therapeutic Target for Parkinson’s Disease. Front. Immunol. 2022, 13, 937555. [Google Scholar] [CrossRef] [PubMed]
  65. 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 Parkinson’s Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
  66. 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]
  67. 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]
  68. 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] [PubMed]
  69. Devos, D.; Lebouvier, T.; Lardeux, B.; Biraud, M.; Rouaud, T.; Pouclet, H.; Coron, E.; Bruley des Varannes, S.; Naveilhan, P.; Nguyen, J.-M.; et al. Colonic inflammation in Parkinson’s disease. Neurobiol. Dis. 2013, 50, 42–48. [Google Scholar] [CrossRef]
  70. 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]
  71. Li, Y.; Chen, Y.; Jiang, L.; Zhang, J.; Tong, X.; Chen, D.; Le, W. Intestinal Inflammation and Parkinson’s Disease. Aging Dis. 2021, 12, 2052–2068. [Google Scholar] [CrossRef]
  72. Lubomski, M.; Tan, A.H.; Lim, S.-Y.; Holmes, A.J.; Davis, R.L.; Sue, C.M. Parkinson’s disease and the gastrointestinal microbiome. J. Neurol. 2020, 267, 2507–2523. [Google Scholar] [CrossRef] [PubMed]
  73. 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]
  74. Brown, G.C. The endotoxin hypothesis of neurodegeneration. J. Neuroinflammation 2019, 16, 180. [Google Scholar] [CrossRef] [PubMed]
  75. Silva, D.F.; Empadinhas, N.; Cardoso, S.M.; Esteves, A.R. Neurodegenerative Microbially-Shaped Diseases: Oxidative Stress Meets Neuroinflammation. Antioxidants 2022, 11, 2141. [Google Scholar] [CrossRef] [PubMed]
  76. Hirayama, M.; Ohno, K. Parkinson’s Disease and Gut Microbiota. Ann. Nutr. Metab. 2021, 77 (Suppl. S2), 28–35. [Google Scholar] [CrossRef] [PubMed]
  77. Çamcı, G.; Oğuz, S. Association between Parkinson’s Disease and Helicobacter Pylori. J. Clin. Neurol. 2016, 12, 147–150. [Google Scholar] [CrossRef] [PubMed]
  78. Deng, I.; Corrigan, F.; Zhai, G.; Zhou, X.-F.; Bobrovskaya, L. Lipopolysaccharide animal models of Parkinson’s disease: Recent progress and relevance to clinical disease. Brain Behav. Immun. Health 2020, 4, 100060. [Google Scholar] [CrossRef] [PubMed]
  79. Cryan, J.F.; O’Riordan, K.J.; Sandhu, K.; Peterson, V.; Dinan, T.G. The gut microbiome in neurological disorders. Lancet Neurol. 2020, 19, 179–194. [Google Scholar] [CrossRef] [PubMed]
  80. Heinzel, S.; Aho, V.T.E.; Suenkel, U.; Von Thaler, A.K.; Schulte, C.; Deuschle, C.; Paulin, L.; Hantunen, S.; Brockmann, K.; Eschweiler, G.W.; et al. Gut Microbiome Signatures of Risk and Prodromal Markers of Parkinson Disease. Ann. Neurol. 2021, 90, E1–E12. [Google Scholar] [CrossRef]
  81. Rani, L.; Mondal, A.C. Unravelling the role of gut microbiota in Parkinson’s disease progression: Pathogenic and therapeutic implications. Neurosci. Res. 2021, 168, 100–112. [Google Scholar] [CrossRef] [PubMed]
  82. Aho, V.T.E.; Houser, M.C.; Pereira, P.A.B.; Chang, J.; Rudi, K.; Paulin, L.; Hertzberg, V.; Auvinen, P.; Tansey, M.G.; Scheperjans, F. Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in Parkinson’s disease. Mol. Neurodegener. 2021, 16, 6. [Google Scholar] [CrossRef] [PubMed]
  83. 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]
  84. Toh, T.S.; Chong, C.W.; Lim, S.-Y.; Bowman, J.; Cirstea, M.; Lin, C.-H.; Chen, C.-C.; Appel-Cresswell, S.; Finlay, B.B.; Tan, A.H. Gut microbiome in Parkinson’s disease: New insights from meta-analysis. Park. Relat. Disord. 2022, 94, 1–9. [Google Scholar] [CrossRef]
  85. 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]
  86. Zhou, S.; Li, B.; Deng, Y.; Yi, J.; Mao, G.; Wang, R.; Zeng, W.; Liu, B.; Wu, D.; Liu, F. Meta-analysis of the relations between gut microbiota and pathogens and Parkinson’s disease. Adv. Clin. Exp. Med. 2023, 32, 613–621. [Google Scholar] [CrossRef] [PubMed]
  87. 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]
  88. 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]
  89. 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]
  90. 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]
  91. 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-naive Parkinson’s disease patients. Genome Med. 2017, 9, 39. [Google Scholar] [CrossRef]
  92. 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]
  93. Petrov, V.A.; Saltykova, I.V.; Zhukova, I.A.; Alifirova, V.M.; Zhukova, N.G.; Dorofeeva, Y.B.; Tyakht, A.V.; Kovarsky, B.A.; Alekseev, D.G.; Kostryukova, E.S.; et al. Analysis of Gut Microbiota in Patients with Parkinson’s Disease. Bull. Exp. Biol. Med. 2017, 162, 734–737. [Google Scholar] [CrossRef] [PubMed]
  94. Caputi, V.; Giron, M.C. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease. Int. J. Mol. Sci. 2018, 19, 1689. [Google Scholar] [CrossRef]
  95. 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]
  96. 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] [PubMed]
  97. Lin, C.-H.; Chen, C.-C.; Chiang, H.-L.; Liou, J.-M.; Chang, C.-M.; Lu, T.-P.; Chuang, E.Y.; Tai, Y.-C.; Cheng, C.; Lin, H.-Y.; et al. Altered gut microbiota and inflammatory cytokine responses in patients with Parkinson’s disease. J. Neuroinflammation 2019, 16, 129. [Google Scholar] [CrossRef] [PubMed]
  98. 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]
  99. 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]
  100. 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]
  101. 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]
  102. 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] [PubMed]
  103. 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] [PubMed]
  104. Mehanna, M.; AbuRaya, S.; Ahmed, S.M.; Ashmawy, G.; Ibrahim, A.; AbdelKhaliq, E. Study of the gut microbiome in Egyptian patients with Parkinson’s Disease. BMC Microbiol. 2023, 23, 196. [Google Scholar] [CrossRef] [PubMed]
  105. Hertel, J.; Harms, A.C.; Heinken, A.; Baldini, F.; Thinnes, C.C.; Glaab, E.; Vasco, D.A.; Pietzner, M.; Stewart, I.; Wareham, N.; 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]
  106. 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]
  107. 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]
  108. Kalyanaraman, B.; Cheng, G.; Hardy, M. Gut microbiome, short-chain fatty acids, alpha-synuclein, neuroinflammation, and ROS/RNS: Relevance to Parkinson’s disease and therapeutic implications. Redox Biol. 2024, 71, 103092. [Google Scholar] [CrossRef]
  109. Schaeffer, E.; Kluge, A.; Bottner, M.; Zunke, F.; Cossais, F.; Berg, D.; Arnold, P. Alpha Synuclein Connects the Gut-Brain Axis in Parkinson’s Disease Patients—A View on Clinical Aspects, Cellular Pathology and Analytical Methodology. Front. Cell. Dev. Biol. 2020, 8, 573696. [Google Scholar] [CrossRef]
  110. Böttner, M.; Zorenkov, D.; Hellwig, I.; Barrenschee, M.; Harde, J.; Fricke, T.; Deuschl, G.; Egberts, J.-H.; Becker, T.; Fritscher-Ravens, A.; et al. Expression pattern and localization of alpha-synuclein in the human enteric nervous system. Neurobiol. Dis. 2012, 48, 474–480. [Google Scholar] [CrossRef]
  111. Barrenschee, M.; Zorenkov, D.; Böttner, M.; Lange, C.; Cossais, F.; Scharf, A.B.; Deuschl, G.; Schneider, S.A.; Ellrichmann, M.; Fritscher-Ravens, A.; et al. Distinct pattern of enteric phospho-alpha-synuclein aggregates and gene expression profiles in patients with Parkinson’s disease. Acta Neuropathol. Commun. 2017, 5, 1. [Google Scholar] [CrossRef]
  112. Sánchez-Ferro, Á.; Rábano, A.; Catalán, M.J.; Rodríguez-Valcárcel, F.C.; Díez, S.F.; Herreros-Rodríguez, J.; García-Cobos, E.; Álvarez-Santullano, M.M.; López-Manzanares, L.; Mosqueira, A.J.; et al. In vivo gastric detection of alpha-synuclein inclusions in Parkinson’s disease. Mov. Disord. 2015, 30, 517–524. [Google Scholar] [CrossRef] [PubMed]
  113. Shannon, K.M.; Keshavarzian, A.; Mutlu, E.; Dodiya, H.B.; Daian, D.; Jaglin, J.A.; Kordower, J.H. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov. Disord. 2012, 27, 709–715. [Google Scholar] [CrossRef]
  114. Fayyad, M.; Salim, S.; Majbour, N.; Erskine, D.; Stoops, E.; Mollenhauer, B.; El-Agnaf, O.M.A. Parkinson’s disease biomarkers based on alpha-synuclein. J. Neurochem. 2019, 150, 626–636. [Google Scholar] [CrossRef] [PubMed]
  115. Hawkes, C.H.; Del Tredici, K.; Braak, H. A timeline for Parkinson’s disease. Park. Relat. Disord. 2010, 16, 79–84. [Google Scholar] [CrossRef]
  116. Schmitt, V.; Masanetz, R.K.; Weidenfeller, M.; Ebbinghaus, L.S.; Süß, P.; Rosshart, S.P.; von Hörsten, S.; Zunke, F.; Winkler, J.; Xiang, W. Gut-to-brain spreading of pathology in synucleinopathies: A focus on molecular signalling mediators. Behav. Brain Res. 2023, 452, 114574. [Google Scholar] [CrossRef]
  117. Braak, H.; Rüb, U.; Gai, W.P.; Del Tredici, K. Idiopathic Parkinson’s disease: Possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm. 2003, 110, 517–536. [Google Scholar] [CrossRef] [PubMed]
  118. Visanji, N.P.; Brooks, P.L.; Hazrati, L.-N.; Lang, A.E. The prion hypothesis in Parkinson’s disease: Braak to the future. Acta Neuropathol. Commun. 2013, 1, 2. [Google Scholar] [CrossRef]
  119. Kim, S.; Kwon, S.-H.; Kam, T.-I.; Panicker, N.; Karuppagounder, S.S.; Lee, S.; Lee, J.H.; Kim, W.R.; Kook, M.; Foss, C.A.; et al. Transneuronal Propagation of Pathologic alpha-Synuclein from the Gut to the Brain Models Parkinson’s Disease. Neuron 2019, 103, 627–641.e7. [Google Scholar] [CrossRef]
  120. Dautan, D.; Paslawski, W.; Montejo, S.G.; Doyon, D.C.; Marangiu, R.; Kaplitt, M.G.; Chen, R.; Dawson, V.L.; Zhang, X.; Dawson, T.M.; et al. Gut-Initiated Alpha Synuclein Fibrils Drive Parkinson’s Disease Phenotypes: Temporal Mapping of non-Motor Symptoms and REM Sleep Behavior Disorder. bioRxiv 2024. [Google Scholar] [CrossRef]
  121. Anis, E.; Xie, A.; Brundin, L.; Brundin, P. Digesting recent findings: Gut alpha-synuclein, microbiome changes in Parkinson’s disease. Trends Endocrinol. Metab. 2023, 34, 426. [Google Scholar] [CrossRef] [PubMed]
  122. Yan, Y.; Ren, S.; Duan, Y.; Lu, C.; Niu, Y.; Wang, Z.; Inglis, B.; Ji, W.; Zheng, Y.; Si, W. Gut microbiota and metabolites of alpha-synuclein transgenic monkey models with early stage of Parkinson’s disease. NPJ Biofilms Microbiomes 2021, 7, 69. [Google Scholar] [CrossRef] [PubMed]
  123. Nielsen, S.D.; Pearson, N.M.; Seidler, K. The link between the gut microbiota and Parkinson’s Disease: A systematic mechanism review with focus on α-synuclein transport. Brain Res. 2021, 1769, 147609. [Google Scholar] [CrossRef] [PubMed]
  124. de Ora, L.O.; Balsamo, J.M.; Uyeda, K.S.; Bess, E.N. Discovery of a Gut Bacterial Metabolic Pathway that Drives alpha-Synuclein Aggregation. ACS Chem. Biol. 2024, 19, 1011–1021. [Google Scholar] [CrossRef] [PubMed]
  125. Parikh, N.S.; Ahlawat, R. Helicobacter Pylori; StatPearls: Treasure Island, FL, USA, 2023. [Google Scholar]
  126. Fernandez-Espejo, E. Microorganisms associated with increased risk of Parkinson’s disease. Neurologia 2022, 38, 495–503. [Google Scholar] [CrossRef] [PubMed]
  127. Hawkes, C.H.; Del Tredici, K.; Braak, H. Parkinson’s disease: A dual-hit hypothesis. Neuropathol. Appl. Neurobiol. 2007, 33, 599–614. [Google Scholar] [CrossRef] [PubMed]
  128. Barnhart, M.M.; Chapman, M.R. Curli biogenesis and function. Annu. Rev. Microbiol. 2006, 60, 131–147. [Google Scholar] [CrossRef] [PubMed]
  129. 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]
  130. 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 alpha-synuclein aggregation and motor impairment in mice. eLife 2020, 9, e53111. [Google Scholar] [CrossRef]
  131. Braak, H.; Del Tredici, K.; Rüb, U.; de Vos, R.A.; Steur, E.N.J.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
  132. Hawkes, C.H.; Del Tredici, K.; Braak, H. Parkinson’s disease: The dual hit theory revisited. Ann. N. Y. Acad. Sci. 2009, 1170, 615–622. [Google Scholar] [CrossRef] [PubMed]
  133. Beach, T.G.; White, C.L.; Hladik, C.L.; Sabbagh, M.N.; Connor, D.J.; Shill, H.A.; Sue, L.I.; Sasse, J.; Bachalakuri, J.; Henry-Watson, J. Olfactory bulb α-synucleinopathy has high specificity and sensitivity for Lewy body disorders. Acta Neuropathol. 2009, 117, 169–174. [Google Scholar] [CrossRef] [PubMed]
  134. Cersosimo, M.G.; Benarroch, E.E. Pathological correlates of gastrointestinal dysfunction in Parkinson’s disease. Neurobiol. Dis. 2012, 46, 559–564. [Google Scholar] [CrossRef] [PubMed]
  135. Doty, R.L. Olfaction in Parkinson’s disease and related disorders. Neurobiol. Dis. 2012, 46, 527–552. [Google Scholar] [CrossRef] [PubMed]
  136. Kuo, Y.-M.; Li, Z.; Jiao, Y.; Gaborit, N.; Pani, A.K.; Orrison, B.M.; Bruneau, B.G.; Giasson, B.I.; Smeyne, R.J.; Gershon, M.D.; et al. Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated α-synuclein gene mutations precede central nervous system changes. Hum. Mol. Genet. 2010, 19, 1633–1650. [Google Scholar] [CrossRef] [PubMed]
  137. Wakabayashi, K.; Takahashi, H.; Takeda, S.; Ohama, E.; Ikuta, F. Parkinson’s disease: The presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol. 1988, 76, 217–221. [Google Scholar] [CrossRef] [PubMed]
  138. Bloch, A.; Probst, A.; Bissig, H.; Adams, H.; Tolnay, M. α-Synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects. Neuropathol. Appl. Neurobiol. 2006, 32, 284–295. [Google Scholar] [CrossRef]
  139. Ross, G.W.; Petrovitch, H.; Abbott, R.D.; Tanner, C.M.; Popper, J.; Masaki, K.; Launer, L.; White, L.R. Association of olfactory dysfunction with risk for future Parkinson’s disease. Ann. Neurol. 2008, 63, 167–173. [Google Scholar] [CrossRef]
  140. Dickson, D.W.; Fujishiro, H.; DelleDonne, A.; Menke, J.; Ahmed, Z.; Klos, K.J.; Josephs, K.A.; Frigerio, R.; Burnett, M.; Parisi, J.E.; et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008, 115, 437–444. [Google Scholar] [CrossRef] [PubMed]
  141. Noorian, A.R.; Rha, J.; Annerino, D.M.; Bernhard, D.; Taylor, G.M.; Greene, J.G. Alpha-synuclein transgenic mice display age-related slowing of gastrointestinal motility associated with transgene expression in the vagal system. Neurobiol. Dis. 2012, 48, 9–19. [Google Scholar] [CrossRef]
  142. Wang, L.; Magen, I.; Yuan, P.; Subramaniam, S.R.; Richter, F.; Chesselet, M.; Taché, Y. Mice overexpressing wild-type human alpha-synuclein display alterations in colonic myenteric ganglia and defecation. Neurogastroenterol. Motil. 2012, 24, e425–e436. [Google Scholar] [CrossRef] [PubMed]
  143. Kelly, L.P.; Carvey, P.M.; Keshavarzian, A.; Shannon, K.M.; Shaikh, M.; Bakay, R.A.E.; Kordower, J.H. Progression of intestinal permeability changes and alpha-synuclein expression in a mouse model of Parkinson’s disease. Mov. Disord. 2014, 29, 999–1009. [Google Scholar] [CrossRef] [PubMed]
  144. Natale, G.; Kastsiushenka, O.; Fulceri, F.; Ruggieri, S.; Paparelli, A.; Fornai, F. MPTP-induced parkinsonism extends to a subclass of TH-positive neurons in the gut. Brain Res. 2010, 1355, 195–206. [Google Scholar] [CrossRef] [PubMed]
  145. Hallett, P.J.; McLean, J.R.; Kartunen, A.; Langston, J.W.; Isacson, O. Alpha-synuclein overexpressing transgenic mice show internal organ pathology and autonomic deficits. Neurobiol. Dis. 2012, 47, 258–267. [Google Scholar] [CrossRef] [PubMed]
  146. Anselmi, L.; Bove, C.; Coleman, F.H.; Le, K.; Subramanian, M.P.; Venkiteswaran, K.; Subramanian, T.; Travagli, R.A. Ingestion of subthreshold doses of environmental toxins induces ascending Parkinsonism in the rat. NPJ Park. Dis 2018, 4, 30. [Google Scholar] [CrossRef] [PubMed]
  147. Donlon, E.; Lynch, R.; Murphy, O.C.; Farrell, M.; Noel, J.; Keogan, M.; O’Connell, M.; Lynch, T. Braak’s Unfinished Hypothesis: A Clinicopathological Case Report of α-Synuclein Peripheral Neuropathy Preceding Parkinsonism by 20 Years. Mov. Disord. Clin. Pract. 2021, 8, 1129–1133. [Google Scholar] [CrossRef] [PubMed]
  148. Harsanyiova, J.; Buday, T.; Trancikova, A.K. Parkinson’s Disease and the Gut: Future Perspectives for Early Diagnosis. Front. Neurosci. 2020, 14, 626. [Google Scholar] [CrossRef] [PubMed]
  149. Chen, M.; Mor, D.E. Gut-to-Brain α-Synuclein Transmission in Parkinson’s Disease: Evidence for Prion-like Mechanisms. Int. J. Mol. Sci. 2023, 24, 7205. [Google Scholar] [CrossRef] [PubMed]
  150. Videlock, E.J.; Xing, T.; Yehya, A.H.S.; Travagli, R.A. Experimental models of gut-first Parkinson’s disease: A systematic review. Neurogastroenterol. Motil. 2023, 35, e14604. [Google Scholar] [CrossRef] [PubMed]
  151. Challis, C.; Hori, A.; Sampson, T.R.; Yoo, B.B.; Challis, R.C.; Hamilton, A.M.; Mazmanian, S.K.; Volpicelli-Daley, L.A.; Gradinaru, V. Gut-seeded α-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat. Neurosci. 2020, 23, 327–336. [Google Scholar] [CrossRef]
  152. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
  153. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant polyphenols: Chemical properties, biological activities, and synthesis. Angew. Chem. Int. Ed. Engl. 2011, 50, 586–621. [Google Scholar] [CrossRef] [PubMed]
  154. Hano, C.; Tungmunnithum, D. Plant Polyphenols, More than Just Simple Natural Antioxidants: Oxidative Stress, Aging and Age-Related Diseases. Medicines 2020, 7, 26. [Google Scholar] [CrossRef] [PubMed]
  155. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef] [PubMed]
  156. Sova, M.; Saso, L. Natural Sources, Pharmacokinetics, Biological Activities and Health Benefits of Hydroxycinnamic Acids and Their Metabolites. Nutrients 2020, 12, 2190. [Google Scholar] [CrossRef]
  157. Aryal, S.; Skinner, T.; Bridges, B.; Weber, J.T. The Pathology of Parkinson’s Disease and Potential Benefit of Dietary Polyphenols. Molecules 2020, 25, 4382. [Google Scholar] [CrossRef] [PubMed]
  158. Martinez, K.B.; Mackert, J.D.; McIntosh, M.K. Polyphenols and Intestinal Health. In Nutritional and Functional Foods for Healthy Aging; Watson, R.R., Ed.; Academic Press: Cambridge, MA, USA, 2017; pp. 191–210. [Google Scholar]
  159. Naczk, M.; Shahidi, F. Phenolics in cereals, fruits and vegetables: Occurrence, extraction and analysis. J. Pharm. Biomed. Anal. 2006, 41, 1523–1542. [Google Scholar] [CrossRef] [PubMed]
  160. Tsao, R.; Deng, Z. Separation procedures for naturally occurring antioxidant phytochemicals. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2004, 812, 85–99. [Google Scholar] [CrossRef] [PubMed]
  161. Liang, T.; Yue, W.; Li, Q. Comparison of the phenolic content and antioxidant activities of Apocynum venetum L. (Luo-Bu-Ma) and two of its alternative species. Int. J. Mol. Sci. 2010, 11, 4452–4464. [Google Scholar] [CrossRef] [PubMed]
  162. PPatil, S.A.; Kandathil, V.; Sobha, A.; Somappa, S.B.; Feldman, M.R.; Bugarin, A.; Patil, S.A. Comprehensive Review on Medicinal Applications of Coumarin-Derived Imine-Metal Complexes. Molecules 2022, 27, 5220. [Google Scholar] [CrossRef]
  163. Sharifi-Rad, J.; Cruz-Martins, N.; López-Jornet, P.; Lopez, E.P.-F.; Harun, N.; Yeskaliyeva, B.; Beyatli, A.; Sytar, O.; Shaheen, S.; Sharopov, F.; et al. Natural Coumarins: Exploring the Pharmacological Complexity and Underlying Molecular Mechanisms. Oxid. Med. Cell. Longev. 2021, 2021, 6492346. [Google Scholar] [CrossRef] [PubMed]
  164. Annunziata, F.; Pinna, C.; Dallavalle, S.; Tamborini, L.; Pinto, A. An Overview of Coumarin as a Versatile and Readily Accessible Scaffold with Broad-Ranging Biological Activities. Int. J. Mol. Sci. 2020, 21, 4618. [Google Scholar] [CrossRef] [PubMed]
  165. Onder, A. Anticancer activity of natural coumarins for biological targets. In Studies in Natural Products Chemistry; Rahman, A.-U., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 85–105. [Google Scholar]
  166. Akkol, E.K.; Genç, Y.; Karpuz, B.; Sobarzo-Sánchez, E.; Capasso, R. Coumarins and Coumarin-Related Compounds in Pharmacotherapy of Cancer. Cancers 2020, 12, 1959. [Google Scholar] [CrossRef] [PubMed]
  167. Abraham, K.; Wöhrlin, F.; Lindtner, O.; Heinemeyer, G.; Lampen, A. Toxicology and risk assessment of coumarin: Focus on human data. Mol. Nutr. Food Res. 2010, 54, 228–239. [Google Scholar] [CrossRef] [PubMed]
  168. De Rosso, M.; Mayr, C.M.; Girardi, G.; Vedova, A.D.; Flamini, R. High-resolution mass spectrometry metabolomics of grape chemical markers to reveal use of not-allowed varieties in the production of Amarone and Recioto wines. Metabolomics 2018, 14, 124. [Google Scholar] [CrossRef] [PubMed]
  169. Shen, T.; Wang, X.-N.; Lou, H.-X. Natural stilbenes: An overview. Nat. Prod. Rep. 2009, 26, 916–935. [Google Scholar] [CrossRef] [PubMed]
  170. Sirerol, J.A.; Rodríguez, M.L.; Mena, S.; Asensi, M.A.; Estrela, J.M.; Ortega, A.L. Role of Natural Stilbenes in the Prevention of Cancer. Oxid. Med. Cell. Longev. 2016, 2016, 3128951. [Google Scholar] [CrossRef]
  171. Li, Y.; Li, S.; Lin, C. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors 2018, 44, 69–82. [Google Scholar] [CrossRef] [PubMed]
  172. Mompeo, O.; Spector, T.D.; Hernandez, M.M.; Le Roy, C.; Istas, G.; Le Sayec, M.; Mangino, M.; Jennings, A.; Rodriguez-Mateos, A.; Valdes, A.M.; et al. Consumption of Stilbenes and Flavonoids is Linked to Reduced Risk of Obesity Independently of Fiber Intake. Nutrients 2020, 12, 1871. [Google Scholar] [CrossRef] [PubMed]
  173. Durazzo, A.; Zaccaria, M.; Polito, A.; Maiani, G.; Carcea, M. Lignan Content in Cereals, Buckwheat and Derived Foods. Foods 2013, 2, 53–63. [Google Scholar] [CrossRef]
  174. Rodriguez-Garcia, C.; Sanchez-Quesada, C.; Toledo, E.; Delgado-Rodriguez, M.; Gaforio, J.J. Naturally Lignan-Rich Foods: A Dietary Tool for Health Promotion? Molecules 2019, 24, 917. [Google Scholar] [CrossRef] [PubMed]
  175. Milder, I.E.J.; Arts, I.C.W.; van de Putte, B.; Venema, D.P.; Hollman, P.C.H. Lignan contents of Dutch plant foods: A database including lariciresinol, pinoresinol, secoisolariciresinol and matairesinol. Br. J. Nutr. 2005, 93, 393–402. [Google Scholar] [CrossRef] [PubMed]
  176. Thompson, L.U.; Boucher, B.A.; Liu, Z.; Cotterchio, M.; Kreiger, N. Phytoestrogen content of foods consumed in Canada, including isoflavones, lignans, and coumestan. Nutr. Cancer 2006, 54, 184–201. [Google Scholar] [CrossRef]
  177. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed]
  178. Li, M.; Xu, Z. Quercetin in a lotus leaves extract may be responsible for antibacterial activity. Arch. Pharm. Res. 2008, 31, 640–644. [Google Scholar] [CrossRef] [PubMed]
  179. Cushnie, T.P.T.; Lamb, A.J. Antimicrobial activity of flavonoids. Int. J. Antimicrob. Agents 2005, 26, 343–356. [Google Scholar] [CrossRef]
  180. Nguyen, W.; Grigori, L.; Just, E.; Santos, C.; Seleem, D. The in vivo anti-Candida albicans activity of flavonoids. J. Oral Biosci. 2021, 63, 120–128. [Google Scholar] [CrossRef] [PubMed]
  181. Kaul, T.N.; Middleton, E., Jr.; Ogra, P.L. Antiviral effect of flavonoids on human viruses. J. Med. Virol. 1985, 15, 71–79. [Google Scholar] [CrossRef]
  182. Badshah, S.L.; Faisal, S.; Muhammad, A.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Antiviral activities of flavonoids. Biomed. Pharmacother. 2021, 140, 111596. [Google Scholar] [CrossRef]
  183. Thompson, L.U.; Yoon, J.H.; Jenkins, D.J.A.; Wolever, T.M.S.; Jenkins, A.L. Relationship between polyphenol intake and blood glucose response of normal and diabetic individuals. Am. J. Clin. Nutr. 1984, 39, 745–751. [Google Scholar] [CrossRef]
  184. Madan, J.; Desai, S.; Moitra, P.; Salis, S.; Agashe, S.; Battalwar, R.; Mehta, A.; Kamble, R.; Kalita, S.; Phatak, A.G.; et al. Effect of Almond Consumption on Metabolic Risk Factors-Glucose Metabolism, Hyperinsulinemia, Selected Markers of Inflammation: A Randomized Controlled Trial in Adolescents and Young Adults. Front. Nutr. 2021, 8, 668622. [Google Scholar] [CrossRef] [PubMed]
  185. Maukonen, J.; Saarela, M. Human gut microbiota: Does diet matter? Proc. Nutr. Soc. 2015, 74, 23–36. [Google Scholar] [CrossRef] [PubMed]
  186. Hurrell, R.F.; Reddy, M.; Cook, J.D. Inhibition of non-haem iron absorption in man by polyphenolic-containing beverages. Br. J. Nutr. 1999, 81, 289–295. [Google Scholar] [CrossRef] [PubMed]
  187. Smith, A.H.; Zoetendal, E.; Mackie, R.I. Bacterial mechanisms to overcome inhibitory effects of dietary tannins. Microb. Ecol. 2005, 50, 197–205. [Google Scholar] [CrossRef] [PubMed]
  188. Ma, Q.; Kim, E.; Lindsay, E.A.; Han, O. Bioactive dietary polyphenols inhibit heme iron absorption in a dose-dependent manner in human intestinal Caco-2 cells. J. Food Sci. 2011, 76, H143–H150. [Google Scholar] [CrossRef]
  189. Duda-Chodak, A.; Tarko, T. Possible Side Effects of Polyphenols and Their Interactions with Medicines. Molecules 2023, 28, 2536. [Google Scholar] [CrossRef] [PubMed]
  190. Kim, E.-Y.; Pai, T.-K.; Han, O. Effect of bioactive dietary polyphenols on zinc transport across the intestinal Caco-2 cell monolayers. J. Agric. Food Chem. 2011, 59, 3606–3612. [Google Scholar] [CrossRef] [PubMed]
  191. Islam, T.; Albracht-Schulte, K.; Ramalingam, L.; Schlabritz-Lutsevich, N.; Park, O.-H.; Zabet-Moghaddam, M.; Kalupahana, N.S.; Moustaid-Moussa, N. Anti-inflammatory mechanisms of polyphenols in adipose tissue: Role of gut microbiota, intestinal barrier integrity and zinc homeostasis. J. Nutr. Biochem. 2023, 115, 109242. [Google Scholar] [CrossRef] [PubMed]
  192. Nouri-Vaskeh, M.; Mahdavi, A.M.; Afshan, H.; Alizadeh, L.; Zarei, M. Effect of curcumin supplementation on disease severity in patients with liver cirrhosis: A randomized controlled trial. Phytother. Res. 2020, 34, 1446–1454. [Google Scholar] [CrossRef]
  193. Mantzorou, M.; Pavlidou, E.; Vasios, G.; Tsagalioti, E.; Giaginis, C. Effects of curcumin consumption on human chronic diseases: A narrative review of the most recent clinical data. Phytother. Res. 2018, 32, 957–975. [Google Scholar] [CrossRef]
  194. Shabbir, U.; Rubab, M.; Daliri, E.B.-M.; Chelliah, R.; Javed, A.; Oh, D.-H. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients 2021, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  195. Perrone, D.; Ardito, F.; Giannatempo, G.; Dioguardi, M.; Troiano, G.; Russo, L.L.; DE Lillo, A.; Laino, L.; Muzio, L.L. Biological and therapeutic activities, and anticancer properties of curcumin. Exp. Ther. Med. 2015, 10, 1615–1623. [Google Scholar] [CrossRef] [PubMed]
  196. Siviero, A.; Gallo, E.; Maggini, V.; Gori, L.; Mugelli, A.; Firenzuoli, F.; Vannacci, A. Curcumin, a golden spice with a low bioavailability. J. Herb. Med. 2015, 5, 57–70. [Google Scholar] [CrossRef]
  197. Cui, C.; Han, Y.; Li, H.; Yu, H.; Zhang, B.; Li, G. Curcumin-driven reprogramming of the gut microbiota and metabolome ameliorates motor deficits and neuroinflammation in a mouse model of Parkinson’s disease. Front. Cell. Infect. Microbiol. 2022, 12, 887407. [Google Scholar] [CrossRef]
  198. Chen, J.; Tang, X.Q.; Zhi, J.L.; Cui, Y.; Yu, H.M.; Tang, E.H.; Sun, S.N.; Feng, J.Q.; Chen, P.X. Curcumin protects PC12 cells against 1-methyl-4-phenylpyridinium ion-induced apoptosis by bcl-2-mitochondria-ROS-iNOS pathway. Apoptosis 2006, 11, 943–953. [Google Scholar] [CrossRef] [PubMed]
  199. Lee, H.S.; Jung, K.K.; Cho, J.Y.; Rhee, M.H.; Hong, S.; Kwon, M.; Kim, S.H.; Kang, S.Y. Neuroprotective effect of curcumin is mainly mediated by blockade of microglial cell activation. Pharmazie 2007, 62, 937–942. [Google Scholar] [PubMed]
  200. Jiang, T.-F.; Zhang, Y.-J.; Zhou, H.-Y.; Wang, H.-M.; Tian, L.-P.; Liu, J.; Ding, J.-Q.; Chen, S.-D. Curcumin ameliorates the neurodegenerative pathology in A53T alpha-synuclein cell model of Parkinson’s disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy. J. Neuroimmune Pharmacol. 2013, 8, 356–369. [Google Scholar] [CrossRef] [PubMed]
  201. Shen, L.; Liu, L.; Ji, H.-F. Regulative effects of curcumin spice administration on gut microbiota and its pharmacological implications. Food Nutr. Res. 2017, 61, 1361780. [Google Scholar] [CrossRef] [PubMed]
  202. Mcfadden, R.M.T.; Larmonier, C.B.; Shehab, K.W.; Midura-Kiela, M.; Ramalingam, R.; Harrison, C.A.; Besselsen, D.G.; Chase, J.H.; Caporaso, J.G.; Jobin, C.; et al. The Role of Curcumin in Modulating Colonic Microbiota During Colitis and Colon Cancer Prevention. Inflamm. Bowel Dis. 2015, 21, 2483–2494. [Google Scholar] [CrossRef]
  203. Zam, W. Gut Microbiota as a Prospective Therapeutic Target for Curcumin: A Review of Mutual Influence. J. Nutr. Metab. 2018, 2018, 1367984. [Google Scholar] [CrossRef]
  204. Yuan, T.; Yin, Z.; Yan, Z.; Hao, Q.; Zeng, J.; Li, L.; Zhao, J. Tetrahydrocurcumin ameliorates diabetes profiles of db/db mice by altering the composition of gut microbiota and up-regulating the expression of GLP-1 in the pancreas. Fitoterapia 2020, 146, 104665. [Google Scholar] [CrossRef]
  205. Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef] [PubMed]
  206. Hidalgo, M.; Oruna-Concha, M.J.; Kolida, S.; Walton, G.E.; Kallithraka, S.; Spencer, J.P.E.; Gibson, G.R.; de Pascual-Teresa, S. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J. Agric. Food Chem. 2012, 60, 3882–3890. [Google Scholar] [CrossRef]
  207. Queipo-Ortuño, M.I.; Boto-Ordóñez, M.; Murri, M.; Gomez-Zumaquero, J.M.; Clemente-Postigo, M.; Estruch, R.; Cardona Diaz, F.; Andrés-Lacueva, C.; Tinahones, F.J. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am. J. Clin. Nutr. 2012, 95, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
  208. Xing, L.; Zhang, H.; Qi, R.; Tsao, R.; Mine, Y. Recent Advances in the Understanding of the Health Benefits and Molecular Mechanisms Associated with Green Tea Polyphenols. J. Agric. Food Chem. 2019, 67, 1029–1043. [Google Scholar] [CrossRef]
  209. Caruana, M.; Vassallo, N. Tea Polyphenols in Parkinson’s Disease. Adv. Exp. Med. Biol. 2015, 863, 117–137. [Google Scholar]
  210. Pérez-Burillo, S.; Navajas-Porras, B.; López-Maldonado, A.; Hinojosa-Nogueira, D.; Pastoriza, S.; Rufián-Henares, J.A. Green Tea and Its Relation to Human Gut Microbiome. Molecules 2021, 26, 3907. [Google Scholar] [CrossRef]
  211. Clifford, M.N. Diet-derived phenols in plasma and tissues and their implications for health. Planta Med. 2004, 70, 1103–1114. [Google Scholar] [CrossRef]
  212. Stangl, V.; Lorenz, M.; Stangl, K. The role of tea and tea flavonoids in cardiovascular health. Mol. Nutr. Food Res. 2006, 50, 218–228. [Google Scholar] [CrossRef] [PubMed]
  213. Shan, Z.; Nisar, M.F.; Li, M.; Zhang, C.; Wan, C.C. Theaflavin Chemistry and Its Health Benefits. Oxid. Med. Cell. Longev. 2021, 2021, 6256618. [Google Scholar] [CrossRef]
  214. Fatima, M.; Kesharwani, R.K.; Misra, K.; Rizvi, S.I. Protective effect of theaflavin on erythrocytes subjected to in vitro oxidative stress. Biochem. Res. Int. 2013, 2013, 649759. [Google Scholar] [CrossRef] [PubMed]
  215. Imran, A.; Butt, M.S.; Arshad, M.S.; Arshad, M.U.; Saeed, F.; Sohaib, M.; Munir, R. Exploring the potential of black tea based flavonoids against hyperlipidemia related disorders. Lipids Health Dis. 2018, 17, 57. [Google Scholar] [CrossRef] [PubMed]
  216. Tanaka, T.; Mine, C.; Inoue, K.; Matsuda, M.; Kouno, I. Synthesis of theaflavin from epicatechin and epigallocatechin by plant homogenates and role of epicatechin quinone in the synthesis and degradation of theaflavin. J. Agric. Food Chem. 2002, 50, 2142–2148. [Google Scholar] [CrossRef] [PubMed]
  217. Ma, H.; Hu, Y.; Zhang, B.; Shao, Z.; Roura, E.; Wang, S. Tea polyphenol-gut microbiota interactions: Hints on improving the metabolic syndrome in a multi-element and multi-target manner. Food Sci. Hum. Wellness 2022, 11, 11–21. [Google Scholar] [CrossRef]
  218. Shi, M.; Lu, Y.; Wu, J.; Zheng, Z.; Lv, C.; Ye, J.; Qin, S.; Zeng, C. Beneficial Effects of Theaflavins on Metabolic Syndrome: From Molecular Evidence to Gut Microbiome. Int. J. Mol. Sci. 2022, 23, 7595. [Google Scholar] [CrossRef] [PubMed]
  219. Liu, Z.; de Bruijn, W.J.C.; Bruins, M.E.; Vincken, J.P. Microbial Metabolism of Theaflavin-3,3′-digallate and Its Gut Microbiota Composition Modulatory Effects. J. Agric. Food Chem. 2021, 69, 232–245. [Google Scholar] [CrossRef] [PubMed]
  220. Yussof, A.; Cammalleri, B.; Fayemiwo, O.; Lopez, S.; Chu, T. Antibacterial and Sporicidal Activity Evaluation of Theaflavin-3,3′-digallate. Int. J. Mol. Sci. 2022, 23, 2153. [Google Scholar] [CrossRef] [PubMed]
  221. Liu, W.; Li, J. Theaflavin-3,3′-Digallate Attenuates Rheumatoid Inflammation in Mice through the Nuclear Factor-kappaB and MAPK Pathways. Arch. Immunol. Ther. Exp. 2019, 67, 153–160. [Google Scholar] [CrossRef] [PubMed]
  222. Chaturvedi, R.; Shukla, S.; Seth, K.; Chauhan, S.; Sinha, C.; Shukla, Y.; Agrawal, A. Neuroprotective and neurorescue effect of black tea extract in 6-hydroxydopamine-lesioned rat model of Parkinson’s disease. Neurobiol. Dis. 2006, 22, 421–434. [Google Scholar] [CrossRef]
  223. Gosslau, A.; Jao, D.L.E.; Huang, M.; Ho, C.; Evans, D.; Rawson, N.E.; Chen, K.Y. Effects of the black tea polyphenol theaflavin-2 on apoptotic and inflammatory pathways in vitro and in vivo. Mol. Nutr. Food Res. 2011, 55, 198–208. [Google Scholar] [CrossRef]
  224. Trautwein, E.A.; Du, Y.; Meynen, E.; Yan, X.; Wen, Y.; Wang, H.; Molhuizen, H.O.F. Purified black tea theaflavins and theaflavins/catechin supplements did not affect serum lipids in healthy individuals with mildly to moderately elevated cholesterol concentrations. Eur. J. Nutr. 2010, 49, 27–35. [Google Scholar] [CrossRef] [PubMed]
  225. Choi, J.-S.; Choi, Y.-J.; Shin, S.-Y.; Li, J.; Kang, S.-W.; Bae, J.-Y.; Kim, D.S.; Ji, G.-E.; Kang, J.-S.; Kang, Y.-H. Dietary flavonoids differentially reduce oxidized LDL-induced apoptosis in human endothelial cells: Role of MAPK- and JAK/STAT-signaling. J. Nutr. 2008, 138, 983–990. [Google Scholar] [CrossRef] [PubMed]
  226. Banerjee, S.; Maity, P.; Mukherjee, S.; Sil, A.K.; Panda, K.; Chattopadhyay, D.; Chatterjee, I.B. Black tea prevents cigarette smoke-induced apoptosis and lung damage. J. Inflamm. 2007, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  227. Chen, H.; Parks, T.A.; Chen, X.; Gillitt, N.D.; Jobin, C.; Sang, S. Structural identification of mouse fecal metabolites of theaflavin 3,3′-digallate using liquid chromatography tandem mass spectrometry. J. Chromatogr. A 2011, 1218, 7297–7306. [Google Scholar] [CrossRef] [PubMed]
  228. Yang, C.S. Tea and health. Nutrition 1999, 15, 946–949. [Google Scholar]
  229. Cabrera, C.; Artacho, R.; Gimenez, R. Beneficial effects of green tea—A review. J. Am. Coll. Nutr. 2006, 25, 79–99. [Google Scholar] [CrossRef] [PubMed]
  230. Yang, C.S.; Wang, X.; Lu, G.; Picinich, S.C. Cancer prevention by tea: Animal studies, molecular mechanisms and human relevance. Nat. Rev. Cancer 2009, 9, 429–439. [Google Scholar] [CrossRef]
  231. Chen, S.-Q.; Wang, Z.-S.; Ma, Y.-X.; Zhang, W.; Lu, J.-L.; Liang, Y.-R.; Zheng, X.-Q. Neuroprotective Effects and Mechanisms of Tea Bioactive Components in Neurodegenerative Diseases. Molecules 2018, 23, 512. [Google Scholar] [CrossRef] [PubMed]
  232. Quintana, J.L.B.; Allam, M.F.; Del Castillo, A.S.; Navajas, R.F.-C. Parkinson’s disease and tea: A quantitative review. J. Am. Coll. Nutr. 2009, 28, 1–6. [Google Scholar] [CrossRef]
  233. Malar, D.S.; Prasanth, M.I.; Brimson, J.M.; Sharika, R.; Sivamaruthi, B.S.; Chaiyasut, C.; Tencomnao, T. Neuroprotective Properties of Green Tea (Camellia sinensis) in Parkinson’s Disease: A Review. Molecules 2020, 25, 3926. [Google Scholar] [CrossRef]
  234. Wang, J.; Tang, L.; Zhou, H.; Zhou, J.; Glenn, T.C.; Shen, C.-L.; Wang, J.-S. Long-term treatment with green tea polyphenols modifies the gut microbiome of female sprague-dawley rats. J. Nutr. Biochem. 2018, 56, 55–64. [Google Scholar] [CrossRef]
  235. Levites, Y.; Weinreb, O.; Maor, G.; Youdim, M.B.; Mandel, S. Green tea polyphenol (−)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neurochem. 2001, 78, 1073–1082. [Google Scholar] [CrossRef]
  236. Dauer, W.; Przedborski, S. Parkinson’s disease: Mechanisms and models. Neuron 2003, 39, 889–909. [Google Scholar] [CrossRef]
  237. Carregosa, D.; Carecho, R.M.; Figueira, I.; Santos, C.N. Low-Molecular Weight Metabolites from Polyphenols as Effectors for Attenuating Neuroinflammation. J. Agric. Food Chem. 2020, 68, 1790–1807. [Google Scholar] [CrossRef]
  238. Pervin, M.; Unno, K.; Takagaki, A.; Isemura, M.; Nakamura, Y. Function of Green Tea Catechins in the Brain: Epigallocatechin Gallate and its Metabolites. Int. J. Mol. Sci. 2019, 20, 3630. [Google Scholar] [CrossRef]
  239. Guo, T.; Song, D.; Cheng, L.; Zhang, X. Interactions of tea catechins with intestinal microbiota and their implication for human health. Food Sci. Biotechnol. 2019, 28, 1617–1625. [Google Scholar] [CrossRef]
  240. Zhou, J.; Tang, L.; Shen, C.-L.; Wang, J.-S. Green tea polyphenols modify gut-microbiota dependent metabolisms of energy, bile constituents and micronutrients in female Sprague-Dawley rats. J. Nutr. Biochem. 2018, 61, 68–81. [Google Scholar] [CrossRef]
  241. Zhou, J.; Tang, L.; Shen, C.-L.; Wang, J.-S. Green tea polyphenols boost gut-microbiota-dependent mitochondrial TCA and urea cycles in Sprague-Dawley rats. J. Nutr. Biochem. 2020, 81, 108395. [Google Scholar] [CrossRef]
  242. Pan, T.; Fei, J.; Zhou, X.; Jankovic, J.; Le, W. Effects of green tea polyphenols on dopamine uptake and on MPP+-induced dopamine neuron injury. Life Sci. 2003, 72, 1073–1083. [Google Scholar] [CrossRef]
  243. Liu, Y.-C.; Li, X.-Y.; Shen, L. Modulation effect of tea consumption on gut microbiota. Appl. Microbiol. Biotechnol. 2020, 104, 981–987. [Google Scholar] [CrossRef]
  244. Liu, Z.; Bruins, M.E.; Ni, L.; Vincken, J.-P. Green and Black Tea Phenolics: Bioavailability, Transformation by Colonic Microbiota, and Modulation of Colonic Microbiota. J. Agric. Food Chem. 2018, 66, 8469–8477. [Google Scholar] [CrossRef]
  245. Yoda, Y.; Hu, Z.-Q.; Shimamura, T.; Zhao, W.-H. Different susceptibilities of Staphylococcus and Gram-negative rods to epigallocatechin gallate. J. Infect. Chemother. 2004, 10, 55–58. [Google Scholar] [CrossRef]
  246. Zhang, X.; Zhu, X.L.; Sun, Y.K.; Hu, B.; Sun, Y.; Jabbar, S.; Zeng, X.X. Fermentation in vitro of EGCG, GCG and EGCG3″ Me isolated from Oolong tea by human intestinal microbiota. Food Res. Int. 2013, 54, 1589–1595. [Google Scholar] [CrossRef]
  247. Teil, M.; Arotcarena, M.-L.; Faggiani, E.; Laferriere, F.; Bezard, E.; Dehay, B. Targeting alpha-synuclein for PD Therapeutics: A Pursuit on All Fronts. Biomolecules 2020, 10, 391. [Google Scholar] [CrossRef]
  248. Kalsoom, I.; Wang, Y.; Li, B.; Wen, H. Research Progress of alpha-Synuclein Aggregation Inhibitors for Potential Parkinson’s Disease Treatment. Mini Rev. Med. Chem. 2023, 23, 1959–1974. [Google Scholar] [CrossRef]
  249. Yamasaki, T.R.; Ono, K.; Ho, L.; Pasinetti, G.M. Gut Microbiome-Modified Polyphenolic Compounds Inhibit alpha-Synuclein Seeding and Spreading in alpha-Synucleinopathies. Front. Neurosci. 2020, 14, 398. [Google Scholar] [CrossRef]
  250. Caruana, M.; Neuner, J.; Högen, T.; Schmidt, F.; Kamp, F.; Scerri, C.; Giese, A.; Vassallo, N. Polyphenolic compounds are novel protective agents against lipid membrane damage by alpha-synuclein aggregates in vitro. Biochim. Biophys. Acta 2012, 1818, 2502–2510. [Google Scholar] [CrossRef]
  251. Caruana, M.; Högen, T.; Levin, J.; Hillmer, A.; Giese, A.; Vassallo, N. Inhibition and disaggregation of alpha-synuclein oligomers by natural polyphenolic compounds. FEBS Lett. 2011, 585, 1113–1120. [Google Scholar] [CrossRef]
  252. Ho, L.; Zhao, D.; Ono, K.; Ruan, K.; Mogno, I.; Tsuji, M.; Carry, E.; Brathwaite, J.; Sims, S.; Frolinger, T.; et al. Heterogeneity in gut microbiota drive polyphenol metabolism that influences alpha-synuclein misfolding and toxicity. J. Nutr. Biochem. 2019, 64, 170–181. [Google Scholar] [CrossRef]
  253. Ehrnhoefer, D.E.; Bieschke, J.; Boeddrich, A.; Herbst, M.; Masino, L.; Lurz, R.; Engemann, S.; Pastore, A.; Wanker, E.E. EGCG redirects amyloidogenic polypeptides into unstructured, off-pathway oligomers. Nat. Struct. Mol. Biol. 2008, 15, 558–566. [Google Scholar] [CrossRef]
  254. Gonçalves, P.B.; Sodero, A.C.R.; Cordeiro, Y. Green Tea Epigallocatechin-3-gallate (EGCG) Targeting Protein Misfolding in Drug Discovery for Neurodegenerative Diseases. Biomolecules 2021, 11, 767. [Google Scholar] [CrossRef] [PubMed]
  255. Fernandes, L.; Cardim-Pires, T.R.; Foguel, D.; Palhano, F.L. Green Tea Polyphenol Epigallocatechin-Gallate in Amyloid Aggregation and Neurodegenerative Diseases. Front. Neurosci. 2021, 15, 718188. [Google Scholar] [CrossRef] [PubMed]
  256. Wang, Y.; Wu, S.; Li, Q.; Lang, W.; Li, W.; Jiang, X.; Wan, Z.; Chen, J.; Wang, H. Epigallocatechin-3-gallate: A phytochemical as a promising drug candidate for the treatment of Parkinson’s disease. Front. Pharmacol. 2022, 13, 977521. [Google Scholar] [CrossRef] [PubMed]
  257. Bieschke, J.; Russ, J.; Friedrich, R.P.; Ehrnhoefer, D.E.; Wobst, H.; Neugebauer, K.; Wanker, E.E. EGCG remodels mature alpha-synuclein and amyloid-beta fibrils and reduces cellular toxicity. Proc. Natl. Acad. Sci. USA 2010, 107, 7710–7715. [Google Scholar] [CrossRef] [PubMed]
  258. Liu, Y.; Ho, L.H.; Carver, J.A.; Pukala, T.L. Ion Mobility Mass Spectrometry Studies of the Inhibition of Alpha Synuclein Amyloid Fibril Formation by (–)-Epigallocatechin-3-Gallate. Aust. J. Chem. 2011, 64, 36–40. [Google Scholar] [CrossRef]
  259. Oliveri, V. Toward the discovery and development of effective modulators of α-synuclein amyloid aggregation. Eur. J. Med. Chem. 2019, 167, 10–36. [Google Scholar] [CrossRef]
  260. Ehrnhoefer, D.E.; Duennwald, M.; Markovic, P.; Wacker, J.L.; Engemann, S.; Roark, M.; Legleiter, J.; Marsh, J.L.; Thompson, L.M.; Lindquist, S.; et al. Green tea (−)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington’s disease models. Hum. Mol. Genet. 2006, 15, 2743–2751. [Google Scholar] [CrossRef] [PubMed]
  261. Konijnenberg, A.; Ranica, S.; Narkiewicz, J.; Legname, G.; Grandori, R.; Sobott, F.; Natalello, A. Opposite Structural Effects of Epigallocatechin-3-gallate and Dopamine Binding to α-Synuclein. Anal. Chem. 2016, 88, 8468–8475. [Google Scholar] [CrossRef]
  262. Palhano, F.L.; Lee, J.; Grimster, N.P.; Kelly, J.W. Toward the molecular mechanism(s) by which EGCG treatment remodels mature amyloid fibrils. J. Am. Chem. Soc. 2013, 135, 7503–7510. [Google Scholar] [CrossRef]
  263. Lorenzen, N.; Nielsen, S.B.; Yoshimura, Y.; Vad, B.S.; Andersen, C.B.; Betzer, C.; Kaspersen, J.D.; Christiansen, G.; Pedersen, J.S.; Jensen, P.H.; et al. How epigallocatechin gallate can inhibit α-synuclein oligomer toxicity in vitro. J. Biol. Chem. 2014, 289, 21299–21310. [Google Scholar] [CrossRef]
  264. Kelley, M.; Sant’anna, R.; Fernandes, L.; Palhano, F.L. Pentameric Thiophene as a Probe to Monitor EGCG’s Remodeling Activity of Mature Amyloid Fibrils: Overcoming Signal Artifacts of Thioflavin T. ACS Omega 2021, 6, 8700–8705. [Google Scholar] [CrossRef] [PubMed]
  265. Nebrisi, E.E. Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature. Int. J. Mol. Sci. 2021, 22, 11248. [Google Scholar] [CrossRef] [PubMed]
  266. Singh, P.K.; Kotia, V.; Ghosh, D.; Mohite, G.M.; Kumar, A.; Maji, S.K. Curcumin modulates alpha-synuclein aggregation and toxicity. ACS Chem. Neurosci. 2013, 4, 393–407. [Google Scholar] [CrossRef] [PubMed]
  267. Pandey, N.; Strider, J.; Nolan, W.C.; Yan, S.X.; Galvin, J.E. Curcumin inhibits aggregation of alpha-synuclein. Acta Neuropathol. 2008, 115, 479–489. [Google Scholar] [CrossRef] [PubMed]
  268. Gautam, S.; Karmakar, S.; Bose, A.; Chowdhury, P.K. Beta-cyclodextrin and curcumin, a potent cocktail for disaggregating and/or inhibiting amyloids: A case study with alpha-synuclein. Biochemistry 2014, 53, 4081–4083. [Google Scholar] [CrossRef] [PubMed]
  269. Ahsan, N.; Mishra, S.; Jain, M.K.; Surolia, A.; Gupta, S. Curcumin Pyrazole and its derivative (N-(3-Nitrophenylpyrazole) Curcumin inhibit aggregation, disrupt fibrils and modulate toxicity of Wild type and Mutant alpha-Synuclein. Sci. Rep. 2015, 5, 9862. [Google Scholar] [CrossRef] [PubMed]
  270. Ahmad, B.; Lapidus, L.J. Curcumin prevents aggregation in alpha-synuclein by increasing reconfiguration rate. J. Biol. Chem. 2012, 287, 9193–9199. [Google Scholar] [CrossRef] [PubMed]
  271. Xu, B.; Chen, J.; Liu, Y. Curcumin Interacts with alpha-Synuclein Condensates to Inhibit Amyloid Aggregation under Phase Separation. ACS Omega 2022, 7, 30281–30290. [Google Scholar] [CrossRef] [PubMed]
  272. Li, J.-F.; Jiang, Z.-Q.; Cao, S.; Zhang, M.-X.; Wang, L.-H.; Liu, J.; Lu, Y.-H.; Wang, H.-Y.; Hong, X.-J.; Wang, Z.-G.; et al. Curcumin Inhibits alpha-Synuclein Aggregation by Acting on Liquid-Liquid Phase Transition. Foods 2024, 13, 1287. [Google Scholar] [CrossRef]
  273. Kamelabad, M.R.; Sardroodi, J.J.; Ebrahimzadeh, A.R.; Ajamgard, M. Influence of curcumin and rosmarinic acid on disrupting the general properties of Alpha-Synuclein oligomer: Molecular dynamics simulation. J. Mol. Graph. Model. 2021, 107, 107963. [Google Scholar] [CrossRef]
  274. Liu, W.; Zhang, W.; Xing, L.-Z.; Zhao, Y.-D.; Xu, J.; Li, R.-J.; Zhang, Y.-X. 4-Arylidene curcumin derivatives in vitro inhibit alpha-Synuclein aggregation and disaggregate the preformed fibril. Bioorg. Med. Chem. 2023, 96, 117529. [Google Scholar] [CrossRef] [PubMed]
  275. Jha, N.N.; Ghosh, D.; Das, S.; Anoop, A.; Jacob, R.S.; Singh, P.K.; Ayyagari, N.; Namboothiri, I.N.N.; Maji, S.K. Effect of curcumin analogs onα-synuclein aggregation and cytotoxicity. Sci. Rep. 2016, 6, 28511. [Google Scholar] [CrossRef] [PubMed]
  276. Wang, M.S.; Boddapati, S.; Emadi, S.; Sierks, M.R. Curcumin reduces alpha-synuclein induced cytotoxicity in Parkinson’s disease cell model. BMC Neurosci. 2010, 11, 57. [Google Scholar] [CrossRef] [PubMed]
  277. Dehghani, Z.; Meratan, A.A.; Saboury, A.A.; Nemat-Gorgani, M. Alpha-Synuclein fibrillation products trigger the release of hexokinase I from mitochondria: Protection by curcumin, and possible role in pathogenesis of Parkinson’s disease. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183251. [Google Scholar] [CrossRef] [PubMed]
  278. Spinelli, K.J.; Osterberg, V.R.; Meshul, C.K.; Soumyanath, A.; Unni, V.K. Curcumin Treatment Improves Motor Behavior in α-Synuclein Transgenic Mice. PLoS ONE 2015, 10, e0128510. [Google Scholar] [CrossRef] [PubMed]
  279. Pandareesh, M.D.; Mythri, R.; Bharath, M.S. Bioavailability of dietary polyphenols: Factors contributing to their clinical application in CNS diseases. Neurochem. Int. 2015, 89, 198–208. [Google Scholar] [CrossRef] [PubMed]
  280. Pogačnik, L.; Pirc, K.; Palmela, I.; Skrt, M.; Kim, K.S.; Brites, D.; Brito, M.A.; Ulrih, N.P.; Silva, R.F. Potential for brain accessibility and analysis of stability of selected flavonoids in relation to neuroprotection in vitro. Brain Res. 2016, 1651, 17–26. [Google Scholar] [CrossRef] [PubMed]
  281. Scholz, S.; Williamson, G. Interactions affecting the bioavailability of dietary polyphenols in vivo. Int. J. Vitam. Nutr. Res. 2007, 77, 224–235. [Google Scholar] [CrossRef] [PubMed]
  282. Scalbert, A.; Williamson, G. Dietary intake and bioavailability of polyphenols. J. Nutr. 2000, 130 (Suppl. S8), 2073s–2085s. [Google Scholar] [CrossRef]
  283. Zhang, W.; Dong, X.; Huang, R. Antiparkinsonian effects of polyphenols: A narrative review with a focus on the modulation of the gut-brain axis. Pharmacol. Res. 2023, 193, 106787. [Google Scholar] [CrossRef]
  284. Láng, L.; McArthur, S.; Lazar, A.S.; Pourtau, L.; Gaudout, D.; Pontifex, M.G.; Müller, M.; Vauzour, D. Dietary (Poly)phenols and the Gut-Brain Axis in Ageing. Nutrients 2024, 16, 1500. [Google Scholar] [CrossRef] [PubMed]
  285. Li, Y.; Peng, Y.; Shen, Y.; Zhang, Y.; Liu, L.; Yang, X. Dietary polyphenols: Regulate the advanced glycation end products-RAGE axis and the microbiota-gut-brain axis to prevent neurodegenerative diseases. Crit. Rev. Food Sci. Nutr. 2023, 63, 9816–9842. [Google Scholar] [CrossRef] [PubMed]
  286. Ross, F.C.; Mayer, D.E.; Horn, J.; Cryan, J.F.; Del Rio, D.; Randolph, E.; Gill, C.I.R.; Gupta, A.; Ross, R.P.; Stanton, C.; et al. Potential of dietary polyphenols for protection from age-related decline and neurodegeneration: A role for gut microbiota? Nutr. Neurosci. 2024, 1–19. [Google Scholar] [CrossRef] [PubMed]
  287. Galkin, M.; Priss, A.; Kyriukha, Y.; Shvadchak, V. Navigating alpha-Synuclein Aggregation Inhibition: Methods, Mechanisms, and Molecular Targets. Chem. Rec. 2024, 24, e202300282. [Google Scholar] [CrossRef]
  288. Kemperman, R.A.; Bolca, S.; Roger, L.C.; Vaughan, E.E. Novel approaches for analysing gut microbes and dietary polyphenols: Challenges and opportunities. Microbiology 2010, 156 Pt 11, 3224–3231. [Google Scholar] [CrossRef] [PubMed]
  289. Williamson, G.; Manach, C. Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am. J. Clin. Nutr. 2005, 81 (Suppl. S1), 243s–255s. [Google Scholar] [CrossRef] [PubMed]
  290. Hollman, P.C.; Katan, M.B. Absorption, metabolism and health effects of dietary flavonoids in man. Biomed. Pharmacother. 1997, 51, 305–310. [Google Scholar] [CrossRef] [PubMed]
  291. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  292. Chen, J.; Deng, Y.; Long, S.-Y.; Xu, H.-Y.; Zeng, Y.-T.; Peng, T.; Yang, C.-M.; Du, J.; Zhang, X.-Y. Metabolic flux and catabolic kinetics of prebiotic-like dietary polyphenol phlorizin in association with gut microbiota in vitro. Food Chem. 2024, 440, 138240. [Google Scholar] [CrossRef]
  293. Shetty, S.S.; Kumari, N.S. Therapeutic efficacy of gut microbiota-derived polyphenol metabolite Urolithin A. Beni-Suef Univ. J. Basic Appl. Sci. 2024, 13, 31. [Google Scholar] [CrossRef]
  294. Gade, A.; Kumar, M.S. Gut microbial metabolites of dietary polyphenols and their potential role in human health and diseases. J. Physiol. Biochem. 2023, 79, 695–718. [Google Scholar] [CrossRef] [PubMed]
  295. Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef] [PubMed]
  296. Frolinger, T.; Sims, S.; Smith, C.; Wang, J.; Cheng, H.; Faith, J.; Ho, L.; Hao, K.; Pasinetti, G.M. The gut microbiota composition affects dietary polyphenols-mediated cognitive resilience in mice by modulating the bioavailability of phenolic acids. Sci. Rep. 2019, 9, 3546. [Google Scholar] [CrossRef] [PubMed]
  297. Pasinetti, G.M.; Singh, R.; Westfall, S.; Herman, F.; Faith, J.; Ho, L. The Role of the Gut Microbiota in the Metabolism of Polyphenols as Characterized by Gnotobiotic Mice. J. Alzheimer’s Dis. 2018, 63, 409–421. [Google Scholar] [CrossRef] [PubMed]
  298. Yan, L.; Guo, M.-S.; Zhang, Y.; Yu, L.; Wu, J.-M.; Tang, Y.; Ai, W.; Zhu, F.-D.; Law, B.Y.-K.; Chen, Q.; et al. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. Oxid. Med. Cell. Longev. 2022, 2022, 5288698. [Google Scholar] [CrossRef] [PubMed]
  299. Leclerc, M.; Dudonne, S.; Calon, F. Can Natural Products Exert Neuroprotection without Crossing the Blood-Brain Barrier? Int. J. Mol. Sci. 2021, 22, 3356. [Google Scholar] [CrossRef] [PubMed]
  300. Roberts, S.B.; Franceschini, M.A.; Silver, R.E.; Taylor, S.F.; De Sa, A.B.; Có, R.; Sonco, A.; Krauss, A.; Taetzsch, A.; Webb, P.; et al. Effects of food supplementation on cognitive function, cerebral blood flow, and nutritional status in young children at risk of undernutrition: Randomized controlled trial. BMJ 2020, 370, m2397. [Google Scholar] [CrossRef] [PubMed]
  301. Burapan, S.; Kim, M.; Han, J. Curcuminoid Demethylation as an Alternative Metabolism by Human Intestinal Microbiota. J. Agric. Food Chem. 2017, 65, 3305–3310. [Google Scholar] [CrossRef] [PubMed]
  302. Grosso Jasutkar, H.; Oh, S.E.; Mouradian, M.M. Therapeutics in the Pipeline Targeting alpha-Synuclein for Parkinson’s Disease. Pharmacol. Rev. 2022, 74, 207–237. [Google Scholar] [CrossRef]
  303. Koudoufio, M.; Desjardins, Y.; Feldman, F.; Spahis, S.; Delvin, E.; Levy, E. Insight into Polyphenol and Gut Microbiota Crosstalk: Are Their Metabolites the Key to Understand Protective Effects against Metabolic Disorders? Antioxidants 2020, 9, 982. [Google Scholar] [CrossRef]
  304. Reddy, V.P.; Aryal, P.; Robinson, S.; Rafiu, R.; Obrenovich, M.; Perry, G. Polyphenols in Alzheimer’s Disease and in the Gut-Brain Axis. Microorganisms 2020, 8, 199. [Google Scholar] [CrossRef] [PubMed]
  305. Wang, D.; Ho, L.; Faith, J.; Ono, K.; Janle, E.M.; Lachcik, P.J.; Cooper, B.R.; Jannasch, A.H.; D’Arcy, B.R.; Williams, B.A.; et al. Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease β-amyloid oligomerization. Mol. Nutr. Food Res. 2015, 59, 1025–1040. [Google Scholar] [CrossRef] [PubMed]
  306. Ali, Y.O.; Escala, W.; Ruan, K.; Zhai, R.G. Assaying locomotor, learning, and memory deficits in Drosophila models of neurodegeneration. J. Vis. Exp. 2011, 49, e2504. [Google Scholar]
  307. Ascherio, A.; Zhang, S.M.; Hernán, M.A.; Kawachi, I.; Colditz, G.A.; Speizer, F.E.; Willett, W.C. Prospective study of caffeine consumption and risk of Parkinson’s disease in men and women. Ann. Neurol. 2001, 50, 56–63. [Google Scholar] [CrossRef]
Figure 1. Scheme of α-synuclein regions with the number of corresponding residues.
Figure 1. Scheme of α-synuclein regions with the number of corresponding residues.
Nutrients 16 02041 g001
Figure 2. The role of α-synuclein in the pathogenesis of PD. DA: dopaminergic (created with BioRender.com on 15 July 2023).
Figure 2. The role of α-synuclein in the pathogenesis of PD. DA: dopaminergic (created with BioRender.com on 15 July 2023).
Nutrients 16 02041 g002
Figure 3. Classification of major dietary polyphenols (created with https://www.biorender.com/, accessed on 17 October 2023).
Figure 3. Classification of major dietary polyphenols (created with https://www.biorender.com/, accessed on 17 October 2023).
Nutrients 16 02041 g003
Figure 4. The interactions between polyphenols, the gut–brain axis, and α-synuclein (created with https://www.biorender.com, on 21 May 2024).
Figure 4. The interactions between polyphenols, the gut–brain axis, and α-synuclein (created with https://www.biorender.com, on 21 May 2024).
Nutrients 16 02041 g004
Table 1. Altered gut microbes in patients with PD compared to healthy controls.
Table 1. Altered gut microbes in patients with PD compared to healthy controls.
PhylaClassOrderFamilyGenusSpeciesIncreasedDecreased
Bacteroidetes
(Bacteroidota)
Phylum Bacteroidetes 87, 100, and 103
BacteroidiaBacteroidalesBacteroidaceaeBacteroides87 and 104
Bacteroidesfragilis 88
Bacteroidescoprocola 93 and 106
Barnesiellaceae98
OdoribacteraceaeButyricimonas97
Odoribacter97
Porphyromonadaceae 100
Porphyromonas89 and 97
Prevotellaceae 44, 87, 94, 100, and 103
Prevotella89 and 9082 and 97
Prevotellacopri 91
RikenellaceaeAlistipes106
Alistipesshahii91
Tannerellaceae 83 and 97
Parabacteroides83 and 97
Firmicutes
(Bacillota)
Phylum Firmicutes91 and 10368, 102, and 104
ErysipelotrichiaErysipelotrichalesCoprobavillaceaeCoprobacillus83 and 97
ErysipelotrichaceaeEubacteriumbiforme 91
hallii 91
rectale 91
eligens 91
ClostridiaEubacterialesClostridiaceaeButyricicoccus 89
Clostridiumsaccharolyticum 91
coccoides 88
Hungatella83
ChristensenellaceaeChristensenella92, 93, 95, and 96
Christensenellaminuta93
CarabacteraceaeCatabacter83 and 93
Catabacterhongkongensis93
Lachnospiraceae 68, 83, 89, 92, 95, 96, 101, and 102
Agathobacter 89
Blautia 68, 89, and 99
Coprococcus 68 and 83
Fusicatenibacter 89
Lachnospira 89
Roseburia 68, 83, 89, 92, 95, and 96
Oscillopiraceae (Ruminococcaceae) 100
Faecalibacterium 83, 89, 92, 95, and 96
Faecalibacteriumprausnitzii 87
Hydrogenoanaerobacterium83 and 100
Oscillospira68, 93, 95, 96, and 10389
Ruminiclostridium83
Ruminococcus95 and 100
Ruminococcusbromii93
Papillibactercinnamivorans93
BacilliLactobacillalesLactobacillaceae 44, 83, 95, 98, and 10187
Lactobacillus88, 89, 93, 96, 97, and 104
Lactobacillusmucosae93
Enterococcaceae 98 and 10187
Enterococcus97 and 99
StreptococcaceaeStreptococcus99
NegativicutesVeillonellalesVeillonellaceaeVeillonella97
Proteobacteria (Pseudomonadota)DeltaproteobacteriaDesulfovibrionalesDesulfovibrionaceaeBilophilia83, 92, 97, and 103
Bilophilawadsworthia92 and 105
GammaproteobacteriaEnterobacteralesEnterobacteriaceae44, 87, 94, 95, and 101
PasteurellalesPasteurellaceae 10083
Haemophilus 83
Actinobacteria (Actinomycetota)ActinomycetiaBifidobacterialesBifidobacteriaceaeBifidobacterium82, 87, 89, 92, 93, 95, 96, and 102104
CorynebacterialesCorynebacteriaceae 95
Corynebacterium89
CoriobacteriiaCoriobacterialesCoriobacteriaceaeCollinsella92, 95, and 103
EggerthellalesEggerthellaceae83 and 103
VerrucomicrobiaPhylum Verrucomicrobia97, 100, 103, and 105
VerrucomicrobiaeVerrucomicrobialesAkkermansiaceaeAkkermansia68, 90, 92, 95, 96, 97, 100, 103, and 106
Akkermansiamuciniphila87, 91, 105, and 106
SynergistetesSynergistiaSynergistalesSynergistaceae83
DeferribacteresDeferribacteresDeferribacteralesMucispirillaceaeMucispirillum97
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Riegelman, E.; Xue, K.S.; Wang, J.-S.; Tang, L. Gut–Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson’s Disease. Nutrients 2024, 16, 2041. https://doi.org/10.3390/nu16132041

AMA Style

Riegelman E, Xue KS, Wang J-S, Tang L. Gut–Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson’s Disease. Nutrients. 2024; 16(13):2041. https://doi.org/10.3390/nu16132041

Chicago/Turabian Style

Riegelman, Elizabeth, Kathy S. Xue, Jia-Sheng Wang, and Lili Tang. 2024. "Gut–Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson’s Disease" Nutrients 16, no. 13: 2041. https://doi.org/10.3390/nu16132041

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