Next Article in Journal
Antioxidant for Neurological Diseases and Neurotrauma and Bioengineering Approaches
Next Article in Special Issue
Nutraceuticals and Physical Activity as Antidepressants: The Central Role of the Gut Microbiota
Previous Article in Journal
Nitric Oxide as a Remedy against Oxidative Damages in Apple Seeds Undergoing Accelerated Ageing
Previous Article in Special Issue
The Emerging Scenario of the Gut–Brain Axis: The Therapeutic Actions of the New Actor Kefir against Neurodegenerative Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Parkinson’s Disease and the Metal–Microbiome–Gut–Brain Axis: A Systems Toxicology Approach

by
Lady Johanna Forero-Rodríguez
1,2,†,
Jonathan Josephs-Spaulding
2,*,†,
Stefano Flor
2,
Andrés Pinzón
1,‡ and
Christoph Kaleta
2,‡
1
Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia
2
Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors jointly supervised this work.
Antioxidants 2022, 11(1), 71; https://doi.org/10.3390/antiox11010071
Submission received: 4 November 2021 / Revised: 2 December 2021 / Accepted: 17 December 2021 / Published: 28 December 2021

Abstract

:
Parkinson’s Disease (PD) is a neurodegenerative disease, leading to motor and non-motor complications. Autonomic alterations, including gastrointestinal symptoms, precede motor defects and act as early warning signs. Chronic exposure to dietary, environmental heavy metals impacts the gastrointestinal system and host-associated microbiome, eventually affecting the central nervous system. The correlation between dysbiosis and PD suggests a functional and bidirectional communication between the gut and the brain. The bioaccumulation of metals promotes stress mechanisms by increasing reactive oxygen species, likely altering the bidirectional gut–brain link. To better understand the differing molecular mechanisms underlying PD, integrative modeling approaches are necessary to connect multifactorial perturbations in this heterogeneous disorder. By exploring the effects of gut microbiota modulation on dietary heavy metal exposure in relation to PD onset, the modification of the host-associated microbiome to mitigate neurological stress may be a future treatment option against neurodegeneration through bioremediation. The progressive movement towards a systems toxicology framework for precision medicine can uncover molecular mechanisms underlying PD onset such as metal regulation and microbial community interactions by developing predictive models to better understand PD etiology to identify options for novel treatments and beyond. Several methodologies recently addressed the complexity of this interaction from different perspectives; however, to date, a comprehensive review of these approaches is still lacking. Therefore, our main aim through this manuscript is to fill this gap in the scientific literature by reviewing recently published papers to address the surrounding questions regarding the underlying molecular mechanisms between metals, microbiota, and the gut–brain-axis, as well as the regulation of this system to prevent neurodegeneration.

1. Introduction

1.1. What Is Parkinson’s Disease?

Parkinson’s disease (PD) is the second most common neurodegenerative disorder and affects nearly 1% of the population above 60 years of age [1]. PD is characterized by motor and non-motor symptoms, characterized by striatal dopamine depletion and alterations in neurochemicals [2]. Although motor impairment is critical to disease development, non-motor symptoms become evident prior to the emergence of motor dysfunction [3]. PD non-motor symptoms are related to gastrointestinal (GI) dysfunction, followed by motor dysfunction, such as excessive salivation, dysphagia, nausea, and constipation in approximately 30–80% of patients [4,5]. The direct cause of PD onset is unknown and is loosely related to genetic background (5% to 10%); environmental factors in idiopathic PD are postulated to trigger pathogenesis [6,7,8].

1.2. Gut–Brain Axis: Braak’s Hypothesis

The GI microbiome is a dynamic ecosystem comprising microbial communities impacted by individual diet, host-derived metabolites, and environmental exposures. More so, the human microbiome is essential in bidirectional gut–brain communication; brain and intestinal development, function, and homeostasis are likely mediated by the gut microbiome [9,10,11,12,13]. Braak and colleagues developed a hypothetical framework to explain the influence of environmental factors on PD development [14]. For example, invasive microbiota from the gut during dysbiosis may induce a pro-inflammatory environment, thereby promoting PD onset [15,16]. Pathological microbial agents retrogradely promote α-synuclein formation and spread into the brainstem, locus coeruleus complex, substantia nigra, and the cerebral cortex [17,18]. However, some studies suggest that the presence of α-synuclein within the enteric nerves of the gut tissues should not be used to infer PD pathology [19]. Overall, in PD patients, microbiota–gut–brain axis communication is likely impaired, whereas GI dysfunction symptoms are observed in over 80% of PD subjects [5,20]. However, general research is necessary to elucidate communication mechanisms between gut microbiota in relation to the gut–brain axis [12].
With regard to recent observations, there are wider theories to inform our understanding of the role of the intestinal environment as a significant influencer in the gut–brain axis theory. Presently, it is well-described that irritable bowel disease (IBD) and other inflammatory diseases of the GI are closely associated with PD risk [21,22,23,24]. For example, within a Danish IBD cohort (n = 76,477), patients had an 22% increased risk for PD; from a USA IBD cohort (n = 144,018) an increased risk of 28%; patients within a Swedish population-based study (n = 39,000) had an incidence rate of 30%; and a Taiwanese nationwide cohort study (n = 8375) suggested that patients with IBD had a 43% increased incidence of PD onset [21,22,23,24]. However, treatment with anti-tumor necrosis factor therapies (anti-TNF) was demonstrated to substantially mitigate the incidence of PD by reducing systemic inflammation, which drives the tandem pathogenesis of IBD and PD, demonstrating a causal link of the gut–brain axis [22]. Specifically, anti-TNF was shown to reduce the incidence of PD by 78%, as compared to individuals who do not take the therapy [22].
Microbiota are also essential for provoking PD or preventing the further progression of disease. For example, in a pilot study to investigate pathogenic growth in relation to PD, it was found that patients with PD had a 10-times-increased relative risk of being colonized by Heliobacter pylori [25]. In a subsequent study investigating the role of eradicating H. pylori from PD patients, with a positive C-urea breath test used to identify H. pylori, it was found that eliminating this pathogenic bacterium improved host response to levodopa (L-DOPA) and numerous patients’ symptoms (motor movement, non-motor functions, and overall quality of life) that are associated with PD onset [26]. In contrast, evidence exists describing some beneficial bacteria in patients with PD. Comparing metagenomic shotgun sequencing of the gut microbiome between 76 PD patients and 78 healthy controls, the authors identified Akkermansia muciniphila as enriched in PD patients after controlling for medication that was provided to intervene against PD [27]. Generally, this microbe is known to confer several benefits to humans such as inciting longer gastrointestinal passage times [28,29], improving the GI barrier functioning [30], and improving anti-inflammatory status [31].
To date, there are a lack of studies investigating the link between metal-induced oxidative stress upon the gut microbiota and neurodegeneration, independently and as a system [16]. Heavy metals promote oxidative stress, thereby altering gut barrier permeability and, subsequently, inflammation, leading to amplified heavy metal absorption and traffic into the brain [32]. The gut microbiota also affects intestinal metal absorption by modulating the barrier function and bioremediation of heavy metals in the gut [33,34]. Therefore, interventions to mitigate dietary toxic metals likely reduce the inflammatory burden on beneficial gut microbiota and potentially PD onset. For example, numerous probiotic strains from the genera Lactobacillus and Bifidobacterium have known bioremediation effects on heavy metals [35,36]. To better decipher the systems-based link between the metal–microbiome–gut–brain axis, it is important to interpret how microbial dysbiosis, in relation to oxidative stress from metals, leads to a pro-inflammatory environment and how the gut microbiome influences the remediation of xenobiotic metals [37]. Therefore, this review will summarize the current understanding of the metal–microbiome–gut–brain axis and propose future perspectives to mitigate heavy metal toxicity through gut-associated microbiome chelation, specifically, the process in which microbiota remove toxic metals from the body by producing molecules, which reduce toxicity and oxidative stress prior to neurodegenerative disorders [38]. Overall, the presented cumulative review is an attempted combination of recent evidence from the two fields of PD and the metal microbiome to identify both overlaps and gaps; our aim is to provide suggestions through the lens of systems toxicology to merge both fields.

1.3. Neurotoxicity and Parkinson’s Disease

Biological organisms require 13 metals, 9 of which are essential trace metals for creating organic building blocks and regulating homeostatic processes such as catalyzing enzymes for basic metabolic or biochemical processes. Approximately one-third of biological proteins and 40% of enzymes rely on metal ions to function [39]. The brain generally contains the greatest concentrations of metals in the human body, including Fe, Cu, Zn, and Mn [40]. For example, both Zn and Mn are found in the midbrain and are essential for neuronal excitement, synaptic transmission, the creation of new myelin, and regulation of oxidative stress [41,42]. Conversely, when these metals are above normal homeostatic balance, they are associated with PD [43]. Specifically, increased occupational exposure to Fe or Mg was found to double the risk of developing PD [44]. Improper metal binding associated with oxidative stress leads to protein misfolding and aggregation [45]. Metals with a high affinity to sulfhydryl groups, alter dopamine neurotransmission, reduce expression of D2 dopamine receptor sites, and impair proteins essential for maintaining cellular homeostasis [46]. Protein misfolding is the hallmark of numerous neurodegenerative diseases such as Alzheimer’s Disease, Lewy Body Dementia, Huntington’s Disease, and prion diseases, and is similarly underlined with metal stress [47]. The association between metal exposure, the accumulation of misfolded proteins (especially α-synuclein) found in Lewy bodies and PD is not well understood at the mechanistic level and is likely associated with an increase in ROS [45]. Regardless, the potential to control metal exposure and maintain cellular homeostasis in PD may prevent improper neuroprotein folding prior to apoptosis. There is a need to review the literature surrounding the topic of metabolic outcomes directed by heavy metals exposure and oxidative stress prior to neuropathology.

1.3.1. Manganese (Mn)

Mn is a trace metal found throughout various tissues and is essential for enzymatic processes, the biosynthesis of amino acids, protein formation, and carbohydrate metabolism [47,48]. Environmental exposure to Mn primarily occurs by ingesting leafy vegetables, grains, and nuts which enter the circulatory system through passive diffusion [49]. Chronic exposure to Mn is positively correlated with the hallmark characteristics of PD pathology, such as dopaminergic nerve deterioration and impairment of the basal ganglia, which includes the globus pallidus, substantia nigra, subthalamic nucleus and striatum [50]. Different valence states or species of Mn (Mn2+ or Mn3+) play significant roles in Mn neurotoxicity, in which Mn3+ is more toxic than Mn2+ [51]. Typically, Mn is found in portal circulation, 80% of Mn2+ is found to be associated with α-macroglobulin or albumin. Toxic Mn3+ species account for 1% of the total Mn found in the body; Mn3+ associates with transferrin endosomes during tissue circulation and Mn2+ diffuses through tissues, including the kidneys, bones, liver, and the brain [52].
Within the brain, Mn3+ accumulates and enters neurons by binding to transferrin endosomes and localizes within brain tissue [48,53]. Specifically, Divalent Metal Transporter 1 (DMT1) is highly expressed in brain tissue, especially in dopamine-rich areas of the basal ganglia and cortex, which explains Mn association with the brain [54,55]. Mn exposure affects the transcription and translation of α-synuclein and through the activation of MAPK, thus beginning the apoptosis cascade signaling, and results in dopaminergic neuron apoptosis [56,57]. Mn stress leads to the dysregulation of key homeostatic functions, which are essential for degrading proteins (such as autophagy, proteasomes, and endosomal trafficking) [58]. Chronic metabolic dysequilibrium linked to Mn initiates conformational changes leading to the aggregation of α-synuclein, which can become harmful to neuronal cells [59]. This suggests that Mn and other metal ions incite the production of ROS, leading to conformation changes and the aggregation of α-synuclein [53,58]. However, recent observations of human α-synuclein have identified a neuroprotective role in the mitigation of dopaminergic degeneration linked to acute Mn exposures [60]. Conversely, neuroprotective effects are lessened when neurons are chronically exposed to Mn, leading to accelerated oxidation and misfolding of α-synuclein [60].
Interestingly, Mn-induced Parkinsonism is not correlated with a significant loss of dopaminergic neurons in non-human primates [61,62]. This highlights that Mn exposure can be regulated at the environmental level to prevent PD outcomes. While occupational exposure to Mn was significantly mitigated through public health interventions and regulations, there is still a great need for the differentiation between chronic Mn neurotoxicity and PD symptoms [63]. Mn neurotoxicity is regularly confused with idiopathic PD symptoms; while both differ in several ways, they also share several similarities. Generally, the distribution of Mn within the brain elicits symptoms that are similar to PD; however, clinical observations of these findings are not as common as a PD diagnosis [63]. For example, Mn impacts the neurons of both the globus pallidus striatum and substantia nigra pars reticulata, but accumulates in a lesser proportion in the substantia nigra pars compacta [64]. This occurs through the induction of alterations in energy deficits, protein aggregation, ubiquitin-proteasome system dysfunction, and altered mitochondria function, all of which are closely related to oxidative stress [45]. Thus, the neurotoxic effects on the globus pallidus, the substantia nigra pars reticulata, and the striatum, which impact the dopaminergic neurons, are typical hallmarks of PD [65,66]. The dysregulation of dopamine transmission from the substantia nigra to the striatum, a region associated with motor symptoms in Mn neurotoxicity, is known to prevent dopamine release, thereby leading to elicit behavioral responses that are similar to both PD and Mn neurotoxicity [64,66]. Additionally, Mn toxicity impairs cells that normally produce the neurotransmitter γ-aminobutyric acid (GABA); this dysfunction is associated with numerous psychiatric disorders such as major depressive disorder, bipolar disorder, and schizophrenia [67,68]. Therefore, there is a need to understand Mn exposure in relation to PD onset in terms of molecular mechanisms. For example, recently, a transcriptomic study investigated expression signatures of human neuroblastoma cells treated with high and low doses of Mn [69,70], revealing perturbed processes involved with nervous system development (axon and cerebral cortex formation). This was manifested as disturbances in the mitochondria cell cycle, ROS production, and chronic inflammatory signaling, which leads to neural cell apoptosis [70].

1.3.2. (Methyl) Mercury (MeHg; Hg)

Hg is a metal that shifts between organic and inorganic phases, thereby inducing differing modes of toxicity. Hg not only bio-accumulates within plants and animals in ecosystems, but is also transported over long distances as a gas following combustion. Hg vapor is stable in a gaseous state and accounts for 90% of Hg within the environment [71]. Inorganic Hg is not physiologically necessary in humans and is toxic in low concentrations. Hg becomes fat-soluble within the tissues of animals after methylation by microbes to become methylmercury (MeHg); this organic state is readily consumed and bioaccumulates from the bottom of food chains. For example, contaminated fish with a high biomass of fatty acids rapidly accumulate MeHg within tissues, as compared to other fish within the same environment. Furthermore, Hg was found to accumulate within the nervous tissue of deceased individuals with neurodegenerative diseases in comparison to healthy controls [72]. Individuals poisoned by chronic MeHg suffer from numerous symptoms linked to central nervous systems (CNS) impairment, such as visual impairments, lethargy, tremors, lack of coordination, and inability to recall memories [73].
Hg differs from other metals with regard to cellular transport; it does not require human macrophages or other immuno-transport mechanisms, rather MeHg is readily able to penetrate the blood–brain barrier (BBB) [74]. Specifically, Hg binds to sulfhydryl groups with a high affinity in comparison to other metals in the human body. When MeHg binds to cysteine groups through molecular mimicry, it is transported through the BBB and directly interacts with glial and neuronal cells [74]. From there, Hg is uptaken through the nerve endings and accumulates in the CNS ganglia where it cannot be detoxified, leading to systemic effects upon the substantia nigra dopaminergic neurons through disruption of tubulin molecules [75,76,77,78]. Dopaminergic neurons consist of axons that are composed of tubulin molecules; however, in the presence of inorganic Hg, this metal binds to the 14-sulfhydryl groups found within the tubulin of the dopaminergic neurons. Through this mechanism, Hg interacts with α-tubulin and β-tubulin, inhibiting the tubulin structural formation of neurons, thus preventing tubulin from binding to Guanosine-5′-triphosphate (GTP) and halting motor neural functions by blocking neurotransmission [79]. Hg further promotes the formation of amyloid-β proteins, the predecessors for amyloid plaques and neurofibrils in the brain; both are associated with the onset of Alzheimer’s Disease and PD [80,81]. Through these pathomechanisms, MeHg exposure leads to apoptosis and neurotoxicity through dysfunctional cell narrowing, chromatin condensation, the modification of cytochrome C flux, and well-described oxidative stress insults to the mitochondria [82].

1.3.3. Iron (Fe)

Fe is a common dietary metal that is essential in humans, assisting in oxygen transport and mitochondrial respiration [83]. Conversely, the over- or underabundance of free Fe in humans is associated with several neurological pathophysiologies [84]. Low Fe, anemia, or low hemoglobin levels throughout the human course of life were correlated with an increased risk of developing PD [85]. Irregular Fe metabolism in humans leads to a lack or excess of Fe in the brain, causing deleterious effects in the PNS or CNS [86]. Incidentally, the argument that Fe accumulation precedes PD and other neurodegenerative diseases is discussed in detail elsewhere [83,87,88].
Dysfunctional Fe metabolism regulates the uptake, sequestration, accumulation, release, and movement of Fe into the substantia nigra pars compacta; these specific pathophysiological mechanisms of Fe metabolism are associated with PD [89]. Specifically, PD patient ferritin, transferrin, and total serum Fe is lower than in healthy controls, illustrating that Fe metabolism and transport is adverse in PD patients [90]. In healthy patients, Fe provides pigmentation and other essential neurophysiological roles; in PD patients, Fe is routinely released from neuromelanin [91]. Approximately 20% of Fe in the substantia nigra is bound to neuromelanin. Impaired Fe metabolism is assessed by the loss of neuromelanin in dopaminergic neurons of PD patients, as observed by magnetic resonance imaging [92,93]. Overall, adverse Fe metabolism highlights the role of metal exposure and dysregulation in individuals, especially in the case of neurodegeneration and idiopathic PD [94].
Fe accumulates within the gray matter of both the basal ganglia and midbrain; this is positively associated with PD onset [95]. Neuronal Fe accumulates in the substantia nigra pars compacta of PD patients, but not in other regions of the brain in postmortem PD patients [96]. High quantities of Fe and a lack of Cu in the substantia nigra pars compacta are typical features of PD onset [31]. When tracking PD patients over 3 years, a longitudinal clinical study identified that Fe increases in the substantia nigra pars compacta and simultaneously decreases in the white matter [97]. A down regulation of DMT1 perturbs Fe metabolism in the brain; this pathology is typically observed in PD brains postmortem. Irregularities in the regulation of this gene lead to chronic ferritin release and neurotoxicity [98].

2. Microbiome–Gut–Brain Communication in Health and Disease

The GI tract is a highly interactive surface area between host and environment, as a barrier it is pivotal in regulating immune homeostasis [99]. Six major host cell types comprise the GI barrier: enterocytes, entero-endocrine cells, goblet cells, paneth cells, microfold cells, and stem cells, which form a monolayer barrier that restricts microbial transport and metal ion movement by the expression of transporters [100,101]. Approximately 80% of GI barrier cells are enterocytes; thus, epithelial cells are intermediates between host and luminal interactions in the maintenance of homeostatic functions [100,102,103]. The GI has three major roles as a semipermeable barrier: selective absorption of nutrients, regulation of environmental antigens, and the transport of microorganisms [101,104]. First, the mechanical barrier consists of intestinal epithelial cells and capillary endothelial cells connected with tight or adherens junctions and cadherin proteins, which regulate nutrients [99]. Second, the GI barrier is an immune barrier for controlling environmental antigens. The third, which is extensively covered in this review, is the biological barrier colonized by gut microbiota. However, recent studies highlight that heavy metal exposure alters both the native gut microbiota and intestinal physiology [105,106,107]. Specifically, additional damage due to heavy metals disruption of the gut epithelium leads to a heightened inflammatory response, thus disrupting GI tight junctions and promoting systemic inflammation by inciting changes in microbial abundance and microbial-mediated metabolic changes [108].
The gut–brain axis concept is a fundamental factor for interpreting bidirectional communication and the mechanisms that modulate gut–brain homeostasis, especially during oxidative stress [12]. The central nervous system (CNS), autonomic nervous system (ANS) and enteric nervous system (ENS) influence GI smooth muscle mobility, mucus secretion, and blood flux, thereby modifying intestinal microbiota abundance and, in turn, brain homeostasis and function [12,109]. Microbiome–gut–brain communication is carried out through pathways involving nervous, endocrine, and immune signaling mechanisms [109]. The GI tract is controlled by both intrinsic and extrinsic innervation, in which intrinsic innervation is regulated by the ENS. The ENS is innervated with extrinsic ANS (parasympathetic and sympathetic systems) [110]. The alteration of microbial communities at the gut epithelial barrier modulate both inflammatory responses and metabolic pathways [111,112].

2.1. Neuroendocrine and Neuroactive Metabolites Crosstalk

Communication between the brain and gut microbiome is likely modulated through the vagus nerve [113]. The commensal relationship between microbiota and host neurotransmitter synthesis through the enteric nerves is potentially an evolutionary adaptation [114,115]. Various GI microbiota produce metabolites that are identical to the chemical structure of host-derived neuronal metabolites [12]. For example, Bacillus, Escherichia, and Proteus are able to produce the neurotransmitters dopamine and norepinephrine, which are uptaken by the host; molecular signals modulate communication between the microbiota–gut–brain axis and have implications for host physiology [116,117,118,119]. For example, human vagus nerve stimulation is a therapeutic intervention to treat various pathologies, such as refractory depression, IBD, and Crohn’s disease [113,120]. The microbial secondary metabolite indole stimulates the vagus nerve by inducing the expression of c-Fos proteins in the dorsal vagal complex, indicating that vagus nerve activation leads to enhanced anxiety-like behaviors [121]. Within the gut barrier, enteroendocrine cells (EECs) represent 1% of the total epithelial cells and are critical for gut maintenance [12]. Type-L enterochromaffin cells are the most common EECs and directly interact with the intestinal lumen, microbiota, and microbially derived metabolites [122]. EECs produce glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), which stimulate satiation and regulate food intake behaviors [123]. The expansion of these molecules by EECs activate food-derived nutrients, in addition to bacteria-derived metabolites such as short-chain fatty acids (SCFAs) and indole, which likely induce GLP-1 secretion in colonic L-cells [12,124]. Evidence from in vitro models highlighted that the SCFA propionate stimulates the secretion of PYY and GLP-1 through interactions with free fatty acid receptors 2 and 3 [125]. Human-derived organoid cultures identified that bacteria-derived secondary bile acids such as lithocholic acid stimulate GLP-1 release by activating the G-protein-coupled bile acid receptor 1 [126]. Gut microbiota, such as Lactobacillus strains in co-culture with EECs, increase GLP-1 secretion by downregulating the expression of adaptor proteins involved in TLR signaling such as MyD88 and CD14 [127]. This may demonstrate the direct effect of lactic acid bacteria (LAB) on host physiology (Figure 1).
Enterochromaffin cells regulate intestinal peristalsis, enzymatic secretions, inflammatory responses, and are major producers of peripheral serotonin, derived from dietary tryptophan [137,138] (Figure 1). Studies in germ-free mice found that the reduction in serotonin in the blood and colon could be restored through microbial recolonization [139]. For example, several strains of bacteria such as Escherichia coli, Streptococcus thermophilus, Lactococcus lactis subsp. cremoris, Morganella morganii, Lactobacillus plantarum and Hafnia alvei are reported to produce serotonin in vitro, which contributes to host serotonin levels [12,117,118,119,140]. Additionally, gut microbiota regulate colonic serotonin biosynthesis and increase intestinal motility by releasing enteric neural serotonin through the serotonin receptor 4 (5-HT4, localized in presynaptic enteric neurons) to promote maturation of intestinal neural networks and neurogenesis in the ENS for gut–brain crosstalk [141,142]. Blatia spp. appears to modulate host serotonin metabolism by increasing tryptophan hydroxylase 1 expression within the gut. This in turn decreases the relative abundance of microbiota related to intestinal dysmotility, impaired gut barrier function, and psychiatric disorders such as depression or autism [143,144]. The colonization effects derived from Clostridium ramosum were attributed to the capability of inducing serotonin production in enterochromaffin cells [145,146,147]. Clostridium spp. likely contributes to serotonin secretion by increasing SCFAs and secondary bile acids [139]. Clostridium spp. also possess a high 7α-dehydroxylation activity which aids in the production of deoxycholate, thereby promoting serotonin synthesis from enterochromaffin cells [139]. This demonstrates the active role of the gut-associated microbiome in maintaining both epithelial integrity and brain health.

2.2. Immunomodulation and Dysregulation

Disorders of the CNS are often accompanied by intestinal inflammation [148,149]. Gut-associated microbiota modulate host immune function and oxidative stress at the local and systemic level [149,150]. The mucosal immune system is composed of the GI tract, lamina propria dendritic cells, gut-associated lymphoid tissue, LP-lymphocytes, and intraepithelial lymphocytes [102]. Hosts tolerate commensal antigens by constantly recognizing and reacting to microbes to ensure the mutualistic nature of the host–microbial relationship [151,152]. Therefore, the immune system is integrated as a bidirectional signaling hub between the CNS and the gut [153]. Microbiota are essential in lymphoid structure development, epithelial, and innate lymphoid cells function, and T-cell subsets at a systemic level. These cells maintain gut mucosa homeostasis by signaling for various proinflammatory (Th1, Th2 and Th17 cells) and anti-inflammatory Foxp3+ Regulatory T-cells (Tregs)) differentiation or response [149,154]. Tregs cells are concentrated in the colon and are influenced by intestinal microbiota [155]. Commensal microbiota likely influence anti-inflammatory Treg cell generation and immune responses through the production of both butyrate and propionate, thereby limiting oxidative-stress-induced inflammation and promoting gut homeostasis [133,149,156] (Figure 1a and Figure 2a). In murine models, the application of the Bifidobacterium, Lactobacillus, and Clostridium genera increase Foxp3 Tregs, which are associated with the protection and attenuation of allergic responses in order to maintain immune homeostasis [157,158,159,160].
PD is associated with intestinal inflammation, in which 80% of patients are observed to have gut microbiota composition and abundance differences compared to non-PD controls [4,169]. For example, an overgrowth of Enterobacteriaceae, as well as decreases in Prevotellaceae or families associated with SCFAs production, are observed in PD [170,171,172,173,174]. Enterobacteriaceae such as E.coli induce Th17 inflammatory responses through inflammasome mechanisms, leading to the production of IL-1β. The induction of Th1 and Th17 crosses the BBB, stimulating the microglia and, consequently, promoting brain inflammatory reactions [175,176]. Furthermore, as PD is regularly characterized as a lack of dopamine biosynthesis in various neuronal cells, patients are generally treated withL-DOPA to mitigate the effects of PD. Most recently, berberine supplementation was reported to increase dopamine levels in the brains of mice with PD by modulating the gut microbiota, such as Enterococcus spp. [177]. In a study employing metagenomics in PD patients who were not taking L-DOPA, it was found that microbiota composition at the taxonomic level was modified [178]. Specifically, there were predicted functional changes that were associated with intestinal barrier and immune functioning, leading to functional differences in β-glucuronate and tryptophan degradation pathways, which were adverse in those not taking L-DOPA[178].
PD patients display high levels of proinflammatory cytokine expression (TNF-α, IFN-γ, IL-1β and IL-6) and glial activation markers (GFAP and Sox-10) in the colon [179]. TNF-α enters the brain and induces inflammation by activating microglia or astrocytes; oligomeric protofibrils are uptaken and presented to CD4+ by antigen presenting cells [175,180,181] (Figure 2B). Oral administration of Proteus mirabilis promotes α-synuclein aggregation in the gut and SN, impairing the colonic barrier, and contributes TNF-α and TLR4-mediated macrophage activation [182]. Thus, the dysbiosis of the gut microbiota induces PD-related pathological changes by increasing gut permeability, promoting TNF-α transit via blood to the SN, as well as triggering α-synuclein aggregation and dopaminergic neuronal damage [175,182,183].

3. Heavy Metals and the Gut–Brain Axis

Various nutrients, vitamins, and reactive metals are consumed daily by humans through the ingestion of contaminated foods or water [95,184,185,186,187,188,189]. Simultaneously, the influx of global industrial emissions of heavy metals into environments is linked to environmentally derived brain pathologies; however, preliminary environmental exposures likely interact with the gut-associated microbiome before the brain [190,191] (Figure 3). Once metals enter the GI system, the gut microbiome potentially mediates metal toxicity through biochemical reactions of oxidation or reduction. However, heavy metals promote oxidative stress and disrupt healthy microbiomes in humans, thereby inciting dysbiosis [113,192]. When protective commensal microbial communities enter a state of dysbiosis, there is an increase in the toxic effects of heavy metals [26] and long-term oxidative stress insults, which are in turn associated with neurobehavioral and neurological pathologies [193]. Heavy metal toxicants, which are not modulated by the host-associated microbiome, transit to the brain and can lead to a wide-range of deleterious effects, such as neuroinflammation and neurodegeneration [22].
The clinical and epidemiological characterization of PD suggest that environmental risk factors such as heavy metal toxicity, occupational farming, and pesticide exposure are major factors in PD pathophysiology [194,195]. Heavy metals derived from these environmental exposures lead to the production of reactive radicals such as ROS or NOS that are normally observed in PD progression [38,196] (Figure 3). Specifically, the acute exposure to divalent metal ions incites oxidative stress and oligomerization risk for α-synuclein formation in the brain [197]. Furthermore, Mn, Pb, and Cu ions increase α-synuclein fibrils formation, which produces to Lewy body formation, a major pathophysiological characteristic of PD [54,198,199]. Moreover, the chronic exposure to high levels of metals leads to tissue accumulation over time, especially in the brain [38,200]. Due to the vulnerability of the CNS to metals and the lack of a natural means to detoxify, excrete, or eliminate metals, neuronal energy homeostasis and antioxidant balance are perturbed, which leads to neurodegeneration [63,201,202,203]. This is the case for dopaminergic nerves of the nigrostriatal system, which are readily stressed by heavy metal exposure and may be associated with PD onset [204]. Thus, there is a necessity to further investigate the study of the prodromal phase of PD in relation to metal toxicity pathomechanisms to better understand the underlying disease etiology.

3.1. Metal Transport, Movement, and Molecular Mimicry

Environmental exposure to heavy metals alters host metal transporters through molecular mimicry by interacting with nutritional targets, resulting in metal uptake into cells and further accumulation by the host [205]. Uptaken metals incite injury by forming complexes that mimic endogenous molecules such as amino acids or peptides, thereby binding to transporters to reach apical membranes and promote oxidative stress [201,206,207]. For example, within the intestinal lumen, Hg2+ forms ligands with S-thiols to stimulate endogenous proteins through molecular mimicry mechanisms. Likewise, CH3Hg-S g mimics GSH, Cys-S-Hg-S-Cys acts as a mimic of cystine; both Cys-S-Cd-S-Cys and G-S-Cd-S g are structurally similar to cysteine and glutathione disulfide [208]. These molecules are then taken up by amino acid transporters, multidrug resistance-associated proteins, or oligopeptides transporters, and then readily trafficked into the brain [54,201,209].
There are a diversity of transporters for the purpose of shuttling endogenic molecules, these are readily employed by heavy metals to exert toxic effects on humans. The best studied is DMT1 which is expressed in the duodenum, erythrocytes, liver, and kidneys; this membrane protein is essential for up-taking divalent metals cations (Pb2+, Hg2+, Fe2+, Mn2+) and increasing metal accumulation in tissues [32,206]. For example, human colorectal cells with DMT1 expression knocked-down have a decreased uptake of Fe2+, Pb2+, and Cd2+ [203,206,207,210,211,212,213]. Organic anion transporters that are involved in intestinal uptake of CH3Hg+ through Zinc carriers (ZIP8 and ZIP14) interact with both Hg2+ and Mn2+. ZIP8 and ZIP14 work in tandem with the zinc-regulated, zinc transporter 1 to transport Cd2+, a mimic of Zn2+. More so, Cd2+ and Mn2+ are transported through Ca2+ channels and via endocytosis (Figure 4, Table 1) [202,203,207,214,215].
Within intestinal mucosal cells, various metal ions (Zn2+, Cd2+, Hg2+, Pb2+, Fe2+) cross the apical membrane and readily bind with metallothioneins due to the high content of thiol groups [241,242]. Metallothioneins (MT) have various roles in homeostasis such as the storage, scavenging, transport, and detoxification of free metal ions, thus mitigating cellular damage [242]. For example, MT1 and MT2 are overexpressed in the duodenum when Cd accumulates in intestinal cells [104]. However, Cd promotes the release of Fe from MTs, leading to ROS production and oxidative stress [241,243]. Other examples include the L-system (LAT2), MRPs (MRP1,3,4 and 5), metal transport protein 1 (MTP1), ferroportin, and hephaestin, which participate in the absorption of dietary Fe (Table 1) [201].
Metal ions are able to permeate into the blood through the intestinal barrier by utilizing several transporters, such as transferrin [205,244]. For example, Pb interacts with both hemoglobin and delta-aminolevulinic acid dehydratase (ALAD); however, both Mn and Mg are able to bind to albumin or transferrin and are transported in the blood [245,246,247]. Albumin binds with numerous metals such as Cu, Hg, and Zn [248] (Figure 4). Through these mechanisms, metals are transported to various target organs such as the liver, kidneys, and brain, where they accumulate [184,199,249,250,251]. Metals do not bind to all molecules, rather many mimic molecules, this in turn can lead to an alternative function and oxidative stress. For example, Pb inactivates the heme-associated ALAD enzyme by binding to thiol-groups, leading to ROS production [225]. Cd, Fe, Cu and Mn bind with apolipoprotein A-I, suggesting that exposure to Cd results in oxidative stress by disturbing lipid metabolism [252].
Endothelial cells are a major component of the BBB, in addition to astrocytes, pericytes, basement membranes, and various proteins. Brain endothelial cells enable the process of transportation, such as allowing for the entry of nutrients and other metabolites into the brain [253]. Similar to the intestinal barrier, endothelial cells and neuronal cells within the brain express transporters to acquire biological metals. Specifically, Zn, Fe, and Cu are trafficked by major brain transporters and are readily uptaken across the BBB [207]. Fe acts as a cofactor, leading metals to enter the brain and cross through endothelial cells by transferrin receptors (TfR). The Fe2-Tf complex, which binds to the TfR, is invaginated in order to fuse with endosomes. Eventually Fe is released into the cytoplasm with the support of DMT1. Cytoplasmic Fe is then transferred into the mitochondria or stored in cytosolic ferritin [254,255,256].
Mn binds to Tf and is transported by Fe or Zn transporters into the brain leading to metal accumulation. While the uptake mechanisms are not clear, dietary Fe may influence Mn transport through non-competitive mechanisms [257,258]. Additional carriers found in the BBB, such as ZIP8 and ZIP14, located in the apical and basolateral membranes, both support the bidirectional flux of Mn2+, while ZIP1 and ZIP 2 support the uptake of Pb [207,215]. Additionally, MeHg likely binds to sulfhydryl groups and forms a complex with L-cysteine, which enables transport across the membranes through the L-type amino acid transporter 1 (LAT1). LAT-1 is located in endothelial and pericyte cell membranes and recruits MeHg into the brain; the shuttling of MeHg into the brain further induces the dysfunction of astrocytes and pericytes by increasing BBB permeability [222,259]. Specifically, MeHg exposure inhibits aquaporin AQP4 in astrocytes, leading to alterations in water balance and, consequently, contributing to neurodegeneration [222,260]. Similarly, Pb is taken up into astrocytes through voltage-dependent calcium channels; however, recent studies highlighted the role of Connexin 43 (Cx43) in relation to the uptake of heavy metals such as Pb [261]. Cx43 is a gap junction protein expressed in endothelial cells, astrocytes, activated microglia, and neurons. This gap junction transports metals throughout neuronal cells and is an essential mechanism for disseminating the deleterious effects of metals upon brain cells [261]. Cd uptake is related to Fe uptake mechanisms through DMT-1. Alterations in tight-junction proteins such as ZO-1 translate to uncontrollable Cd uptake in the brain, which in turn induces the impairment of neural tissue functioning [249,262].

3.2. Oxidative Stress and Inflammation in the Gut and Brain

ROS (superoxide, hydrogen peroxide, and hydroxyl) produced from metals generate deleterious products which impair key enzymes of the mitochondrial electron transport chain, leading to alterations in protein functions [263,264,265]. For example, the hippocampus, which is associated with memory, is impacted by heavy metals affecting various regions of the brain in PD patients [15]. Metal stress upon the hippocampus is linked to accelerated aging, memory loss, and dementia [266,267]. The hippocampus contains higher than average levels of glutamate and glucocorticoid receptors, which can predispose the hippocampus to metal stress [268].
Heavy metals further induce oxidative stress by generating ROS, which impair tight junctions that regulate barrier and permeability functions of both the GI tract and BBB by modifying phosphorylated junction proteins, thereby inducing chronic inflammation (Table 2) [28,201,269,270,271,272,273]. However, oxidative stress caused by ROS can be counteracted via antioxidants. Across various organisms, glutathione (GSH) mitigates cellular damage by ROS. Specifically, metal ions accelerate hydroxyl radical production through Fenton reactions, leading to GSH reductase inhibition and, eventually, a decrease in GSH concentration [274] (Figure 4 and Figure 5). Specifically, when ROS levels are greater than antioxidant capacity, oxidative stress is generated. Thus, the increase in ROS triggers cell damage through lipid peroxidation, DNA fragmentation, mitochondrial damage, and other cellular alterations that disrupt barrier tight junctions [273].
The human GI tract contains a diversity of metabolites produced by the microbiota. These microbiota metabolites mediate gut–brain communication by transiting to the CNS, thereby modulating oxidative stress and promoting microglia activation [192,295]. Oxidative stress disrupts gut and BBB barriers, leading to α-synuclein misfolding, aggregation, and subsequent neuronal damage in both ENS and CNS [183]. Increased intestinal permeability allows for the translocation of heavy metal ions and bacterial antigens, both are critical in promoting intestinal neuronal oxidative injury. Synthesis of nitric oxide (NO) in the GI tract by inhibitory motor neurons is readily translocated into the brain through the vagus nerve [296]. On the other hand, NO that is produced by the gut microbiota, is scavenged by hemoglobin and is later diffused. This in turn provides free radicals which can interact with α-synuclein, microbes, and immune signaling, eventually leading to a perturbed BBB and neurodegenerative disorders [148].
Microbial dysbiosis of the GI system promotes a pro-inflammatory environment and alters barrier permeability. This factor, in addition to free heavy metal ions which leak from the gut, likely induces downstream oxidative stress in the ENS, leading to α-synuclein misfolding and aggregation [54,297]. Alterations in microbiota stimulate ROS production by activating the cytoplasmic NLRP3-associated inflammasome, regulating the maturation and secretion of pro-inflammatory cytokines, such as IL-1β in epithelial cells, thereby promoting Th17 cell differentiation [156,298]. Gut-mucosa Th17, an inflammatory subset of T-helper cells, are associated with the development of autoimmune disorders, in addition to PD [175,299,300]. Caspase-1-deficient and stressed mice which lack inflammasome activation, elicit a reduction in depressive-like behaviors and have an altered fecal microbiome. The changes in the microbiome are related to beneficial effects, such as rebalancing in gut microbial communities, and the attenuation of inflammation, all highlight that modulation of the gut microbiota via inflammasome signaling likely alters brain functioning [301]. Proinflammatory signaling from LPS and TNF directs nitric oxide synthase (iNOS) upregulation by inducing the oxidation and nitration of the actin cytoskeleton, thereby disrupting the GI mucosal barrier by depleting both occludin and zonula occludens-1 [136,302,303,304]. Due to the fact that the GI submucosal neurons and terminal axons are proximal to the gut lumen, this may lead to the spread of α-synuclein from the ENS to the CNS through the vagus nerve pathway [297]. Furthermore, when dopaminergic nerve cells are exposed to chronic oxidative stress, this insult eventually leads to the characteristic motor symptoms of PD [20,25]. These tandem reactions trigger neuroinflammation and the onset of neurodegeneration [305].
Heavy metal exposure and ROS by-products promote neurotoxic disturbances such as cognitive impairments, learning/memory dysfunctions, movement disorders, loss of language skills, nervousness, and emotional instability [28,204]. Human brains are especially susceptible to fatty acid oxidation due to high rates of oxygen consumption within this organ and ROS stress caused by heavy metals. Specifically, when fatty acids contained in vascular endothelial cells, neurons, and astrocytes are exposed to MeHg, these cells become impaired and cannot uptake antioxidants; this cascade further leads to chronic oxidative stress, inflammation, and endothelial dysfunction [203,306].
Mn exposure has systemic effects on ROS regulation and production, primarily upon enzymes that control both protein folding and transcription [307,308]. A recent RNA-Seq study investigating the acute and chronic effects of Mn on human-derived neuroblastoma cells observed that Mn at any dose incites damages to the nervous system by upregulating oxidative stress, leading to mitochondrial dysfunction and a heightened inflammatory response [68]. Mn prevents neurotoxin clearance by promoting cellular oxidative stress and impairing crosstalk between both neurons and astrocytes by dysregulating calcium signaling, the glutamate–glutamine pathway, and glutamate–GABA cycle [281,295,309,310]. These metabolic disturbances drive oxidative stress, mitochondrial dysfunction by mitigating ATP levels, induce protein misfolding, and eventually, neurotoxicity through neuroinflammation, leading to nigrostriatal cell death [40,281]. Mitochondrial oxidative stress further exacerbates ROS production and drives neuroinflammation in the brain by perturbing muscarinic and dopaminergic receptors [311,312]. Furthermore, the Mn3+ oxidation of dopamine increases the relative concentration of localized, oxidized dopamine, resulting in increased oxidative stress [60,313]. While Mn impacts neurotransmitter uptake and trafficking through neuronal oxidative stress causes impairments in cellular metabolism, this pathomechanism is shared between both Mn neurotoxicity and PD onset; however, there is a lack of mechanistic evidence connecting Mn exposure to PD causation [314,315,316,317,318] (Figure 5A).
Inorganic Hg2+ that is oxidized by ROS has an increased half-life in the brain and binds to the sulfhydryls of thiols, leading to disruptions in protein conformation or enzyme functions [340,341]. By binding to GSH and the cysteine residues of hormones, essential functions such as GSH upregulation, mitochondrial functions, and the inhibition of the NFkB pathway are blocked, thereby leading to neuronal damage [342,343,344,345]. For example, MeHg causes apoptosis within 18 h of exposure by impairing mitochondrial mRNA expression, inciting the mutation of mtDNA, and leading to the excessive production of ROS [346,347]. When heavy metals damage mitochondria, and GSH concentrations are inhibited, Hg is able to concentrate in the midbrain and promote ROS production prior to PD onset by depleting dopamine production of dopaminergic cells [348,349,350]. This mechanism of neurodegeneration occurs due to the fact that Hg has a high affinity for dopamine receptors as compared to other metals, thus preventing GSH from mitigating ROS and leading to the impairment of numerous neuronal cellular processes and, eventually, hallmark neurological pathologies preceding PD [38,351].
Fe exerts neurotoxic effects through the production of ROS and reacts with hydroxyl radicals; the production of hydrogen peroxide interacts with ferroptosis to release Fe ions that oxidize dopamine and induce dopamine catabolism [352,353,354,355]. The tandem increase in both Fe and ROS in the brain of PD patients is associated with substantia nigra damage [356]. This oxidative stress cascade can result in: neural degeneration, the loss of dopaminergic cells and apoptosis; the overall effect of chronic oxidative stress leads to the exacerbation of neuroinflammation in PD patients [319,320,321]. Chronic inflammation perpetuates Fe ion accumulation in the brain and modifies proteins which metabolize Fe; this further drives neuronal apoptosis by inhibiting mitochondrial complex I [322,357,358,359,360]. Over time, the chronic inflammation by free Fe ions creates ROS, increases the presence of both IL-6 and L-Ferritin in the cerebrospinal fluid, and, finally, directs neuronal apoptosis, which is presented as recurrent tremors [84,361].
BBB dysequilibrium allows for the entry of metals by increasing peripheral inflammatory immune cell infiltration into the CNS, further promoting PD development [362]. Microglia are able to sense diverse stimuli which disrupt CNS homeostasis, such as toxicants, neuronal damage, microbial infection, and α-synuclein [326] (Figure 5c). Initial microglial activation allows for leukocytes to enter the BBB, thereby recruiting Th1 and Th17 cells to produce cytokines that secrete inflammatory molecules such as cytokines, chemokines, reactive free radicals, and others (e.g., iNOS, NO, IL-1beta, IL-6, TNF-α, IFN-γ, IL-8, TGF-b, prostaglandins, cyclooxygenases) [159,160,161]. This in turn may worsen neurodegenerative conditions, inducing neuronal inflammation by stimulating the inflammatory M1 phenotype in the microglia, thereby contributing to neuroinflammation and neurodegeneration [327,328,329,330,331,332,333]. Overall, the chronic activation of M1 microglia is implicated as a key component in the development of neurodegenerative diseases [333].
Astrocyte activation through chronic metal exposure promotes the secretion of pro-inflammatory TNF-α, IL-6, and IL-10, which result in BBB tight junction impairment [363,364,365] (Figure 5e). The protective role of astrocytes, which effectively clears α-synuclein and neurotrophic factors, is impaired following metal alterations of glycogen consumption by inducing ROS production, further promoting astrogliosis and dopaminergic neuron degeneration [366,367]. This mechanism is essential in PD onset due to the fact that if α-synuclein is overproduced and taken up by astrocytes, this likely leads to astrogliosis and neurodegeneration [339]. Thus, the overactivation, abnormal influx, and loss of regulatory activities within astrocytes is a significant contributing factor in the progression of immune-mediated PD [338,339].

3.3. Microbes and Heavy Metals in Gut–Brain Bioremediation

Minerals and trace elements (Ca, P, Mg, Fe, Cu and Zn) are essential for intestinal homeostasis and absorption [368]. However, diets contaminated with heavy metals can result in adverse effects through a variety of mechanisms [369]. The transport of nutrients occurs in the small intestines; the contact or accumulation of metal ions within the GI epithelium induces oxidative stress, cellular injury, dysbiosis, and increases the abundance of facultative anaerobes [207,370]. As a consequence, the available epithelial oxygen increases, leading to the depletion of anaerobic SCFA-producing microbiota, thus reducing the production of anti-inflammatory or antioxidant metabolites. In a healthy environment, oxidative stress is mitigated through chelation or the microbial-mediated reduction, uptake, and eventual clearing of heavy metals (Table 3) [99,371].
Chronic exposure to toxic metals, such as Cu, Pb, Zn, and Cd, alter the gut microbiome of individuals who reside in metal-polluted environments by shifting gut-associated microbiota towards a relatively high abundance of Lachnospiraceae spp., Eubacterium eligens, Ruminococcaceae UGG-014, Erysipelotrichaceae UCG-003, and Tyzzerella spp., as well as significantly decreasing Prevotellaceae [399]. Overall, this change in microbial composition by metal exposure incites metabolic changes in gut microbiota, affecting host metabolism. A stable and functional gut-associated microbial community is necessary to remediate xenobiotic metals. Gut microbiome commensals induce numerous factors which protect hosts from metal stress through the expression of endogenous metallothioneins [400], the upregulation of glutathione (GSH) for anti-oxidative activities [401], and other relevant processes [105]. These factors act in orchestration to limit heavy metal uptake to modulate metal-associated pathogenesis. For example, gnotobiotic mice exposed to Cd and Pb via oral ingestion, have a significant 5–30 and 7–40-fold increase in metal accumulation compared to controls in fecal samples, respectively [27]. These results suggest that host-associated microbiota are essential barriers to mitigate and bioremediate heavy metals in the gut and brain.
A broad group of anaerobic digesting microorganisms described as LAB are key colonizers of the human GI tract. LAB generally act as probiotics and human symbionts, which lack deleterious effects on human health. LAB tolerates acids, degrades different carbohydrates, binds metals to decrease intestinal metal absorption, and reduces tissue metal accumulation, thereby mitigating oxidative stress [402,403]. LAB metal bioremediation properties also create a strong competition in human-associated niches as probiotics [106,392,404]. Human gut LAB likely prevents and bioremediate metal toxicity associated with neurodegenerative disease in at-risk populations [405,406]. LAB responds to various dietary heavy metals, and this is indicative of a potential intervention strategy as a neurological probiotic for the clearance of metals (Table 4).
Probiotic Bifidobacterium and Lactobacillus are found in human infants and increase both gut microbiota diversity and healthy immune functioning [415,416]. L. rhamnosus GR-1 mitigates metal translocation by employing mechanisms of heavy metal tolerance by binding, sequestering, and clearing excess metals for the host [417]. Organic acids produced by LAB such as Lactobacillus, chelate toxic metals by decreasing pH and increasing metal solubility, resulting in the formation of metallo-organic molecules [418,419,420]. This occurs through three mechanisms: ion exchange, precipitation through nucleation reactions, and by creating metal nanoparticles in bacteria cell walls [24,421,422,423].
Probiotic LAB uniquely ferments complex fibers to produce SCFAs (acetate, butyrate, and propionate), thereby providing nutritional supplements for colonocytes to support gut barrier maintenance and prevent invasive pathogens [125,424]. Cd-exposed mice are observed to have a decreased abundance and growth of the genera Lactobacillus and Bifidobacterium, resulting in an impaired GI barrier and Cd accumulation [425]. Low butyrate production increases pH in the gut, creating conditions favoring the overgrowth of invasive pathogens [91,107,164]. Thus, decreased SCFA production promotes increased gut permeability, the growth of opportunistic bacteria, increased uptake of metals, and triggers chronic inflammation [425,426]. When SCFAs are limited, both intestinal dysbiosis and chronic inflammation are heightened [104]. The oral administration of L. plantarum is beneficial in the remediation of Cd, Pb and Cu toxicity; this mechanism inhibits metal uptake by intestinal cells and allows for metal excretion in the feces [389,427,428]. L. rhamnosus GR-1 (LGR-1), which is supplemented in yogurt, demonstrated a decrease in the levels of both As and Hg in the blood of pregnant women and children. These findings suggest that various Lactobacillus strains are potential therapeutic candidates in the remediation of heavy metals to prevent diverse diseases, such as neurodegenerative pathologies [429].
LAB strains also synthesize branched-chain amino acids (BCAAs). These amino acids modulate host physiology by increasing mitochondrial biogenesis, leading to increased antioxidant effects against ROS, and are able to penetrate the BBB [430,431,432,433]. Low plasma BCAAs were associated with autism; however, dietary supplementation with BCAAs reverses some neurological phenotypes [434]. With regard to PD, recent studies identified that PD patients generally have reduced concentrations of both BCAAs and aromatic amino acids compared with controls [435,436], while other studies observed increased BCAA concentrations in PD patients as compared to controls [437,438]. These conflicting observations on the effects of BCAAs, which are derived from LAB, elicit the need to further investigate the mechanistic effect of BCAA upon neurodegeneration and PD onset.
LAB supports the production of various gut-associated secondary metabolites, which can potentially influence neuronal health. For example, L. rhamnosus JB-1 was successfully employed as a therapeutic probiotic to reduce stress, anxiety, and depressive behaviors in mice [439]. Furthermore, decreases in microbiome-derived acetate in mice treated with the antibiotic vancomycin demonstrates a reduction in synaptophysin in the hippocampus, leading to impairments in learning and memory [424,440]. Propionate from LAB interacts with FFAR3 in the ANS, PNS, and CNS to induce intestinal gluconeogenesis [424]. Probiotic strains such as Lactobacillus, Bifidobacterium, and Bacteroidetes are also a significant source of bile salt hydrolase; this allows for the deconjugation of bile acids from taurine and glycine [12,441]. Recently, it was hypothesized that bile acids modulate neuronal function through the stimulation of Fibroblast Growth Factor Receptor (FGF)15 secretion by Farnesoid X receptor (FXR) in the GI, silencing hypothalamic Agouti-related protein/neuropeptide Y neurons through FGF receptors [442].
A primary inhibitory neurotransmitter in the brain, GABA is produced by both the host and GI-associated microbiota [443]. Alterations in GABA and glutamate metabolism were identified during Mn accumulation [444]. The neurotoxic effects include effects in the dopaminergic function in the striatal zone on the brain, where high rates of both oxygen consumption and metabolic rates are crucial factors for increasing brain sensitivity to oxidative damage. [444,445]. Additionally, alterations in GABA receptor expression are related to anxiety and depression symptoms, which occur in tandem with IBD [147,446]. Mice supplemented with probiotic L. rhamnosus were found to have decreased anxiety levels in addition to increased gene expression of both GABAAα1 and GABAAα2 in the hippocampus[432]. Histamine, synthesized by both microbiota and epithelial gut cells of the host have essential roles for mediating gut microbiome and host neurocommunication [447,448]. Application of the probiotic L.reuteri activates histamine receptor 2, thereby modulating host mucosal responses to inflammation and highlighting the role of probiotic microbiota in gut-brain communication [449].

4. Systems Toxicology

4.1. Towards Systems Toxicology

In our day-to-day life, humans are exposed to an environmental cocktail of heavy metals through their diet. Conversely, classical toxicity testing and the risk assessment of metals typically investigates a singular exposure. Therefore, the consideration of metal mixtures upon multiple target organ systems is lacking in detail. Specifically, the application of “Omics” methods can be utilized to increase the mechanistic understanding of metal mixtures toxicity upon biological pathways such as oxidative stress, mRNA splicing, and ETC dysfunction in relation to neurodegeneration and PD [259,450]. As extensively discussed in previous sections of this review, heavy metals toxicity influences neurodegeneration via the microbiota through a wide variety of pathomechanisms. The utilization of different “omics” technologies such as transcriptomics and metagenomics, needed to generate large “molecular snapshots” of various conditions underlying neurodegeneration in the gut–brain axis, are essential [451,452]. While these approaches provide the means to obtain sizable quantitative datasets describing a variety of molecular functions that underlie neurodegeneration, a major hurdle still exists: the data-driven integration of multi-omics and microbiome data into a large-scale system toxicology analysis through modeling approaches [453].
Systems biological knowledge bases and models can be readily employed to integrate and interpret these data at a systems level, providing mechanistic hypotheses about the interactions between the large variety of biological components involved. For example, the alterations that metal toxicants induce between different biological processes, such as oxidative stress responses, mRNA splicing, and energy metabolism and their relation to neurodegeneration [450,454] can be characterized at the levels of genes, proteins, and metabolites. Systems biology models can be employed in this case to infer systems-level activity changes to characterize the interplay between metal toxicants, gut bacteria, and human tissues. In the next section, we introduce a metabolic modeling approach which is specifically designed to simulate the metabolic interactions occurring between multiple human organs, including the gut and its bacterial community. We will speculate about how this approach can be employed to study the role of the microbiota in modulating heavy metal uptake and toxicity effects.

4.2. Whole-Body Models Integration with Heavy Metals Bioremediation

Various systems biology tools such as Constraint Based Reconstruction and Analysis (COBRA) [455,456] have proved their worth by deciphering the metabolic activity of single organisms, as well as multi-species metabolic cross-talk. One of the most frequently employed COBRA methods is flux-balance analysis (FBA) [457], which is used to computationally estimate and characterize biochemical reactions activity in reconstructed metabolic networks of the studied organisms under metabolic steady-states. This framework requires the specific definition of a growth environment (e.g., which metabolites are taken up by the modeled organism), which is dependent upon molecules that are known to be available within the environment under investigation.
Over time, these applications have led to the development of whole-body metabolic models (WBM models) [458]. These models represent multiple individual tissues of an organism by their respective metabolic network along with their unique metabolic connections (e.g., via the bloodstream or the lymphatic system). Additionally, previously published WBM models also contain a specific colon luminal compartment with metabolic networks for the gut microbiota [458]. Moreover, depending on the specific questions to be answered, physiological constraints can be integrated into these models to include information such as blood flow and kidney filtration [458]. These applications led to the recapitulation of interorgan interactions such as the lactic acid and alanine/glucose-alanine cycles through simulations by using the WBM approach [458]. Overall, WBM models highlight a promising approach to predict the interactions between multiple organ systems and microbes within specific contexts, which can include toxic exposure as an important factor for the emergence of PD and related diseases.
Additionally, heavy metals have a detrimental effect on the bacterial community [459], which is important for maintaining gut–brain axis homeostasis [12]; microbial communities are, therefore, strong mediators in the pathophysiological balance between health and disease statuses. While the influence of heavy metals on gut-associated microbiota in directing changes in microbial community composition were studied experimentally [107,460], computational studies which propose metabolic interventions to revert those changes are still required. For example, a list of potentially beneficial metabolites could be generated by simulating the effects of several metabolic interventions on the structure of microbiomes altered by heavy metals. This would allow for the screening of specific metabolites that are able to revert a dysbiotic, host-associated microbial composition back towards a healthy state.
In the context of heavy metal toxicity that is associated with neurodegeneration and PD, WBM models could be employed to investigate the extent to which microbial communities could either mitigate or exacerbate brain toxicity effects. It is already known that some specific bacteria or a combination of them, as shown in Table 3, are able to reduce the toxicity of several contaminants; Streptomyces werraensis for example is known to absorb hexavalent chromium [461]. The combined effect of multiple bacterial species and their communication within host tissues could be studied through the WBM models approach. Additionally, as WBM models allow to account for sex-specific differences, it will be possible to investigate whether sex-specific trajectories of heavy metal toxicity on the host microbiome [370,462] are related to differences in host metabolism. Other potential applications of metabolic modeling approaches include the assessment of the mechanistic bases which underlie complex biological relationships, such as the effect of gut microbiome taxonomic changes and the influence of IBD treatments upon PD incidence, such as the case for anti-TNF therapy [22,178]. A list of metabolic-dependent heavy metal uptake and transformations pathways that can be employed by the bacteria to alter heavy metals availability in the surrounding environment is extensively explained in Chandran et al. 2020 [406]. An important example of these transformations is the methylation of heavy metals, which increases their solubility, and thus the potential transport to the brain. Specific models that include heavy metal metabolism need to be established, and such models’ creation and validation is of paramount importance for systems toxicology study and will require a large amount of work. The use of models specifically curated to include this information will allow for the estimation of microbiome-specific metal transformation capabilities. Those values could be compared to investigate whether the activity of heavy metals transformation mechanisms correlate with PD severity. Overall, the integration of a gut-associated microbial compartment can allow for the further investigation of host–gut microbial co-metabolism within a systems toxicology context. To provide some examples of similar concepts on a topic that differ from heavy metals and PD, it could be important to take inspiration from systems biology. For example, results from integrated WBM models illustrated that the presence of specific host microbiota does not only bolster the production of essential neurotransmitters, but also recapitulates gut–liver alcohol exchange and colonocytes uptake of bacterial butyrate [458], the latter being essential in maintaining the homeostasis of colon epithelial cells. Moreover, this framework allowed for the modeling of the competition between bacterial metabolites and drug detoxification in the liver [458]. Overall, these results suggest that important metabolic exchanges between microbiome, gut, and brain can be modeled with this approach and could be specifically tailored to include information about heavy metal trafficking throughout the gut–brain axis.

5. Conclusions: Fermented Functional Probiotics and Oxidative Stress

Industrialization and urbanization contribute to the release of heavy metals in food and water. Contamination with heavy metals is a serious ecological problem for humans and animals. Although metals are biologically important, they are usually required in trace amounts, excessive metal accumulation in various organs induces various detrimental intracellular events (oxidative stress, mitochondrial dysfunction, DNA fragmentation, protein misfolding, endoplasmic reticulum (ER) stress, autophagy dysregulation, and the activation of apoptosis). Metal accumulation in tissues is known to disrupt the homeostatic functioning of biological systems [405,419]. Thus, harmful compounds can cross organismic barriers leading to alterations within microbiome–gut–brain axis communication; chronic metal toxicity promotes oxidative stress of the gut and brain, eventually leading to PD-associated pathologies [27,38].
The gut barrier, as the first line of defense, largely depends on microbiome–host interactions to control the entry of ingested toxicants. Recent studies illustrated the gut microbiome’s role in modifying the bioavailability and toxicity of heavy metals. Specifically, microbial communities were shown to decrease metal accumulation in both the blood and organs [105]. In this regard, the integration of both in vitro or in vivo studies of toxic exposures with computational modeling allows for the study of community composition, microbiome-specific metals transformation capabilities, gut–brain communication and neurotoxicity from a systems toxicology point of view. This is important due to applications of WBM models that include the prediction of interactions between multiple organ systems and microbes. Tailoring WBM models to account for chronic toxic exposures within specific contexts can facilitate the identification of the mechanistic foundation which underlies these interactions. Moreover, the utilization of novel modeling approaches such as WBM models can be employed to screen for metabolites that appear to be important in the gut-associated microbiome structure and improve the bioremediation potential of microbes, allowing for new strategies to decrease metal uptake and accumulation to improve human health. Thus, potential bioprocessing capabilities of LAB can be estimated as probable interventions to alleviate oxidative stress and metal toxicity, which are significant causative agents of neurodegeneration and the onset of PD.

Author Contributions

L.J.F.-R. and J.J.-S. equally drafted and refined the manuscript, tables, and figures. S.F., A.P. and C.K. provided extensive input and materials into the support of this work. A.P. and C.K. both supervised this work. All authors have read and agreed to the published version of the manuscript.

Funding

C.K. acknowledges support by the German Research Foundation within the scope of collaborative research center “Metaorganisms” (DFG support code SFB1182), as well as the Excellence Cluster “Precision medicine in chronic inflammation” (DFG support code EXC2187) and the European Union within the Marie Skłodowska-Curie Action “SmartAge” (Grant agreement N° 859890). S.F. acknowledges funding support from ITN (Marie Skłodowska-Curie Training Network “SmartAge”, EU grant agreement number 859890). L.J.F.-R. received funds from COLCIENCIAS (National PhD scholarship Conv. 647-2015) (https://www.minciencias.gov.co/, accessed on 3 November 2021). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Acknowledgments

The authors are grateful for institutional support during the preparation of this manuscript. The authors are also thankful to both Johannes Delp and Johannes Zimmermann for providing constructive feedback on this manuscript.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Tysnes, O.B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
  2. Qamar, M.A.; Sauerbier, A.; Politis, M.; Carr, H.; Loehrer, P.; Chaudhuri, K.R. Presynaptic dopaminergic terminal imaging and non-motor symptoms assessment of Parkinson’s disease: Evidence for dopaminergic basis? NPJ Parkinsons Dis. 2017, 3, 5. [Google Scholar] [CrossRef]
  3. Pfeiffer, R.F. Non-motor symptoms in Parkinson’s disease. Park. Relat. Disord. 2015, 22, S119–S122. [Google Scholar] [CrossRef]
  4. Brudek, T. Inflammatory Bowel Diseases and Parkinson’s Disease. J. Parkinsons Dis. 2019, 9, S331–S344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609. [Google Scholar] [CrossRef] [PubMed]
  6. Bellou, V.; Belbasis, L.; Tzoulaki, I.; Evangelou, E.; Ioannidis, J.P.A. Environmental risk factors and Parkinson’s disease: An umbrella review of meta-analyses. Parkinsonism Relat. Disord. 2015, 23, 1–9. [Google Scholar] [CrossRef] [Green Version]
  7. Chen, H.; Ritz, B. The Search for Environmental Causes of Parkinson’s Disease: Moving Forward. J. Parkinson Dis. 2018, 8, S9–S17. [Google Scholar] [CrossRef] [Green Version]
  8. Kouli, A.; Torsney, K.M.; Kuan, W.-L. Parkinson’s Disease: Etiology, Neuropathology, and Pathogenesis. In Parkinson’s Disease: Pathogenesis and Clinical Aspects; Exon Publications: Brisbane, Australia, 2018; ISBN 9780994438164. [Google Scholar]
  9. 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] [PubMed] [Green Version]
  10. Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef]
  11. Calvani, R.; Picca, A.; Lo Monaco, M.R.; Landi, F.; Bernabei, R.; Marzetti, E. Of Microbes and Minds: A Narrative Review on the Second Brain Aging. Front. Med. 2018, 5, 53. [Google Scholar] [CrossRef] [PubMed]
  12. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; 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] [PubMed]
  13. Suganya, K.; Koo, B.-S. Gut–Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef] [PubMed]
  14. 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]
  15. 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]
  16. Ghaisas, S.; Maher, J.; Kanthasamy, A. Gut microbiome in health and disease: Linking the microbiome–gut–brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol. Ther. 2015, 158, 52–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. 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] [Green Version]
  18. Oertel, W.H. Recent advances in treating Parkinson’s disease. F1000Research 2017, 6, 260. [Google Scholar] [CrossRef] [Green Version]
  19. Chung, S.J.; Kim, J.; Lee, H.J.; Ryu, H.-S.; Kim, K.; Lee, J.H.; Jung, K.W.; Kim, M.J.; Kim, Y.J.; Yun, S.-C.; et al. Alpha-synuclein in gastric and colonic mucosa in Parkinson’s disease: Limited role as a biomarker. Mov. Disord. 2015, 31, 241–249. [Google Scholar] [CrossRef]
  20. Lebouvier, T.; Chaumette, T.; Paillusson, S.; Duyckaerts, C.; Bruley Des Varannes, S.; Neunlist, M.; Derkinderen, P. The second brain and Parkinson’s disease. Eur. J. Neurosci. 2009, 30, 735–741. [Google Scholar] [CrossRef]
  21. Lin, J.-C.; Lin, C.-S.; Hsu, C.-W.; Lin, C.-L.; Kao, C.-H. Association between Parkinson’s Disease and Inflammatory Bowel Disease: A nationwide Taiwanese retrospective cohort study. Inflamm. Bowel Dis. 2016, 22, 1049–1055. [Google Scholar] [CrossRef] [Green Version]
  22. Peter, I.; Dubinsky, M.; Bressman, S.; Park, A.; Lu, C.; Chen, N.; Wang, A. Anti–Tumor Necrosis Factor Therapy and Incidence of Parkinson Disease Among Patients With Inflammatory Bowel Disease. JAMA Neurol. 2018, 75, 939–946. [Google Scholar] [CrossRef]
  23. Villumsen, M.; Aznar, S.; Pakkenberg, B.; Jess, T.; Brudek, T. Inflammatory bowel disease increases the risk of Parkinson’s disease: A Danish nationwide cohort study 1977–2014. Gut 2018, 68, 18–24. [Google Scholar] [CrossRef] [PubMed]
  24. Weimers, P.; Halfvarson, J.; Sachs, M.C.; Saunders-Pullman, R.; Ludvigsson, J.F.; Peter, I.; Burisch, J.; Olén, O. Inflammatory Bowel Disease and Parkinson’s Disease: A Nationwide Swedish Cohort Study. Inflamm. Bowel Dis. 2018, 25, 111–123. [Google Scholar] [CrossRef]
  25. Blaecher, C.; Smet, A.; Flahou, B.; Pasmans, F.; Ducatelle, R.; Taylor, D.; Weller, C.; Bjarnason, I.; Charlett, A.; Lawson, A.J.; et al. Significantly higher frequency ofHelicobacter suisin patients with idiopathic parkinsonism than in control patients. Aliment. Pharmacol. Ther. 2013, 38, 1347–1353. [Google Scholar] [CrossRef] [Green Version]
  26. Hashim, H.; Azmin, S.; Razlan, H.; Yahya, N.W.; Tan, H.J.; Manaf, M.R.A.; Ibrahim, N.M. Eradication of Helicobacter pylori Infection Improves Levodopa Action, Clinical Symptoms and Quality of Life in Patients with Parkinson’s Disease. PLoS ONE 2014, 9, e112330. [Google Scholar] [CrossRef] [Green Version]
  27. 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. 2017, 33, 88–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Vandeputte, D.; Falony, G.; Vieira-Silva, S.; Tito, R.Y.; Joossens, M.; Raes, J. Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut 2016, 65, 57–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Gobert, A.P.; Sagrestani, G.; Delmas, E.; Wilson, K.T.; Verriere, T.G.; Dapoigny, M.; Del’Homme, C.; Bernalier-Donadille, A. The human intestinal microbiota of constipated-predominant irritable bowel syndrome patients exhibits anti-inflammatory properties. Sci. Rep. 2016, 6, 39399. [Google Scholar] [CrossRef]
  30. Reunanen, J.; Kainulainen, V.; Huuskonen, L.; Ottman, N.; Belzer, C.; Huhtinen, H.; de Vos, W.M.; Satokari, R. Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer. Appl. Environ. Microbiol. 2015, 81, 3655–3662. [Google Scholar] [CrossRef] [Green Version]
  31. Ottman, N.; Reunanen, J.; Meijerink, M.; Pietilä, T.E.; Kainulainen, V.; Klievink, J.; Huuskonen, L.; Aalvink, S.; Skurnik, M.; Boeren, S.; et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017, 12, e0173004. [Google Scholar] [CrossRef] [PubMed]
  32. Rolli-Derkinderen, M.; Leclair-Visonneau, L.; Bourreille, A.; Coron, E.; Neunlist, M.; Derkinderen, P. Is Parkinson’s disease a chronic low-grade inflammatory bowel disease? J. Neurol. 2020, 267, 2207–2213. [Google Scholar] [CrossRef]
  33. Feng, P.; Ye, Z.; Kakade, A.; Virk, A.K.; Li, X.; Liu, P. A Review on Gut Remediation of Selected Environmental Contaminants: Possible Roles of Probiotics and Gut Microbiota. Nutrients 2019, 11, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Mrvčić, J.; Stanzer, D.; Šolić, E.; Stehlik-Tomas, V. Interaction of lactic acid bacteria with metal ions: Opportunities for improving food safety and quality. World J. Microbiol. Biotechnol. 2012, 28, 2771–2782. [Google Scholar] [CrossRef]
  35. Claus, S.P.; Guillou, H.; Ellero-Simatos, S. The gut microbiota: A major player in the toxicity of environmental pollutants? NPJ Biofilms Microbiomes 2016, 2, 16003. [Google Scholar] [CrossRef]
  36. Daisley, B.A.; Monachese, M.; Trinder, M.; Bisanz, J.E.; Chmiel, J.A.; Burton, J.P.; Reid, G. Immobilization of cadmium and lead by Lactobacillus rhamnosus GR-1 mitigates apical-to-basolateral heavy metal translocation in a Caco-2 model of the intestinal epithelium. Gut Microbes 2019, 10, 321–333. [Google Scholar] [CrossRef] [Green Version]
  37. Monroy-Torres, R.; Antonio Hernández-Luna, M.; Sofía Ramírez-Gómez, X.; López-Briones, S. Role of the Microbiome as the First Metal Detoxification Mechanism. In Prebiotics and Probiotics—Potential Benefits in Nutrition and Health; IntechOpen: London, UK, 2020; Available online: https://www.intechopen.com/chapters/69664 (accessed on 3 November 2021). [CrossRef] [Green Version]
  38. Breton, J.; Daniel, C.; Vignal, C.; Body-Malapel, M.; Garat, A.; Plé, C.; Foligne, B. Does oral exposure to cadmium and lead mediate susceptibility to colitis? The dark-and-bright sides of heavy metals in gut ecology. Sci. Rep. 2016, 6, 19200. [Google Scholar] [CrossRef] [Green Version]
  39. Andreini, C.; Bertini, I.; Cavallaro, G.; Holliday, G.L.; Thornton, J.M. Metal ions in biological catalysis: From enzyme databases to general principles. JBIC J. Biol. Inorg. Chem. 2008, 13, 1205–1218. [Google Scholar] [CrossRef] [PubMed]
  40. Genoud, S.; Roberts, B.R.; Gunn, A.P.; Halliday, G.M.; Lewis, S.J.G.; Ball, H.J.; Hare, D.J.; Double, K.L. Subcellular compartmentalisation of copper, iron, manganese, and zinc in the Parkinson’s disease brain. Metallomics 2017, 9, 1447–1455. [Google Scholar] [CrossRef] [Green Version]
  41. Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative Stress: An Essential Factor in the Pathogenesis of Gastrointestinal Mucosal Diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Möller, H.E.; Bossoni, L.; Connor, J.R.; Crichton, R.R.; Does, M.D.; Ward, R.J.; Zecca, L.; Zucca, F.A.; Ronen, I. Iron, Myelin, and the Brain: Neuroimaging Meets Neurobiology. Trends Neurosci. 2019, 42, 384–401. [Google Scholar] [CrossRef]
  43. Barnham, K.J.; Bush, A.I. Metals in Alzheimer’s and Parkinson’s Diseases. Curr. Opin. Chem. Biol. 2008, 12, 222–228. [Google Scholar] [CrossRef]
  44. Hegde, M.L.; Shanmugavelu, P.; Vengamma, B.; Rao, T.S.S.; Menon, R.B.; Rao, R.V.; Rao, K.S.J. Serum trace element levels and the complexity of inter-element relations in patients with Parkinson’s disease. J. Trace Elem. Med. Biol. 2004, 18, 163–171. [Google Scholar] [CrossRef]
  45. Abramov, A.Y.; Potapova, E.V.; Dremin, V.V.; Dunaev, A.V. Interaction of Oxidative Stress and Misfolded Proteins in the Mechanism of Neurodegeneration. Life 2020, 10, 101. [Google Scholar] [CrossRef]
  46. Carmona, A.; Roudeau, S.; Ortega, R. Molecular Mechanisms of Environmental Metal Neurotoxicity: A Focus on the Interactions of Metals with Synapse Structure and Function. Toxics 2021, 9, 198. [Google Scholar] [CrossRef] [PubMed]
  47. Harischandra, D.S.; Ghaisas, S.; Zenitsky, G.; Jin, H.; Kanthasamy, A.; Anantharam, V.; Kanthasamy, A.G. Manganese-Induced Neurotoxicity: New Insights into the Triad of Protein Misfolding, Mitochondrial Impairment, and Neuroinflammation. Front. Neurosci. 2019, 13, 654. [Google Scholar] [CrossRef] [Green Version]
  48. Fitsanakis, V.A.; Piccola, G.; Dos Santos, A.P.M.; Aschner, J.L.; Aschner, M. Putative proteins involved in manganese transport across the blood-brain barrier. Hum. Exp. Toxicol. 2007, 26, 295–302. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, P.; Bornhorst, J.; Aschner, M. Manganese metabolism in humans. Front. Biosci. 2018, 23, 1655–1679. [Google Scholar] [CrossRef] [Green Version]
  50. Bouabid, S.; Delaville, C.; De Deurwaerdère, P.; Lakhdar-Ghazal, N.; Benazzouz, A. Manganese-Induced Atypical Parkinsonism Is Associated with Altered Basal Ganglia Activity and Changes in Tissue Levels of Monoamines in the Rat. PLoS ONE 2014, 9, e98952. [Google Scholar] [CrossRef] [Green Version]
  51. O’Neal, S.L.; Zheng, W. Manganese Toxicity upon Overexposure: A Decade in Review. Curr. Environ. Health Rep. 2015, 2, 315–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Gunter, T.E.; Gerstner, B.; Gunter, K.K.; Malecki, J.; Gelein, R.; Valentine, W.M.; Aschner, M.; Yule, D.I. Manganese transport via the transferrin mechanism. Neurotoxicology 2013, 34, 118–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Martinez-Finley, E.J.; Chakraborty, S.; Fretham, S.J.B.; Aschner, M. Cellular transport and homeostasis of essential and nonessential metals. Metallomics 2012, 4, 593–605. [Google Scholar] [CrossRef] [Green Version]
  54. Ingrassia, R.; Garavaglia, B.; Memo, M. DMT1 Expression and Iron Levels at the Crossroads between Aging and Neurodegeneration. Front. Neurosci. 2019, 13, 575. [Google Scholar] [CrossRef] [Green Version]
  55. Tai, Y.K.; Chew, K.C.M.; Tan, B.W.Q.; Lim, K.-L.; Soong, T.W. Iron mitigates DMT1-mediated manganese cytotoxicity via the ASK1-JNK signaling axis: Implications of iron supplementation for manganese toxicity. Sci. Rep. 2016, 6, 21113. [Google Scholar] [CrossRef] [Green Version]
  56. Li, Y.; Sun, L.; Cai, T.; Zhang, Y.; Lv, S.; Wang, Y.; Ye, L. α-Synuclein overexpression during manganese-induced apoptosis in SH-SY5Y neuroblastoma cells. Brain Res. Bull. 2010, 81, 428–433. [Google Scholar] [CrossRef] [PubMed]
  57. Prabhakaran, K.; Chapman, G.D.; Gunasekar, P.G. α-Synuclein overexpression enhances manganese-induced neurotoxicity through the NF-κB-mediated pathway. Toxicol. Mech. Methods 2011, 21, 435–443. [Google Scholar] [CrossRef] [PubMed]
  58. Moons, R.; Konijnenberg, A.; Mensch, C.; Van Elzen, R.; Johannessen, C.; Maudsley, S.; Lambeir, A.-M.; Sobott, F. Metal ions shape α-synuclein. Sci. Rep. 2020, 10, 16293. [Google Scholar] [CrossRef] [PubMed]
  59. Cai, T.; Yao, T.; Zheng, G.; Chen, Y.; Du, K.; Cao, Y.; Shen, X.; Chen, J.; Luo, W. Manganese induces the overexpression of α-synuclein in PC12 cells via ERK activation. Brain Res. 2010, 1359, 201–207. [Google Scholar] [CrossRef] [PubMed]
  60. Harischandra, D.S.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. α-Synuclein Protects Against Manganese Neurotoxic Insult During the Early Stages of Exposure in a Dopaminergic Cell Model of Parkinson’s Disease. Toxicol. Sci. 2014, 143, 454–468. [Google Scholar] [CrossRef] [Green Version]
  61. Guilarte, T.R.; Burton, N.C.; McGlothan, J.L.; Verina, T.; Zhou, Y.; Alexander, M.; Pham, L.; Griswold, M.; Wong, D.F.; Syversen, T.; et al. Impairment of nigrostriatal dopamine neurotransmission by manganese is mediated by pre-synaptic mechanism(s): Implications to manganese-induced parkinsonism. J. Neurochem. 2008, 107, 1236–1247. [Google Scholar] [CrossRef] [Green Version]
  62. Guilarte, T.R.; Chen, M.-K.; McGlothan, J.L.; Verina, T.; Wong, D.F.; Zhou, Y.; Alexander, M.; Rohde, C.A.; Syversen, T.; DeCamp, E. Nigrostriatal dopamine system dysfunction and subtle motor deficits in manganese-exposed non-human primates. Exp. Neurol. 2006, 202, 381–390. [Google Scholar] [CrossRef]
  63. Ratner, M.H.; Fitzgerald, E. Understanding of the role of manganese in parkinsonism and Parkinson disease. Neurology 2016, 88, 338–339. [Google Scholar] [CrossRef]
  64. Mallet, N.; Delgado, L.; Chazalon, M.; Miguelez, C.; Baufreton, J. Cellular and Synaptic Dysfunctions in Parkinson’s Disease: Stepping out of the Striatum. Cells 2019, 8, 1005. [Google Scholar] [CrossRef] [Green Version]
  65. Aschner, M.; Erikson, K.M.; Hernández, E.H.; Tjalkens, R. Manganese and its role in Parkinson’s disease: From transport to neuropathology. Neuromol. Med. 2009, 11, 252–266. [Google Scholar] [CrossRef] [PubMed]
  66. Roth, J.A.; Li, Z.; Sridhar, S.; Khoshbouei, H. The effect of manganese on dopamine toxicity and dopamine transporter (DAT) in control and DAT transfected HEK cells. Neurotoxicology 2013, 35, 121–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Torrey, E.F.; Barci, B.M.; Webster, M.J.; Bartko, J.J.; Meador-Woodruff, J.H.; Knable, M.B. Neurochemical markers for schizophrenia, bipolar disorder, and major depression in postmortem brains. Biol. Psychiatry 2005, 57, 252–260. [Google Scholar] [CrossRef]
  68. Fitsanakis, V.A.; Au, C.; Erikson, K.M.; Aschner, M. The effects of manganese on glutamate, dopamine and γ-aminobutyric acid regulation. Neurochem. Int. 2006, 48, 426–433. [Google Scholar] [CrossRef]
  69. Gandhi, D.; Sivanesan, S.; Kannan, K. Manganese-Induced Neurotoxicity and Alterations in Gene Expression in Human Neuroblastoma SH-SY5Y Cells. Biol. Trace Elem. Res. 2017, 183, 245–253. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, S.; Chen, L.; Chen, T.; Zhang, Y.; Ma, J.; Ji, H.; Jia, J.; Huang, Y.; Guo, C.; Xiao, Z.; et al. Expression Profiles of mRNAs in Manganese-Induced Acute and Chronic Neurotoxicity by Bioinformatics Analysis of Gene Microarray. SSRN Electron. J. 2020. [Google Scholar] [CrossRef]
  71. Lin, Y.; Vogt, R.; Larssen, T. Environmental mercury in China: A review. Environ. Toxicol. Chem. 2012, 31, 2431–2444. [Google Scholar] [CrossRef] [Green Version]
  72. Fung, Y.K.; Meade, A.G.; Rack, E.P.; Blotcky, A.J. Brain Mercury in Neurodegenerative Disorders. J. Toxicol. Clin. Toxicol. 1997, 35, 49–54. [Google Scholar] [CrossRef] [PubMed]
  73. Finkelman, R.B.; Orem, W.; Castranova, V.; Tatu, C.A.; Belkin, H.E.; Zheng, B.; Lerch, H.E.; Maharaj, S.V.; Bates, A.L. Health impacts of coal and coal use: Possible solutions. Int. J. Coal Geol. 2002, 50, 425–443. [Google Scholar] [CrossRef]
  74. Cariccio, V.L.; Samà, A.; Bramanti, P.; Mazzon, E. Mercury Involvement in Neuronal Damage and in Neurodegenerative Diseases. Biol. Trace Elem. Res. 2018, 187, 341–356. [Google Scholar] [CrossRef]
  75. Arvidson, B. Accumulation of mercury in brainstem nuclei of mice after retrograde axonal transport. Acta Neurol. Scand. 2009, 82, 234–237. [Google Scholar] [CrossRef]
  76. Arvidson, B. A review of axonal transport of metals. Toxicology 1994, 88, 1–14. [Google Scholar] [CrossRef]
  77. Sarafian, T.A.; Bredesen, D.E.; Verity, M.A. Cellular resistance to methylmercury. Neurotoxicology 1996, 17, 27–36. [Google Scholar]
  78. Pendergrass, J.C.; Haley, B.E. Inhibition of brain tubulin-guanosine 5′-triphosphate interactions by mercury: Similarity to observations in Alzheimer’s diseased brain. Met. Ions Boil. Syst. 1997, 34, 461–478. [Google Scholar]
  79. Pendergrass, J.C.; Haley, B.E.; Vimy, M.J.; Winfield, S.A.; Lorscheider, F.L. Mercury vapor inhalation inhibits binding of GTP to tubulin in rat brain: Similarity to a molecular lesion in Alzheimer diseased brain. Neurotoxicology 1997, 18, 315–324. [Google Scholar] [PubMed]
  80. Breydo, L.; Wu, J.W.; Uversky, V.N. α-Synuclein misfolding and Parkinson’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2012, 1822, 261–285. [Google Scholar] [CrossRef] [Green Version]
  81. Wallin, C.; Friedemann, M.; Sholts, S.B.; Noormägi, A.; Svantesson, T.; Jarvet, J.; Roos, P.M.; Palumaa, P.; Gräslund, A.; Wärmländer, S.K.T.S. Mercury and Alzheimer’s Disease: Hg(II) Ions Display Specific Binding to the Amyloid-β Peptide and Hinder Its Fibrillization. Biomolecules 2020, 10, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ceccatelli, S.; Daré, E.; Moors, M. Methylmercury-induced neurotoxicity and apoptosis. Chem. Biol. Interact. 2010, 188, 301–308. [Google Scholar] [CrossRef] [PubMed]
  83. Sian-Hülsmann, J.; Mandel, S.; Youdim, M.B.H.; Riederer, P. The relevance of iron in the pathogenesis of Parkinson’s disease. J. Neurochem. 2010, 118, 939–957. [Google Scholar] [CrossRef]
  84. Muñoz, P.; Humeres, A. Iron deficiency on neuronal function. BioMetals 2012, 25, 825–835. [Google Scholar] [CrossRef] [PubMed]
  85. Savica, R.; Grossardt, B.R.; Carlin, J.M.; Icen, M.; Bower, J.H.; Ahlskog, J.E.; Maraganore, D.M.; Steensma, D.P.; Rocca, W.A. Anemia or low hemoglobin levels preceding Parkinson disease: A case-control study. Neurology 2009, 73, 1381–1387. [Google Scholar] [CrossRef] [Green Version]
  86. Dev, S.; Babitt, J.L. Overview of iron metabolism in health and disease. Hemodial. Int. 2017, 21, S6–S20. [Google Scholar] [CrossRef]
  87. Hare, D.J.; Arora, M.; Jenkins, N.L.; Finkelstein, D.I.; Doble, P.A.; Bush, A.I. Is early-life iron exposure critical in neurodegeneration? Nat. Rev. Neurol. 2015, 11, 536–544. [Google Scholar] [CrossRef] [PubMed]
  88. Hare, D.J.; Cardoso, B.R.; Raven, E.P.; Double, K.L.; Finkelstein, D.I.; Szymlek-Gay, E.A.; Biggs, B.-A. Excessive early-life dietary exposure: A potential source of elevated brain iron and a risk factor for Parkinson’s disease. NPJ Parkinson Dis. 2017, 3, 1. [Google Scholar] [CrossRef] [PubMed]
  89. Costa-Mallen, P.; Gatenby, C.; Friend, S.; Maravilla, K.R.; Hu, S.-C.; Cain, K.C.; Agarwal, P.; Anzai, Y. Brain iron concentrations in regions of interest and relation with serum iron levels in Parkinson disease. J. Neurol. Sci. 2017, 378, 38–44. [Google Scholar] [CrossRef]
  90. Logroscino, G.; Marder, K.; Graziano, J.; Freyer, G.; Slavkovich, V.; LoIacono, N.; Cote, L.; Mayeux, R. Altered systemic iron metabolism in Parkinson’s disease. Neurology 1997, 49, 714–717. [Google Scholar] [CrossRef]
  91. Fasano, M.; Bergamasco, B.; Lopiano, L. Modifications of the iron-neuromelanin system in Parkinson’s disease. J. Neurochem. 2006, 96, 909–916. [Google Scholar] [CrossRef] [PubMed]
  92. Gerlach, M.; Double, K.L.; Youdim, M.B.H.; Riederer, P. Potential sources of increased iron in the substantia nigra of parkinsonian patients. Parkinson Dis. Relat. Disord. 2006, 70, 133–142. [Google Scholar] [CrossRef]
  93. Lotfipour, A.K.; Wharton, S.; Schwarz, S.T.; Gontu, V.; Schäfer, A.; Peters, A.M.; Bowtell, R.W.; Auer, D.P.; Gowland, P.A.; Bajaj, N.P.S. High resolution magnetic susceptibility mapping of the substantia nigra in Parkinson’s disease. J. Magn. Reson. Imaging 2012, 35, 48–55. [Google Scholar] [CrossRef]
  94. Levi, S.; Finazzi, D. Neurodegeneration with brain iron accumulation: Update on pathogenic mechanisms. Front. Pharmacol. 2014, 5, 99. [Google Scholar] [CrossRef] [Green Version]
  95. Xuan, M.; Guan, X.; Gu, Q.; Shen, Z.; Yu, X.; Qiu, T.; Luo, X.; Song, R.; Jiaerken, Y.; Xu, X.; et al. Different iron deposition patterns in early- and middle-late-onset Parkinson’s disease. Parkinsonism Relat. Disord. 2017, 44, 23–27. [Google Scholar] [CrossRef]
  96. Dexter, D.T.; Wells, F.R.; Lee, A.J.; Agid, F.; Agid, Y.; Jenner, P.; Marsden, C.D. Increased Nigral Iron Content and Alterations in Other Metal Ions Occurring in Brain in Parkinson’s Disease. J. Neurochem. 1989, 52, 1830–1836. [Google Scholar] [CrossRef]
  97. Ulla, M.; Bonny, J.M.; Ouchchane, L.; Rieu, I.; Claise, B.; Durif, F. Is R2* a New MRI Biomarker for the Progression of Parkinson’s Disease? A Longitudinal Follow-Up. PLoS ONE 2013, 8, e57904. [Google Scholar] [CrossRef] [PubMed]
  98. Yu, X.; Du, T.; Song, N.; He, Q.; Shen, Y.; Jiang, H.; Xie, J. Decreased iron levels in the temporal cortex in postmortem human brains with Parkinson disease. Neurology 2013, 80, 492–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
  100. Turner, J.R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef] [PubMed]
  101. Takiishi, T.; Fenero, C.I.M.; Câmara, N.O.S. Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef] [PubMed]
  102. Pott, J.; Hornef, M. Innate immune signalling at the intestinal epithelium in homeostasis and disease. EMBO Rep. 2012, 13, 684–698. [Google Scholar] [CrossRef] [Green Version]
  103. Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kainulainen, V.; Jalanka, J.; Satokari, R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018, 10, 988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Romero, E.S.; Cotoner, C.A.; Camacho, C.P.; Bedmar, M.C.; Vicario, M. The intestinal barrier function and its involvement in digestive disease. Rev. Esp. Enferm. Dig. 2015, 108, 686–696. [Google Scholar] [CrossRef] [Green Version]
  105. Breton, J.Ô.; Daniel, C.; Dewulf, J.; Pothion, S.; Froux, N.; Sauty, M.; Thomas, P.; Pot, B.; Foligne, B. Gut microbiota limits heavy metals burden caused by chronic oral exposure. Toxicol. Lett. 2013, 222, 132–138. [Google Scholar] [CrossRef] [PubMed]
  106. Breton, J.; Le Clère, K.; Daniel, C.; Sauty, M.; Nakab, L.; Chassat, T.; Dewulf, J.; Penet, S.; Carnoy, C.; Thomas, P.; et al. Chronic ingestion of cadmium and lead alters the bioavailability of essential and heavy metals, gene expression pathways and genotoxicity in mouse intestine. Arch. Toxicol. 2013, 87, 1787–1795. [Google Scholar] [CrossRef]
  107. Li, X.; Brejnrod, A.; Ernst, M.; Rykær, M.; Herschend, J.; Olsen, N.M.C.; Dorrestein, P.C.; Rensing, C.; Sørensen, S.J. Heavy metal exposure causes changes in the metabolic health-associated gut microbiome and metabolites. Environ. Int. 2019, 126, 454–467. [Google Scholar] [CrossRef] [PubMed]
  108. Tinkov, A.A.; Gritsenko, V.A.; Skalnaya, M.G.; Cherkasov, S.V.; Aaseth, J.; Skalny, A.V. Gut as a target for cadmium toxicity. Environ. Pollut. 2018, 235, 429–434. [Google Scholar] [CrossRef]
  109. Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The Brain-Gut-Microbiome Axis. Cell. Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef] [Green Version]
  110. 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]
  111. Coryell, M.; Roggenbeck, B.A.; Walk, S.T. The Human Gut Microbiome’s Influence on Arsenic Toxicity. Curr. Pharmacol. Rep. 2019, 5, 491–504. [Google Scholar] [CrossRef] [Green Version]
  112. Assefa, S.; Köhler, G. Intestinal microbiome and metal toxicity. Curr. Opin. Toxicol. 2019, 19, 21–27. [Google Scholar] [CrossRef]
  113. Breit, S.; Kupferberg, A.; Rogler, G.; Hasler, G. Vagus Nerve as Modulator of the Brain–Gut Axis in Psychiatric and Inflammatory Disorders. Front. Psychiatry 2018, 9, 44. [Google Scholar] [CrossRef] [Green Version]
  114. Galland, L. The Gut Microbiome and the Brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef] [PubMed]
  115. Olmo, B.M.G.; Butler, M.J.; Barrientos, R.M. Evolution of the Human Diet and Its Impact on Gut Microbiota, Immune Responses, and Brain Health. Nutrients 2021, 13, 196. [Google Scholar] [CrossRef]
  116. Tsavkelova, E.A.; Botvinko, I.V.; Kudrin, V.S.; Oleskin, A.V. Detection of neurotransmitter amines in microorganisms with the use of high-performance liquid chromatography. Dokl. Biochem. 2000, 372, 115–117. [Google Scholar] [PubMed]
  117. Özoğul, F. Production of biogenic amines by Morganella morganii, Klebsiella pneumoniae and Hafnia alvei using a rapid HPLC method. Eur. Food Res. Technol. 2004, 219, 465–469. [Google Scholar] [CrossRef]
  118. Shishov, V.A.; Kirovskaya, T.A.; Kudrin, V.S.; Oleskin, A.V. Amine neuromediators, their precursors, and oxidation products in the culture of Escherichia coli K-12. Appl. Biochem. Microbiol. 2009, 45, 494–497. [Google Scholar] [CrossRef]
  119. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  120. Bonaz, B.; Sinniger, V.; Hoffmann, D.; Clarençon, D.; Mathieu, N.; Dantzer, C.; Vercueil, L.; Picq, C.; Trocmé, C.; Faure, P.; et al. Chronic vagus nerve stimulation in Crohn’s disease: A 6-month follow-up pilot study. Neurogastroenterol. Motil. 2016, 28, 948–953. [Google Scholar] [CrossRef] [PubMed]
  121. Jaglin, M.; Rhimi, M.; Philippe, C.; Pons, N.; Bruneau, A.; Goustard, B.; Daugé, V.; Maguin, E.; Naudon, L.; Rabot, S. Indole, a Signaling Molecule Produced by the Gut Microbiota, Negatively Impacts Emotional Behaviors in Rats. Front. Neurosci. 2018, 12, 216. [Google Scholar] [CrossRef]
  122. Woźniak, D.; Cichy, W.; Przysławski, J.; Drzymała-Czyż, S. The role of microbiota and enteroendocrine cells in maintaining homeostasis in the human digestive tract. Adv. Med. Sci. 2021, 66, 284–292. [Google Scholar] [CrossRef]
  123. Latorre, R.; Sternini, C.; de Giorgio, R.; Meerveld, B.G.-V. Enteroendocrine cells: A review of their role in brain-gut communication. Neurogastroenterol. Motil. 2015, 28, 620–630. [Google Scholar] [CrossRef] [Green Version]
  124. Chimerel, C.; Emery, E.; Summers, D.K.; Keyser, U.; Gribble, F.M.; Reimann, F. Bacterial Metabolite Indole Modulates Incretin Secretion from Intestinal Enteroendocrine L Cells. Cell Rep. 2014, 9, 1202–1208. [Google Scholar] [CrossRef] [Green Version]
  125. Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424–429. [Google Scholar] [CrossRef] [Green Version]
  126. Lund, M.L.; Sorrentino, G.; Egerod, K.L.; Kroone, C.; Mortensen, B.; Knop, F.K.; Reimann, F.; Gribble, F.M.; Drucker, D.J.; de Koning, E.J.P.; et al. L-Cell Differentiation Is Induced by Bile Acids Through GPBAR1 and Paracrine GLP-1 and Serotonin Signaling. Diabetes 2020, 69, 614–623. [Google Scholar] [CrossRef]
  127. Panwar, H.; Calderwood, D.; Gillespie, A.L.; Wylie, A.R.; Graham, S.F.; Grant, I.R.; Grover, S.; Green, B.D. Identification of lactic acid bacteria strains modulating incretin hormone secretion and gene expression in enteroendocrine cells. J. Funct. Foods 2016, 23, 348–358. [Google Scholar] [CrossRef] [Green Version]
  128. Ticho, A.L.; Malhotra, P.; Dudeja, P.K.; Gill, R.K.; Alrefai, W.A. Bile acid receptors and gastrointestinal functions. Liver Res. 2019, 3, 31–39. [Google Scholar] [CrossRef]
  129. Alemi, F.; Poole, D.P.; Chiu, J.; Schoonjans, K.; Cattaruzza, F.; Grider, J.R.; Bunnett, N.W.; Corvera, C.U. The receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is required for normal defecation in mice. Gastroenterology 2013, 144, 145–154. [Google Scholar] [CrossRef]
  130. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. (Lausanne) 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
  131. Covasa, M.; Stephens, R.W.; Toderean, R.; Cobuz, C. Intestinal sensing by gut microbiota: Targeting gut peptides. Front. Endocrinol. (Lausanne) 2019, 10, 82. [Google Scholar] [CrossRef] [Green Version]
  132. Mishra, S.P.; Karunakar, P.; Taraphder, S.; Yadav, H. Free fatty acid receptors 2 and 3 as microbial metabolite sensors to shape host health: Pharmacophysiological view. Biomedicines 2020, 8, 154. [Google Scholar] [CrossRef]
  133. Venegas, D.P.; De La Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short chain fatty acids (SCFAs)mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  134. Overby, H.B.; Ferguson, J.F. Gut Microbiota-Derived Short-Chain Fatty Acids Facilitate Microbiota: Host Cross talk and Modulate Obesity and Hypertension. Curr. Hypertens. Rep. 2021, 23, 8. [Google Scholar] [CrossRef]
  135. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [Green Version]
  136. Litvak, Y.; Byndloss, M.X.; Bäumler, A.J. Colonocyte metabolism shapes the gut microbiota. Science 2018, 362, eaat9076. [Google Scholar] [CrossRef] [Green Version]
  137. Gershon, M.D.; Tack, J. The Serotonin Signaling System: From Basic Understanding To Drug Development for Functional GI Disorders. Gastroenterology 2007, 132, 397–414. [Google Scholar] [CrossRef]
  138. Mawe, G.M.; Hoffman, J.M. Serotonin Signaling in the Gastrointestinal Tract: Functions, dysfunctions, and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 22, 313–333. [Google Scholar] [CrossRef]
  139. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous Bacteria from the Gut Microbiota Regulate Host Serotonin Biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [Green Version]
  140. Ozogul, F.; Kuley, E.; Ozogul, Y.; Özoğul, I. The Function of Lactic Acid Bacteria on Biogenic Amines Production by Food-Borne Pathogens in Arginine Decarboxylase Broth. Food Sci. Technol. Res. 2012, 18, 795–804. [Google Scholar] [CrossRef] [Green Version]
  141. Terry, N.; Margolis, K.G. Serotonergic Mechanisms Regulating the GI Tract: Experimental Evidence and Therapeutic Relevance. Gastrointest. Pharmacol. 2016, 239, 319–342. [Google Scholar] [CrossRef] [Green Version]
  142. De Vadder, F.; Grasset, E.; Holm, L.M.; Karsenty, G.; MacPherson, A.J.; Olofsson, L.E.; Bäckhed, F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc. Natl. Acad. Sci. USA 2018, 115, 6458–6463. [Google Scholar] [CrossRef] [Green Version]
  143. Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef] [Green Version]
  144. Golubeva, A.V.; Joyce, S.A.; Moloney, G.; Burokas, A.; Sherwin, E.; Arboleya, S.; Flynn, I.; Khochanskiy, D.; Moya-Pérez, A.; Peterson, V.; et al. Microbiota-related Changes in Bile Acid & Tryptophan Metabolism are Associated with Gastrointestinal Dysfunction in a Mouse Model of Autism. EBioMedicine 2017, 24, 166–178. [Google Scholar] [CrossRef] [Green Version]
  145. Wikoff, W.R.; Anfora, A.T.; Liu, J.; Schultz, P.G.; Lesley, S.A.; Peters, E.C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. USA 2009, 106, 3698–3703. [Google Scholar] [CrossRef] [Green Version]
  146. Sjögren, K.; Engdahl, C.; Henning, P.; Lerner, U.H.; Tremaroli, V.; Lagerquist, M.K.; Bäckhed, F.; Ohlsson, C. The gut microbiota regulates bone mass in mice. J. Bone Miner. Res. 2012, 27, 1357–1367. [Google Scholar] [CrossRef] [Green Version]
  147. Mandić, A.D.; Woting, A.; Jaenicke, T.; Sander, A.; Sabrowski, W.; Rolle-Kampcyk, U.; Von Bergen, M.; Blaut, M. Clostridium ramosum regulates enterochromaffin cell development and serotonin release. Sci. Rep. 2019, 9, 1177. [Google Scholar] [CrossRef] [Green Version]
  148. Fond, G.; Loundou, A.; Hamdani, N.; Boukouaci, W.; Dargel, A.; Oliveira, J.; Roger, M.; Tamouza, R.; Leboyer, M.; Boyer, L. Anxiety and depression comorbidities in irritable bowel syndrome (IBS): A systematic review and meta-analysis. Eur. Arch. Psychiatry Clin. Neurosci. 2014, 264, 651–660. [Google Scholar] [CrossRef]
  149. Fung, T.C. The microbiota-immune axis as a central mediator of gut-brain communication. Neurobiol. Dis. 2019, 136, 104714. [Google Scholar] [CrossRef]
  150. Dumitrescu, L.; Popescu-Olaru, I.; Cozma, L.; Tulbă, D.; Hinescu, M.E.; Ceafalan, L.C.; Gherghiceanu, M.; Popescu, B.O. Oxidative Stress and the Microbiota-Gut-Brain Axis. Oxidative Med. Cell. Longev. 2018, 2018, 2406594. [Google Scholar] [CrossRef] [Green Version]
  151. MacDonald, T.T.; Monteleone, G. Immunity, Inflammation, and Allergy in the Gut. Science 2005, 307, 1920–1925. [Google Scholar] [CrossRef] [Green Version]
  152. Hooper, L.V.; Littman, D.R.; MacPherson, A.J. Interactions between the Microbiota and the Immune System. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [Green Version]
  153. Powell, N.; Walker, M.M.; Talley, N.J. The mucosal immune system: Master regulator of bidirectional gut–brain communications. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 143–159. [Google Scholar] [CrossRef]
  154. Sommer, F.; Bäckhed, F. The gut microbiota—Masters of host development and physiology. Nat. Rev. Microbiol. 2013, 11, 227–238. [Google Scholar] [CrossRef] [PubMed]
  155. Hall, J.A.; Bouladoux, N.; Sun, C.M.; Wohlfert, E.A.; Blank, R.B.; Zhu, Q.; Grigg, M.E.; Berzofsky, J.A.; Belkaid, Y. Commensal DNA Limits Regulatory T Cell Conversion and Is a Natural Adjuvant of Intestinal Immune Responses. Immunity 2008, 29, 637–649. [Google Scholar] [CrossRef] [Green Version]
  156. Cheng, H.; Guan, X.; Chen, D.; Ma, W. The th17/treg cell balance: A gut microbiota-modulated story. Microorganisms 2019, 7, 583. [Google Scholar] [CrossRef] [Green Version]
  157. Karimi, K.; Inman, M.D.; Bienenstock, J.; Forsythe, P. Lactobacillus reuteri–induced Regulatory T cells Protect against an Allergic Airway Response in Mice. Am. J. Respir. Crit. Care Med. 2009, 179, 186–193. [Google Scholar] [CrossRef] [PubMed]
  158. Lyons, A.; O’Mahony, D.; O’Brien, F.; Mac Sharry, J.; Sheil, B.; Ceddia, M.; Russell, W.M.; Forsythe, P.; Bienenstock, J.; Kiely, B.; et al. Bacterial strain-specific induction of Foxp3+T regulatory cells is protective in murine allergy models. Clin. Exp. Allergy 2010, 40, 811–819. [Google Scholar] [CrossRef]
  159. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous clostridium species. Science 2011, 331, 337–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Lu, L.; Barbi, J.; Pan, F. The regulation of immune tolerance by FOXP3. Nat. Rev. Immunol. 2017, 17, 703–717. [Google Scholar] [CrossRef] [PubMed]
  161. Bain, C.C.; Schridde, A. Origin, differentiation, and function of intestinal macrophages. Front. Immunol. 2018. [Google Scholar] [CrossRef]
  162. Jacobson, A.; Yang, D.; Vella, M.; Chiu, I.M. The intestinal neuro-immune axis: Crosstalk between neurons, immune cells, and microbes. Mucosal Immunol. 2021, 14, 555–565. [Google Scholar] [CrossRef]
  163. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef] [Green Version]
  164. Muller, P.A.; Koscsó, B.; Rajani, G.M.; Stevanovic, K.; Berres, M.L.; Hashimoto, D.; Mortha, A.; Leboeuf, M.; Li, X.M.; Mucida, D.; et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell 2014, 158, 300–313. [Google Scholar] [CrossRef] [Green Version]
  165. Obata, Y.; Pachnis, V. The Effect of Microbiota and the Immune System on the Development and Organization of the Enteric Nervous System. Gastroenterology 2016, 151, 836–844. [Google Scholar] [CrossRef] [Green Version]
  166. Gabanyi, I.; Muller, P.A.; Feighery, L.; Oliveira, T.Y.; Costa-Pinto, F.A.; Mucida, D. Neuro-immune Interactions Drive Tissue Programming in Intestinal Macrophages. Cell 2016, 164, 378–391. [Google Scholar] [CrossRef] [Green Version]
  167. Lycke, N.Y.; Bemark, M. The regulation of gut mucosal IgA B-cell responses: Recent developments. Mucosal Immunol. 2017, 10, 1361–1374. [Google Scholar] [CrossRef]
  168. Zheng, W.; He, R.; Yan, Z.; Huang, Y.; Huang, W.; Cai, Z.; Su, Y.; Liu, S.; Deng, Y.; Wang, Q.; et al. Regulation of immune-driven pathogenesis in Parkinson’s disease by gut microbiota. Brain. Behav. Immun. 2020, 87, 890–897. [Google Scholar] [CrossRef]
  169. Chen, Q.-Q.; Haikal, C.; Li, W.; Li, J.-Y. Gut Inflammation in Association with Pathogenesis of Parkinson’s Disease. Front. Mol. Neurosci. 2019, 12, 218. [Google Scholar] [CrossRef] [Green Version]
  170. 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]
  171. Scheperjans, F.; Aho, V.; Pereira, P.A.; 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]
  172. 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. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
  173. Li, W.; Wu, X.L.; Hu, X.; Wang, T.; Liang, S.; Duan, Y.F.; 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]
  174. 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. 2018, 34, 396–405. [Google Scholar] [CrossRef] [PubMed]
  175. Campos-Acuña, J.; Elgueta, D.; Pacheco, R. T-Cell-Driven Inflammation as a Mediator of the Gut-Brain Axis Involved in Parkinson’s Disease. Front. Immunol. 2019, 10, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Mosley, R.L.; Hutter-Saunders, J.A.; Stone, D.K.; Gendelman, H.E. Inflammation and Adaptive Immunity in Parkinson’s Disease. Cold Spring Harb. Perspect. Med. 2011, 2, a009381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Jahromi, M.F.; Liang, J.B.; Ebrahimi, R.; Soleimani, A.F.; Rezaeizadeh, A.; Abdullah, N.; Shokryazdan, P. Protective potential of Lactobacillus species in lead toxicity model in broiler chickens. Animal 2017, 11, 755–761. [Google Scholar] [CrossRef] [Green Version]
  178. Bedarf, J.R.; Hildebrand, F.; Coelho, L.P.; Sunagawa, S.; Bahram, M.; Goeser, F.; Bork, P.; Wüllner, U. Functional implications of microbial and viral gut metagenome changes in early stage L-DOPA-naïve Parkinson’s disease patients. Genome Med. 2017, 9, 39. [Google Scholar] [CrossRef]
  179. Devos, D.; Lebouvier, T.; Lardeux, B.; Biraud, M.; Rouaud, T.; Pouclet, H.; Coron, E.; Varannes, S.B.D.; Naveilhan, P.; Nguyen, J.-M.; et al. Colonic inflammation in Parkinson’s disease. Neurobiol. Dis. 2013, 50, 42–48. [Google Scholar] [CrossRef]
  180. Osburg, B.; Peiser, C.; Dömling, D.; Schomburg, L.; Ko, Y.T.; Voigt, K.; Bickel, U. Effect of endotoxin on expression of TNF receptors and transport of TNF-α at the blood-brain barrier of the rat. Am. J. Physiol. Metab. 2002, 283, E899–E908. [Google Scholar] [CrossRef] [Green Version]
  181. Benner, E.J.; Banerjee, R.; Reynolds, A.D.; Sherman, S.; Pisarev, V.M.; Tsiperson, V.; Nemachek, C.; Ciborowski, P.; Przedborski, S.; Mosley, R.L.; et al. Nitrated α–Synuclein Immunity Accelerates Degeneration of Nigral Dopaminergic Neurons. PLoS ONE 2008, 3, e1376. [Google Scholar] [CrossRef]
  182. Choi, J.G.; Kim, N.; Ju, I.G.; Eo, H.; Lim, S.-M.; Jang, S.-E.; Kim, D.-H.; Oh, M.S. Oral administration of Proteus mirabilis damages dopaminergic neurons and motor functions in mice. Sci. Rep. 2018, 8, 1275. [Google Scholar] [CrossRef] [Green Version]
  183. Chen, Q.; Chen, Y.; Zhang, Y.; Wang, F.; Yu, H.; Zhang, C.; Jiang, Z.; Luo, W. Iron deposition in Parkinson’s disease by quantitative susceptibility mapping. BMC Neurosci. 2019, 20, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Miranda, M.; López-Alonso, M.; Castillo, C.; Hernández, J.L.; Benedito, J. Effects of moderate pollution on toxic and trace metal levels in calves from a polluted area of northern Spain. Environ. Int. 2005, 31, 543–548. [Google Scholar] [CrossRef]
  185. McConnell, J.R.; Edwards, R. Coal burning leaves toxic heavy metal legacy in the Arctic. Proc. Natl. Acad. Sci. USA 2008, 105, 12140–12144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Petersen, M.S.; Halling, J.; Bech, S.; Wermuth, L.; Weihe, P.; Nielsen, F.; Jørgensen, P.J.; Budtz-Jørgensen, E.; Grandjean, P. Impact of dietary exposure to food contaminants on the risk of Parkinson’s disease. Neurotoxicology 2008, 29, 584–590. [Google Scholar] [CrossRef]
  187. Hadayat, N.; De Oliveira, L.M.; Da Silva, E.; Han, L.; Hussain, M.; Liu, X.; Ma, L.Q. Assessment of trace metals in five most-consumed vegetables in the US: Conventional vs. organic. Environ. Pollut. 2018, 243, 292–300. [Google Scholar] [CrossRef]
  188. Fu, L.; Lu, X.; Niu, K.; Tan, J.; Chen, J. Bioaccumulation and human health implications of essential and toxic metals in freshwater products of Northeast China. Sci. Total Environ. 2019, 673, 768–776. [Google Scholar] [CrossRef]
  189. Miclean, M.; Cadar, O.; Levei, E.A.; Roman, R.; Ozunu, A.; Levei, L. Metal (Pb, Cu, Cd, and Zn) Transfer along Food Chain and Health Risk Assessment through Raw Milk Consumption from Free-Range Cows. Int. J. Environ. Res. Public Health 2019, 16, 4064. [Google Scholar] [CrossRef] [Green Version]
  190. Tshala-Katumbay, D.; Mwanza, J.-C.; Rohlman, D.S.; Maestre, G.E.; Oriá, R. A global perspective on the influence of environmental exposures on the nervous system. Nature 2015, 527, S187–S192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Duan, H.; Yu, L.; Tian, F.; Zhai, Q.; Fan, L.; Chen, W. Gut microbiota: A target for heavy metal toxicity and a probiotic protective strategy. Sci. Total Environ. 2020, 742, 140429. [Google Scholar] [CrossRef]
  192. Giambò, F.; Italia, S.; Teodoro, M.; Briguglio, G.; Furnari, N.; Catanoso, R.; Costa, C.; Fenga, C. Influence of toxic metal exposure on the gut microbiota (Review). World Acad. Sci. J. 2021, 3, 1. [Google Scholar] [CrossRef]
  193. Rosenfeld, C.S. Gut Dysbiosis in Animals Due to Environmental Chemical Exposures. Front. Cell. Infect. Microbiol. 2017, 7, 396. [Google Scholar] [CrossRef] [Green Version]
  194. Priyadarshi, A.; Khuder, S.A.; Schaub, E.A.; Priyadarshi, S.S. Environmental Risk Factors and Parkinson’s Disease: A Metaanalysis. Environ. Res. 2001, 86, 122–127. [Google Scholar] [CrossRef] [Green Version]
  195. Dick, F.D.; De Palma, G.; Ahmadi, A.; Scott, N.W.; Prescott, G.J.; Bennett, J.; Semple, S.; Dick, S.; Counsell, C.; Mozzoni, P.; et al. Environmental risk factors for Parkinson’s disease and parkinsonism: The Geoparkinson study. Occup. Environ. Med. 2007, 64, 666–672. [Google Scholar] [CrossRef] [Green Version]
  196. Seidler, A.; Hellenbrand, W.; Robra, B.-P.; Vieregge, P.; Nischan, P.; Joerg, J.; Oertel, W.H.; Ulm, G.; Schneider, E. Possible environmental, occupational, and other etiologic factors for Parkinson’s disease: A case-control study in Germany. Neurology 1996, 46, 1275. [Google Scholar] [CrossRef]
  197. Jinsmaa, Y.; Sullivan, P.; Gross, D.; Cooney, A.; Sharabi, Y.; Goldstein, D.S. Divalent metal ions enhance DOPAL-induced oligomerization of alpha-synuclein. Neurosci. Lett. 2014, 569, 27–32. [Google Scholar] [CrossRef] [Green Version]
  198. Zhang, J.; Cai, T.; Zhao, F.; Yao, T.; Chen, Y.; Liu, X.; Luo, W.; Chen, J. The Role of α-synuclein and Tau Hyperphosphorylation-Mediated Autophagy and Apoptosis in Lead-induced Learning and Memory Injury. Int. J. Biol. Sci. 2012, 8, 935–944. [Google Scholar] [CrossRef]
  199. Kwakye, G.F.; Paoliello, M.M.B.; Mukhopadhyay, S.; Bowman, A.B.; Aschner, M. Manganese-induced Parkinsonism and Parkinson’s disease: Shared and distinguishable features. Int. J. Environ. Res. Public Health 2015, 12, 7519–7540. [Google Scholar] [CrossRef] [Green Version]
  200. Miller, K.; Ochudło, S.; Opala, G.; Smolicha, W.; Siuda, J. Parkinsonism in chronic occupational metallic mercury intoxication. Neurol. Neurochir. Polska 2003, 37, 31–38. [Google Scholar]
  201. Bridges, C.C.; Zalups, R.K. Molecular and ionic mimicry and the transport of toxic metals. Toxicol. Appl. Pharmacol. 2005, 204, 274–308. [Google Scholar] [CrossRef] [Green Version]
  202. Fujishiro, H.; Okugaki, S.; Kubota, K.; Fujiyama, T.; Miyataka, H.; Himeno, S. The role of ZIP8 down-regulation in cadmium-resistant metallothionein-null cells. J. Appl. Toxicol. 2009, 29, 367–373. [Google Scholar] [CrossRef]
  203. Vesey, D.A. Transport pathways for cadmium in the intestine and kidney proximal tubule: Focus on the interaction with essential metals. Toxicol. Lett. 2010, 198, 13–19. [Google Scholar] [CrossRef] [PubMed]
  204. Caito, S.; Aschner, M. Neurotoxicity of metals. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2015. [Google Scholar]
  205. Roth, J.; Ponzoni, S.; Aschner, M. Manganese Homeostasis and Transport. Met. Cell 2012, 12, 169–201. [Google Scholar] [CrossRef]
  206. Ballatori, N. Transport of toxic metals by molecular mimicry. Environ. Health Perspect. 2002, 110, 689–694. [Google Scholar] [CrossRef] [Green Version]
  207. Bressler, J.P.; Olivi, L.; Cheong, J.H.; Kim, Y.; Maerten, A.; Bannon, D. Metal transporters in intestine and brain: Their involvement in metal-associated neurotoxicities. Hum. Exp. Toxicol. 2007, 26, 221–229. [Google Scholar] [CrossRef] [PubMed]
  208. Dubey, V.; Mishra, A.K.; Ghosh, A.R.; Mandal, B.K. Probiotic Pediococcus pentosaceus GS4 shields brush border membrane and alleviates liver toxicity imposed by chronic cadmium exposure in Swiss albino mice. J. Appl. Microbiol. 2019, 126, 1233–1244. [Google Scholar] [CrossRef]
  209. Tulpule, K.; Robinson, S.R.; Bishop, G.M.; Dringen, R. Uptake of ferrous iron by cultured rat astrocytes. J. Neurosci. Res. 2010, 88, 563–571. [Google Scholar] [CrossRef] [PubMed]
  210. Bannon, D.I.; Abounader, R.; Lees, P.S.J.; Bressler, J.P. Effect of DMT1 knockdown on iron, cadmium, and lead uptake in Caco-2 cells. Am. J. Physiol. Cell Physiol. 2003, 284, C44–C50. [Google Scholar] [CrossRef] [Green Version]
  211. Singh, N. The Role of Iron in Prion Disease and Other Neurodegenerative Diseases. PLoS Pathog. 2014, 10, e1004335. [Google Scholar] [CrossRef]
  212. Chiacchia, M.; Cerutti, C.; Gromnicova, R.; Rietdorf, K.; Romero, I.A.; Bradshaw, D. Zinc-imidazolate polymers (ZIPs) as a potential carrier to brain capillary endothelial cells. J. Mater. Chem. B 2015, 3, 9053–9059. [Google Scholar] [CrossRef] [Green Version]
  213. Xu, H.; Wang, Y.; Song, N.; Wang, J.; Jiang, H.; Xie, J. New Progress on the Role of Glia in Iron Metabolism and Iron-Induced Degeneration of Dopamine Neurons in Parkinson’s Disease. Front. Mol. Neurosci. 2018, 10, 455. [Google Scholar] [CrossRef]
  214. Hoepken, H.H.; Korten, T.; Robinson, S.R.; Dringen, R. Iron accumulation, iron-mediated toxicity and altered levels of ferritin and transferrin receptor in cultured astrocytes during incubation with ferric ammonium citrate. J. Neurochem. 2004, 88, 1194–1202. [Google Scholar] [CrossRef]
  215. Steimle, B.L.; Smith, F.M.; Kosman, D.J. The solute carriers ZIP8 and ZIP14 regulate manganese accumulation in brain microvascular endothelial cells and control brain manganese levels. J. Biol. Chem. 2019, 294, 19197–19208. [Google Scholar] [CrossRef]
  216. Kumar, V. Mechanism of Microbial Heavy Metal Accumulation from a Polluted Environment and Bioremediation. In Microbial Cell Factories; Deepansh Sharma, B.S.S., Ed.; CRC Press: Boca Raton, FL, USA, 2018; pp. 156–158. [Google Scholar]
  217. Yokooji, T.; Murakami, T.; Yumoto, R.; Nagai, J.; Takano, M. Site-specific bidirectional efflux of 2,4-dinitrophenyl-S-glutathione, a substrate of multidrug resistance-associated proteins, in rat intestine and Caco-2 cells. J. Pharm. Pharmacol. 2007, 59, 513–520. [Google Scholar] [CrossRef]
  218. Calatayud, M.; Barrios, J.A.; Vélez, D.; Devesa, V. In Vitro Study of Transporters Involved in Intestinal Absorption of Inorganic Arsenic. Chem. Res. Toxicol. 2012, 25, 446–453. [Google Scholar] [CrossRef]
  219. Maciaszczyk-Dziubinska, E.; Wawrzycka, D.; Wysocki, R. Arsenic and Antimony Transporters in Eukaryotes. Int. J. Mol. Sci. 2012, 13, 3527–3548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Nico, B.; Frigeri, A.; Nicchia, G.P.; Corsi, P.; Ribatti, D.; Quondamatteo, F.; Herken, R.; Girolamo, F.; Marzullo, A.; Svelto, M.; et al. Severe alterations of endothelial and glial cells in the blood-brain barrier of dystrophic mdx mice. Glia 2003, 42, 235–251. [Google Scholar] [CrossRef]
  221. Zheng, W.; Aschner, M.; Ghersi-Egea, J.-F. Brain barrier systems: A new frontier in metal neurotoxicological research. Toxicol. Appl. Pharmacol. 2003, 192, 1–11. [Google Scholar] [CrossRef] [Green Version]
  222. Takahashi, T.; Shimohata, T. Vascular Dysfunction Induced by Mercury Exposure. Int. J. Mol. Sci. 2019, 20, 2435. [Google Scholar] [CrossRef] [Green Version]
  223. Cragg, R.A.; Christie, G.R.; Phillips, S.R.; Russi, R.M.; Küry, S.; Mathers, J.C.; Taylor, P.M.; Ford, D. A Novel Zinc-regulated Human Zinc Transporter, hZTL1, Is Localized to the Enterocyte Apical Membrane. J. Biol. Chem. 2002, 277, 22789–22797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Garza-Lombó, C.; Posadas, Y.; Quintanar, L.; Gonsebatt, M.E.; Franco, R. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid. Redox Signal. 2018, 28, 1669–1703. [Google Scholar] [CrossRef] [Green Version]
  225. La-Llave-León, O.; Méndez-Hernández, E.M.; Castellanos-Juárez, F.X.; Esquivel-Rodríguez, E.; Vázquez-Alaniz, F.; Sandoval-Carrillo, A.; García-Vargas, G.; Duarte-Sustaita, J.; Candelas-Rangel, J.L.; Salas-Pacheco, J.M. Association between Blood Lead Levels and Delta-Aminolevulinic Acid Dehydratase in Pregnant Women. Int. J. Environ. Res. Public Health 2017, 14, 432. [Google Scholar] [CrossRef] [Green Version]
  226. Dallas, S.; Zhu, X.; Baruchel, S.; Schlichter, L.; Bendayan, R. Functional Expression of the Multidrug Resistance Protein 1 in Microglia. J. Pharmacol. Exp. Ther. 2003, 307, 282–290. [Google Scholar] [CrossRef] [Green Version]
  227. Meyer, N.; Koehler, Y.; Tulpule, K.; Dringen, R. Arsenate accumulation and arsenate-induced glutathione export in astrocyte-rich primary cultures. Neurochem. Int. 2013, 62, 1012–1019. [Google Scholar] [CrossRef]
  228. Su, W.; Pasternak, G.W. The role of multidrug resistance-associated protein in the blood-brain barrier and opioid analgesia. Synapse 2013, 67, 609–619. [Google Scholar] [CrossRef] [Green Version]
  229. Sabath, E.; Robles-Osorio, M.L. Medio ambiente y riñón: Nefrotoxicidad por metales pesados. Nefrologia 2012, 32, 279286. [Google Scholar]
  230. Zalups, R.K.; Koropatnick, J. Cellular and Molecular Biology of Metals; CRC Press: Boca Raton, FL, USA, 2010; ISBN 9781420059984. [Google Scholar]
  231. Bridges, C.C.; Zalups, R.K. Mechanisms involved in the transport of mercuric ions in target tissues. Arch. Toxicol. 2016, 91, 63–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Li, T.-T.; An, J.-X.; Xu, J.-Y.; Tuo, B.-G. Overview of organic anion transporters and organic anion transporter polypeptides and their roles in the liver. World J. Clin. Cases 2019, 7, 3915–3933. [Google Scholar] [CrossRef]
  233. Menon, A.V.; Chang, J.O.; Kim, J. Mechanisms of divalent metal toxicity in affective disorders. Toxicology 2015, 339, 58–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Peer, W.A.; Baxter, I.R.; Richards, E.L.; Freeman, J.L.; Murphy, A.S. Molecular Biology of Metal Homeostasis and Detoxification; Springer: Berlin/Heidelberg, Germany; New York, NY, USA, 2006; ISBN 3-540-22175-1. [Google Scholar]
  235. Wu, L.J.-C.; Leenders, A.G.M.; Cooperman, S.; Meyron-Holtz, E.; Smith, S.; Land, W.; Tsai, R.Y.L.; Berger, U.V.; Sheng, Z.-H.; Rouault, T.A. Expression of the iron transporter ferroportin in synaptic vesicles and the blood–brain barrier. Brain Res. 2004, 1001, 108–117. [Google Scholar] [CrossRef]
  236. Villa-Bellosta, R.; Sorribas, V. Arsenate transport by sodium/phosphate cotransporter type IIb. Toxicol. Appl. Pharmacol. 2010. [Google Scholar] [CrossRef]
  237. Miyamoto, K.I.; Haito-Sugino, S.; Kuwahara, S.; Ohi, A.; Nomura, K.; Ito, M.; Kuwahata, M.; Kido, S.; Tatsumi, S.; Kaneko, I.; et al. Sodium-dependent phosphate cotransporters: Lessons from gene knockout and mutation studies. J. Pharm. Sci. 2011, 100, 3719–3730. [Google Scholar]
  238. Mukhopadhyay, R.; Bhattacharjee, H.; Rosen, B.P. Aquaglyceroporins: Generalized metalloid channels. Biochim. Biophys. Acta Gen. Subj. 2014, 1840, 1583–1591. [Google Scholar] [CrossRef] [Green Version]
  239. Bhattacharjee, H.; Rosen, B.P.; Mukhopadhyay, R. Aquaglyceroporins and Metalloid Transport: Implications in Human Diseases. Aquaporins 2009, 190, 309–325. [Google Scholar] [CrossRef] [Green Version]
  240. Ximenes-Da-Silva, A. Metal Ion Toxins and Brain Aquaporin-4 Expression: An Overview. Front. Neurosci. 2016, 10, 233. [Google Scholar] [CrossRef] [Green Version]
  241. Sabolić, I.; Breljak, D.; Škarica, M.; Herak-Kramberger, C.M. Role of metallothionein in cadmium traffic and toxicity in kidneys and other mammalian organs. Biometals 2010, 23, 897–926. [Google Scholar] [CrossRef]
  242. Ruttkay-Nedecky, B.; Nejdl, L.; Gumulec, J.; Zitka, O.; Masarik, M.; Eckschlager, T.; Stiborova, M.; Adam, V.; Kizek, R. The Role of Metallothionein in Oxidative Stress. Int. J. Mol. Sci. 2013, 14, 6044–6066. [Google Scholar] [CrossRef] [Green Version]
  243. Petering, D.H.; Krezoski, S.; Tabatabai, N.M. 12 Metallothionein Toxicology: Metal Ion Trafficking and Cellular Protection. In Metallothioneins and Related Chelators; De Gruyter: Berlin, Germany, 2015. [Google Scholar]
  244. Anderson, G.J.; Vulpe, C.D. Mammalian iron transport. Cell. Mol. Life Sci. 2009, 66, 3241–3261. [Google Scholar] [CrossRef]
  245. Gonick, H.C. Lead-Binding Proteins: A Review. J. Toxicol. 2011, 2011, 686050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Fanali, G.; Cao, Y.; Ascenzi, P.; Fasano, M. Mn(II) binding to human serum albumin: A 1H-NMR relaxometric study. J. Inorg. Biochem. 2012, 117, 198–203. [Google Scholar] [CrossRef] [PubMed]
  247. Yun, Z.; Li, L.; Liu, L.; He, B.; Zhao, X.; Jiang, G. Characterization of mercury-containing protein in human plasma. Metallomics 2013, 5, 821–827. [Google Scholar] [CrossRef] [PubMed]
  248. Briner, W. The Role of Metal Regulatory Proteins in Brain Oxidative Stress: A Tutorial. Oxidative Med. Cell. Longev. 2012, 2012, 981561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Pacini, A.; Branca, J.J.V.; Morucci, G. Cadmium-induced neurotoxicity: Still much ado. Neural Regen. Res. 2018, 13, 1879–1882. [Google Scholar] [CrossRef] [PubMed]
  250. Hussain, A.; Raveendran, V.A.; Kundu, S.; Samanta, T.; Shunmugam, R.; Pal, D.; Sarma, J. Das Mechanisms of Arsenic-Induced Toxicity with Special Emphasis on Arsenic-Binding Proteins. In Arsenic—Analytical and Toxicological Studies; IntechOpen: London, UK, 2018; Available online: https://www.intechopen.com/chapters/60425 (accessed on 3 November 2021). [CrossRef] [Green Version]
  251. Ball, N.; Teo, W.-P.; Chandra, S.; Chapman, J. Parkinson’s Disease and the Environment. Front. Neurol. 2019, 10, 218. [Google Scholar] [CrossRef] [Green Version]
  252. Prabu, S.M.; Shagirtha, K.; Renugadevi, J. Amelioration of Cadmium-Induced Oxidative Stress, Impairment in Lipids and Plasma Lipoproteins by the Combined Treatment with Quercetin and α-Tocopherol in Rats. J. Food Sci. 2010, 75, T132–T140. [Google Scholar] [CrossRef]
  253. Obermeier, B.; Verma, A.; Ransohoff, R.M. The blood-brain barrier. Handb Clin Neurol. 2016, 133, 39–59. [Google Scholar] [CrossRef]
  254. Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.V.; Ganz, T.; Kaplan, J. Hepcidin Regulates Cellular Iron Efflux by Binding to Ferroportin and Inducing Its Internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef] [Green Version]
  255. Crichton, R.; Ward, R. Role of Metal Ions in Brain Function, Metal Transport, Storage and Homoeostasis. In Metal-Based Neurodegeneration; John Wiley and Sons Ltd.: Hoboken, NJ, USA, 2013. [Google Scholar]
  256. Chiou, B.; Neal, E.H.; Bowman, A.B.; Lippmann, E.S.; Simpson, I.A.; Connor, J.R. Endothelial cells are critical regulators of iron transport in a model of the human blood–brain barrier. J. Cereb. Blood Flow Metab. 2018, 39, 2117–2131. [Google Scholar] [CrossRef] [PubMed]
  257. Crossgrove, J.S.; Allen, D.D.; Bukaveckas, B.L.; Rhineheimer, S.S.; Yokel, R.A. Manganese Distribution Across the Blood–Brain Barrier: I. Evidence for Carrier-Mediated Influx of Manganese Citrate as Well as Manganese and Manganese Transferrin. Neurotoxicology 2003, 24, 3–13. [Google Scholar] [CrossRef]
  258. Ye, Q.; Park, J.E.; Gugnani, K.; Betharia, S.; Pino-Figueroa, A.; Kim, J. Influence of iron metabolism on manganese transport and toxicity. Metallomics 2017, 9, 1028–1046. [Google Scholar] [CrossRef]
  259. Karri, V.; Schuhmacher, M.; Kumar, V. Heavy metals (Pb, Cd, As and MeHg) as risk factors for cognitive dysfunction: A general review of metal mixture mechanism in brain. Environ. Toxicol. Pharmacol. 2016, 48, 203–213. [Google Scholar] [CrossRef] [Green Version]
  260. Yukutake, Y.; Tsuji, S.; Hirano, Y.; Adachi, T.; Takahashi, T.; Fujihara, K.; Agre, P.; Yasui, M.; Suematsu, M. Mercury chloride decreases the water permeability of aquaporin-4-reconstituted proteoliposomes. Biol. Cell 2008, 100, 355–363. [Google Scholar] [CrossRef]
  261. Song, H.; Zheng, G.; Liu, Y.; Shen, X.-F.; Zhao, Z.-H.; Aschner, M.; Luo, W.-J.; Chen, J.-Y. Cellular uptake of lead in the blood-cerebrospinal fluid barrier: Novel roles of Connexin 43 hemichannel and its down-regulations via Erk phosphorylation. Toxicol. Appl. Pharmacol. 2016, 297, 1–11. [Google Scholar] [CrossRef] [PubMed]
  262. Branca, J.J.V.; Maresca, M.; Morucci, G.; Mello, T.; Becatti, M.; Pazzagli, L.; Colzi, I.; Gonnelli, C.; Carrino, D.; Paternostro, F.; et al. Effects of Cadmium on ZO-1 Tight Junction Integrity of the Blood Brain Barrier. Int. J. Mol. Sci. 2019, 20, 6010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Chtourou, Y.; Trabelsi, K.; Fetoui, H.; Mkannez, G.; Kallel, H.; Zeghal, N. Manganese Induces Oxidative Stress, Redox State Unbalance and Disrupts Membrane Bound ATPases on Murine Neuroblastoma Cells In Vitro: Protective Role of Silymarin. Neurochem. Res. 2011, 36, 1546–1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell 2019, 18, e13031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Prakash, C.; Soni, M.; Kumar, V. Mitochondrial oxidative stress and dysfunction in arsenic neurotoxicity: A review. J. Appl. Toxicol. 2015, 36, 179–188. [Google Scholar] [CrossRef]
  266. Schneider, C.B.; Donix, M.; Linse, K.; Werner, A.; Fauser, M.; Klingelhoefer, L.; Löhle, M.; Von Kummer, R.; Reichmann, H.; Storch, A.; et al. Accelerated Age-Dependent Hippocampal Volume Loss in Parkinson Disease with Mild Cognitive Impairment. Am. J. Alzheimer Dis. Other Dement. 2017, 32, 313–319. [Google Scholar] [CrossRef]
  267. Lamtai, M.; Zghari, O.; Ouakki, S.; Marmouzi, I.; Mesfioui, A.; El Hessni, A.; Ouichou, A. Chronic copper exposure leads to hippocampus oxidative stress and impaired learning and memory in male and female rats. Toxicol. Res. 2020, 36, 359–366. [Google Scholar] [CrossRef]
  268. Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef] [PubMed]
  269. Rao, R.K.; Li, L.; Baker, R.D.; Baker, S.S.; Gupta, A. Glutathione oxidation and PTPase inhibition by hydrogen peroxide in Caco-2 cell monolayer. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 279, G332–G340. [Google Scholar] [CrossRef]
  270. Jaishankar, M.; Tseten, T.; Anbalagan, N.; Mathew, B.B.; Beeregowda, K.N. Toxicity, mechanism and health effects of some heavy metals. Interdiscip. Toxicol. 2014, 7, 60–72. [Google Scholar] [CrossRef] [Green Version]
  271. Ferruzza, S.; Scacchi, M.; Scarino, M.L.; Sambuy, Y. Iron and copper alter tight junction permeability in human intestinal Caco-2 cells by distinct mechanisms. Toxicol. Vitr. 2002, 16, 399–404. [Google Scholar] [CrossRef]
  272. Valko, M.; Morris, H.; Cronin, M.T.D. Metals, Toxicity and Oxidative Stress. Curr. Med. Chem. 2005, 12, 1161–1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Rao, R. Oxidative stress-induced disruption of epithelial and endothelial tight junctions. Front. Biosci. 2008, 13, 7210–7226. [Google Scholar] [CrossRef] [Green Version]
  274. Bonetto, J.; Villaamil-Lepori, E. Update on the oxidative stress associated with arsenic exposure. Curr. Top. Toxicol. 2014, 10, 37–48. [Google Scholar]
  275. Flora, G.; Gupta, D.; Tiwari, A. Toxicity of lead: A review with recent updates. Interdiscip. Toxicol. 2012, 5, 47–58. [Google Scholar] [CrossRef] [PubMed]
  276. Neal, A.P.; Guilarte, T.R. Mechanisms of lead and manganese neurotoxicity. Toxicol. Res. 2013, 2, 99–114. [Google Scholar] [CrossRef]
  277. Nemmiche, S. Oxidative Signaling Response to Cadmium Exposure. Toxicol. Sci. 2017, 156, 4–10. [Google Scholar] [CrossRef] [Green Version]
  278. Patra, R.C.; Rautray, A.K.; Swarup, D. Oxidative Stress in Lead and Cadmium Toxicity and Its Amelioration. Vet. Med. Int. 2011, 2011, 457327. [Google Scholar] [CrossRef] [Green Version]
  279. Wu, X.; Yalowich, J.C.; Hasinoff, B.B. Cadmium is a catalytic inhibitor of DNA topoisomerase II. J. Inorg. Biochem. 2011, 105, 833–838. [Google Scholar] [CrossRef] [Green Version]
  280. Branco, V.; Canário, J.; Lu, J.; Holmgren, A.; Carvalho, C. Mercury and selenium interaction in vivo: Effects on thioredoxin reductase and glutathione peroxidase. Free. Radic. Biol. Med. 2011, 52, 781–793. [Google Scholar] [CrossRef]
  281. Farina, M.; Avila, D.S.; da Rocha, J.B.T.; Aschner, M. Metals, oxidative stress and neurodegeneration: A focus on iron, manganese and mercury. Neurochem. Int. 2012, 62, 575–594. [Google Scholar] [CrossRef] [Green Version]
  282. Fretham, S.J.B.; Caito, S.; Martinez-Finley, E.J.; Aschner, M. Mechanisms and modifiers of methylmercury-induced neurotoxicity. Toxicol. Res. 2012, 1, 32–38. [Google Scholar] [CrossRef] [Green Version]
  283. Glaser, V.; Leipnitz, G.; Straliotto, M.R.; de Oliveira, J.; Dos Santos, V.V.; Wannmacher, C.M.D.; De Bem, A.F.; da Rocha, J.B.T.; Farina, M.; Latini, A. Oxidative stress-mediated inhibition of brain creatine kinase activity by methylmercury. Neurotoxicology 2010, 31, 454–460. [Google Scholar] [CrossRef]
  284. Mazerik, J.N.; Mikkilineni, H.; Kuppusamy, V.A.; Steinhour, E.; Peltz, A.; Marsh, C.B.; Kuppusamy, P.; Parinandi, N.L. Mercury Activates Phospholipase A2and Induces Formation of Arachidonic Acid Metabolites in Vascular Endothelial Cells. Toxicol. Mech. Methods 2007, 17, 541–557. [Google Scholar] [CrossRef] [PubMed]
  285. Ueda, K.; Makino, R.; Tobe, T.; Okamoto, Y.; Kojima, N. Effects of organic and inorganic mercury(II) on gene expression via DNA conformational changes. Fundam. Toxicol. Sci. 2014, 1, 73–79. [Google Scholar] [CrossRef] [Green Version]
  286. Chen, J.Y.; Tsao, G.C.; Zhao, Q.; Zheng, W. Differential Cytotoxicity of Mn(II) and Mn(III): Special Reference to Mitochondrial [Fe-S] Containing Enzymes. Toxicol. Appl. Pharmacol. 2001, 175, 160–168. [Google Scholar] [CrossRef] [Green Version]
  287. Gunter, T.E.; Gerstner, B.; Lester, T.; Wojtovich, A.P.; Malecki, J.; Swarts, S.G.; Brookes, P.S.; Gavin, C.E.; Gunter, K.K. An analysis of the effects of Mn2+ on oxidative phosphorylation in liver, brain, and heart mitochondria using state 3 oxidation rate assays. Toxicol. Appl. Pharmacol. 2010, 249, 65–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Harischandra, D.S.; Rokad, D.; Neal, M.L.; Ghaisas, S.; Manne, S.; Sarkar, S.; Panicker, N.; Zenitsky, G.; Jin, H.; Lewis, M.; et al. Manganese promotes the aggregation and prion-like cell-to-cell exosomal transmission of α-synuclein. Sci. Signal. 2019, 12, eaau4543. [Google Scholar] [CrossRef]
  289. Zheng, W.; Ren, S.; Graziano, J.H. Manganese inhibits mitochondrial aconitase: A mechanism of manganese neurotoxicity1Published on the World Wide Web on 3 June 1998.1. Brain Res. 1998, 799, 334–342. [Google Scholar] [CrossRef]
  290. Abeyawardhane, D.L.; Lucas, H.R. Iron Redox Chemistry and Implications in the Parkinson’s Disease Brain. Oxidative Med. Cell. Longev. 2019, 2019, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  291. Joppe, K.; Roser, A.-E.; Maass, F.; Lingor, P. The Contribution of Iron to Protein Aggregation Disorders in the Central Nervous System. Front. Neurosci. 2019, 13, 15. [Google Scholar] [CrossRef] [Green Version]
  292. Zhao, Z. Iron and oxidizing species in oxidative stress and Alzheimer’s disease. Aging Med. 2019, 2, 82–87. [Google Scholar] [CrossRef]
  293. Garza-Lombo, C.; Pappa, A.; Panayiotidis, M.I.; Gonsebatt, M.E.; Franco, R. Arsenic-induced neurotoxicity: A mechanistic appraisal. JBIC J. Biol. Inorg. Chem. 2019, 24, 1305–1316. [Google Scholar] [CrossRef]
  294. Tamás, M.J.; Sharma, S.K.; Ibstedt, S.; Jacobson, T.; Christen, P. Heavy Metals and Metalloids As a Cause for Protein Misfolding and Aggregation. Biomolecules 2014, 4, 252–267. [Google Scholar] [CrossRef] [Green Version]
  295. Kwon, H.S.; Koh, S.-H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
  296. Sanders, K.M.; Ward, S.M. Nitric oxide and its role as a non-adrenergic, non-cholinergic inhibitory neurotransmitter in the gastrointestinal tract. Br. J. Pharmacol. 2019, 176, 212–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. Friedland, R.P.; Chapman, M.R. The role of microbial amyloid in neurodegeneration. PLoS Pathog. 2017, 13, e1006654. [Google Scholar] [CrossRef]
  298. Tschopp, J.; Schroder, K. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 2010, 10, 210–215. [Google Scholar] [CrossRef] [PubMed]
  299. Lee, Y.K.; Menezes, J.S.; Umesaki, Y.; Mazmanian, S.K. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 2010, 108, 4615–4622. [Google Scholar] [CrossRef] [Green Version]
  300. Kustrimovic, N.; Comi, C.; Magistrelli, L.; Rasini, E.; Legnaro, M.; Bombelli, R.; Aleksic, I.; Blandini, F.; Minafra, B.; Riboldazzi, G.; et al. Parkinson’s disease patients have a complex phenotypic and functional Th1 bias: Cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naïve and drug-treated patients. J. Neuroinflammation 2018, 15, 205. [Google Scholar] [CrossRef] [PubMed]
  301. Wong, M.-L.; Inserra, A.; Lewis, M.D.; Mastronardi, C.A.; Leong, L.E.X.; Choo, J.; Kentish, S.; Xie, P.; Morrison, M.; Wesselingh, S.L.; et al. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol. Psychiatry 2016, 21, 797–805. [Google Scholar] [CrossRef] [PubMed]
  302. Banan, A.; Fields, J.Z.; Zhang, Y.; Keshavarzian, A. iNOS upregulation mediates oxidant-induced disruption of F-actin and barrier of intestinal monolayers. Am. J. Physiol. Liver Physiol. 2001, 280, G1234–G1246. [Google Scholar] [CrossRef]
  303. Byndloss, M.X.; Olsan, E.E.; Rivera-Chávez, F.; Tiffany, C.R.; Cevallos, S.A.; Lokken, K.L.; Torres, T.P.; Byndloss, A.J.; Faber, F.; Gao, Y.; et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [Google Scholar] [CrossRef] [PubMed]
  304. Hu, Y.; Chen, D.; Zheng, P.; Yu, J.; He, J.; Mao, X.; Yu, B. The Bidirectional Interactions between Resveratrol and Gut Microbiota: An Insight into Oxidative Stress and Inflammatory Bowel Disease Therapy. BioMed Res. Int. 2019, 2019, 5403761. [Google Scholar] [CrossRef]
  305. Hawkes, C.H.; Del Tredici, K.; Braak, H. A timeline for Parkinson’s disease. Parkinsonism Relat. Disord. 2010, 16, 79–84. [Google Scholar] [CrossRef]
  306. Dos Santos, A.A.; Ferrer, B.; Gonçalves, F.M.; Tsatsakis, A.M.; Renieri, E.A.; Skalny, A.V.; Farina, M.; Rocha, J.B.T.; Aschner, M. Oxidative Stress in Methylmercury-Induced Cell Toxicity. Toxics 2018, 6, 47. [Google Scholar] [CrossRef] [Green Version]
  307. Fernandes, J.; Hao, L.; Bijli, K.M.; Chandler, J.D.; Orr, M.; Hu, X.; Jones, D.P.; Go, Y.-M. From the Cover: Manganese Stimulates Mitochondrial H2O2Production in SH-SY5Y Human Neuroblastoma Cells OVER Physiologic as well as Toxicologic Range. Toxicol. Sci. 2017, 155, 213–223. [Google Scholar] [CrossRef] [Green Version]
  308. Fernandes, J.; Chandler, J.D.; Lili, L.N.; Uppal, K.; Hu, X.; Hao, L.; Go, Y.-M.; Jones, D.P. Transcriptome Analysis Reveals Distinct Responses to Physiologic versus Toxic Manganese Exposure in Human Neuroblastoma Cells. Front. Genet. 2019, 10, 676. [Google Scholar] [CrossRef] [Green Version]
  309. Sidoryk-Wegrzynowicz, M.; Aschner, M. Manganese toxicity in the central nervous system: The glutamine/glutamate-γ-aminobutyric acid cycle. J. Intern. Med. 2013, 273, 466–477. [Google Scholar] [CrossRef] [Green Version]
  310. Ke, T.; Sidoryk-Wegrzynowicz, M.; Pajarillo, E.; Rizor, A.; Soares, F.A.A.; Lee, E.; Aschner, M. Role of Astrocytes in Manganese Neurotoxicity Revisited. Neurochem. Res. 2019, 44, 2449–2459. [Google Scholar] [CrossRef] [PubMed]
  311. Raj, K.; Kaur, P.; Gupta, G.D.; Singh, S. Metals associated neurodegeneration in Parkinson’s disease: Insight to physiological, pathological mechanisms and management. Neurosci. Lett. 2021, 753, 135873. [Google Scholar] [CrossRef]
  312. Azevedo, B.F.; Furieri, L.B.; Peçanha, F.M.; Wiggers, G.; Vassallo, P.F.; Simões, M.R.; Fiorim, J.; De Batista, P.R.; Fioresi, M.; Rossoni, L.; et al. Toxic Effects of Mercury on the Cardiovascular and Central Nervous Systems. J. Biomed. Biotechnol. 2012, 2012, 949048. [Google Scholar] [CrossRef]
  313. Segura-Aguilar, J.; Metodiewa, D.; Welch, C.J. Metabolic activation of dopamine o-quinones to o-semiquinones by NADPH cytochrome P450 reductase may play an important role in oxidative stress and apoptotic effects. Biochim. Biophys. Acta Gen. Subj. 1998, 1381, 1–6. [Google Scholar] [CrossRef]
  314. Desole, M.S.; Esposito, G.; Migheli, R.; Fresu, L.; Sircana, S.; Zangani, D.; Miele, M.; Miele, E. Cellular defence mechanisms in the striatum of young and aged rats subchronically exposed to manganese. Neuropharmacology 1995, 34, 289–295. [Google Scholar] [CrossRef]
  315. Serra, P.A.; Esposito, G.; Enrico, P.; Mura, M.A.; Migheli, R.; Delogu, M.R.; Miele, M.; Desole, M.S.; Grella, G.; Miele, E. Manganese increases L-DOPA auto-oxidation in the striatum of the freely moving rat: Potential implications to L-DOPA long-term therapy of Parkinson’s disease. Br. J. Pharmacol. 2000, 130, 937–945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Desole, M.S.; Sciola, G.L.; Delogu, M.R.; Sircana, S.; Migheli, R. Manganese and 1-methyl-4-(2′-ethylphenyl)-1,2,3,6-tetrahydropyridine induce apoptosis in PC12 cells. Neurosci. Lett. 1996, 209, 193–196. [Google Scholar] [CrossRef]
  317. Fitsanakis, V.A.; Aschner, M. The importance of glutamate, glycine, and γ-aminobutyric acid transport and regulation in manganese, mercury and lead neurotoxicity. Toxicol. Appl. Pharmacol. 2005, 204, 343–354. [Google Scholar] [CrossRef]
  318. Racette, B.A.; Nielsen, S.S.; Criswell, S.R.; Sheppard, L.; Seixas, N.; Warden, M.N.; Checkoway, H. Dose-dependent progression of parkinsonism in manganese-exposed welders. Neurology 2017, 88, 344–351. [Google Scholar] [CrossRef] [Green Version]
  319. Block, M.L.; Zecca, L.; Hong, J.-S. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nat. Rev. Neurosci. 2007, 8, 57–69. [Google Scholar] [CrossRef] [PubMed]
  320. Hirsch, E.C.; Hunot, S. Neuroinflammation in Parkinson’s disease: A target for neuroprotection? Lancet Neurol. 2009, 8, 382–397. [Google Scholar] [CrossRef]
  321. Tiwari, P.C.; Pal, R. The potential role of neuroinflammation and transcription factors in Parkinson disease. Dialogues Clin. Neurosci. 2017, 19, 71–80. [Google Scholar] [CrossRef]
  322. Moos, T.; Nielsen, T.R.; Skjørringe, T.; Morgan, E.H. Iron trafficking inside the brain. J. Neurochem. 2007, 103, 1730–1740. [Google Scholar] [CrossRef] [PubMed]
  323. Neal, A.P.; Guilarte, T.R. Molecular neurobiology of lead (Pb2+): Effects on synaptic function. Mol. Neurobiol. 2010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Florea, A.-M.; Taban, J.; Varghese, E.; Alost, B.T.; Moreno, S.; Büsselberg, D. Lead (Pb2+) neurotoxicity: Ion-mimicry with calcium (Ca2+) impairs synaptic transmission. A review with animated illustrations of the pre- and post-synaptic effects of lead. J. Local Glob. Heal. Sci. 2013. [Google Scholar] [CrossRef]
  325. Chater, T.E.; Goda, Y. The role of AMPA receptors in postsynaptic mechanisms of synaptic plasticity. Front. Cell. Neurosci. 2014. [Google Scholar] [CrossRef] [PubMed]
  326. Hanisch, U.-K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  327. Nagatsu, T.; Mogi, M.; Ichinose, H.; Togari, A.; Riederer, P. Cytokines in Parkinson’s disease. Neurosci. News 1999, 58, 143–151. [Google Scholar]
  328. Kebir, H.; Kreymborg, K.; Ifergan, I.; Dodelet-Devillers, A.; Cayrol, R.; Bernard, M.; Giuliani, F.; Arbour, N.; Becher, B.; Prat, A. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat. Med. 2007, 13, 1173–1175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Dardalhon, V.; Korn, T.; Kuchroo, V.K.; Anderson, A.C. Role of Th1 and Th17 cells in organ-specific autoimmunity. J. Autoimmun. 2008, 31, 252–256. [Google Scholar] [CrossRef] [Green Version]
  330. Barcia, C.; Ros, F.; Annese, V.; Gómez, C.M.R.; Bernal, F.R.; Aguado-Llera, D.; Pagán, M.E.M.; De Pablos, V.; Villalba, E.F.; Herrero, M.T. IFN-γ signaling, with the synergistic contribution of TNF-α, mediates cell specific microglial and astroglial activation in experimental models of Parkinson’s disease. Cell Death Dis. 2011, 2, e142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Tanaka, T.; Kai, S.; Matsuyama, T.; Adachi, T.; Fukuda, K.; Hirota, K. General Anesthetics Inhibit LPS-Induced IL-1β Expression in Glial Cells. PLoS ONE 2013, 8, e82930. [Google Scholar] [CrossRef] [Green Version]
  332. Wang, Q.; Liu, Y.; Zhou, J. Neuroinflammation in Parkinson’s disease and its potential as therapeutic target. Transl. Neurodegener. 2015, 4, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Troncoso-Escudero, P.; Parra, A.; Nassif, M.; Vidal, R.L. Outside in: Unraveling the Role of Neuroinflammation in the Progression of Parkinson’s Disease. Front. Neurol. 2018, 9, 860. [Google Scholar] [CrossRef] [PubMed]
  334. Pajarillo, E.; Nyarko-Danquah, I.; Adinew, G.; Rizor, A.; Aschner, M.; Lee, E. Neurotoxicity Mechanisms of Manganese in the Central Nervous System; Elsevier: Amsterdam, The Netherlands, 2021. [Google Scholar]
  335. Li, B.; Xia, M.; Zorec, R.; Parpura, V.; Verkhratsky, A. Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain Res. 2021, 1752, 147234. [Google Scholar] [CrossRef]
  336. Johnson, J.; Pajarillo, E.A.B.; Taka, E.; Reams, R.; Son, D.S.; Aschner, M.; Lee, E. Valproate and sodium butyrate attenuate manganese-decreased locomotor activity and astrocytic glutamate transporters expression in mice. Neurotoxicology 2018, 64, 230–239. [Google Scholar] [CrossRef] [Green Version]
  337. Deng, Y.; Xu, Z.; Xu, B.; Xu, D.; Tian, Y.; Feng, W. The protective effects of riluzole on manganese-induced disruption of glutamate transporters and glutamine synthetase in the cultured astrocytes. Biol. Trace Elem. Res. 2012, 148, 242–249. [Google Scholar] [CrossRef]
  338. Gu, X.-L.; Long, C.-X.; Sun, L.; Xie, C.; Lin, X.; Cai, H. Astrocytic expression of Parkinson’s disease-related A53T α-synuclein causes neurodegeneration in mice. Mol. Brain 2010, 3, 12. [Google Scholar] [CrossRef] [Green Version]
  339. Gelders, G.; Baekelandt, V.; Van Der Perren, A. Linking Neuroinflammation and Neurodegeneration in Parkinson’s Disease. J. Immunol. Res. 2018, 2018, 4784268. [Google Scholar] [CrossRef] [Green Version]
  340. Clarkson, T.W. The three modern faces of mercury. Environ. Health Perspect. 2002, 110, 11–23. [Google Scholar] [CrossRef] [Green Version]
  341. Rooney, J.P.K. The role of thiols, dithiols, nutritional factors and interacting ligands in the toxicology of mercury. Toxicology 2007, 234, 145–156. [Google Scholar] [CrossRef] [PubMed]
  342. Atchison, W.D.; Hare, M.F. Mechanisms of methylmercury-induced neurotoxicity. FASEB J. 1994, 8, 622–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Olivieri, G.; Brack, C.; Müller-Spahn, F.; Stähelin, H.B.; Herrmann, M.; Renard, P.; Brockhaus, M.; Hock, C. Mercury Induces Cell Cytotoxicity and Oxidative Stress and Increases β-Amyloid Secretion and Tau Phosphorylation in SHSY5Y Neuroblastoma Cells. J. Neurochem. 2000, 74, 231–236. [Google Scholar] [CrossRef]
  344. Kim, S.H.; Johnson, V.J.; Sharma, R.P. Mercury inhibits nitric oxide production but activates proinflammatory cytokine expression in murine macrophage: Differential modulation of NF-κB and p38 MAPK signaling pathways. Nitric Oxide 2002, 7, 67–74. [Google Scholar] [CrossRef]
  345. Fonnum, F.; Lock, E.A. The contributions of excitotoxicity, glutathione depletion and DNA repair in chemically induced injury to neurones: Exemplified with toxic effects on cerebellar granule cells. J. Neurochem. 2003, 88, 513–531. [Google Scholar] [CrossRef] [Green Version]
  346. Castoldi, A.F.; Barni, S.; Turin, I.; Gandini, C.; Manzo, L. Early acute necrosis, delayed apoptosis and cytoskeletal breakdown in cultured cerebellar granule neurons exposed to methylmercury. J. Neurosci. Res. 2000, 59, 775–787. [Google Scholar] [CrossRef]
  347. Wang, X.; Yan, M.; Zhao, L.; Wu, Q.; Wu, C.; Chang, X.; Zhou, Z. Low-Dose Methylmercury-Induced Apoptosis and Mitochondrial DNA Mutation in Human Embryonic Neural Progenitor Cells. Oxidative Med. Cell. Longev. 2016, 2016, 5137042. [Google Scholar] [CrossRef] [Green Version]
  348. Perry, T.L.; Godin, D.V.; Hansen, S. Parkinson’s disease: A disorder due to nigral glutathione deficiency? Neurosci. Lett. 1982, 33, 305–310. [Google Scholar] [CrossRef]
  349. Serru, V.; Baudin, B.; Ziegler, F.; David, J.-P.; Cals, M.-J.; Vaubourdolle, M.; Mario, N. Quantification of Reduced and Oxidized Glutathione in Whole Blood Samples by Capillary Electrophoresis. Clin. Chem. 2001, 47, 1321–1324. [Google Scholar] [CrossRef]
  350. Hsu, M.; Srinivas, B.; Kumar, J.K.; Subramanian, R.; Andersen, J. Glutathione depletion resulting in selective mitochondrial complex I inhibition in dopaminergic cells is via an NO-mediated pathway not involving peroxynitrite: Implications for Parkinson’s disease: Glutathione Affects Mitochondrial Complex I via NO Not ONOO. J. Neurochem. 2005, 92, 1091–1103. [Google Scholar] [CrossRef]
  351. Ballatori, N.; Krance, S.M.; Notenboom, S.; Shi, S.; Tieu, K.; Hammond, C.L. Glutathione dysregulation and the etiology and progression of human diseases. Biol. Chem. 2009, 390, 191–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  352. Götz, M.E.; Double, K.; Gerlach, M.; Youdim, M.B.H.; Riederere, P. The Relevance of Iron in the Pathogenesis of Parkinson’s Disease. Ann. N. Y. Acad. Sci. 2004, 1012, 193–208. [Google Scholar] [CrossRef]
  353. Hare, D.J.; Lei, P.; Ayton, S.; Roberts, B.R.; Grimm, R.; George, J.L.; Bishop, D.P.; Beavis, A.D.; Donovan, S.J.; McColl, G.; et al. An iron–dopamine index predicts risk of parkinsonian neurodegeneration in the substantia nigra pars compacta. Chem. Sci. 2014, 5, 2160–2169. [Google Scholar] [CrossRef] [Green Version]
  354. Hare, D.J.; Double, K.L. Iron and dopamine: A toxic couple. Brain 2016, 139, 1026–1035. [Google Scholar] [CrossRef] [PubMed]
  355. Guiney, S.J.; Adlard, P.A.; Bush, A.I.; Finkelstein, D.I.; Ayton, S. Ferroptosis and cell death mechanisms in Parkinson’s disease. Neurochem. Int. 2017, 104, 34–48. [Google Scholar] [CrossRef]
  356. Ma, L.; Azad, M.G.; Dharmasivam, M.; Richardson, V.; Quinn, R.J.; Feng, Y.; Pountney, D.L.; Tonissen, K.F.; Mellick, G.D.; Yanatori, I.; et al. Parkinson’s disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol. 2021, 41, 101896. [Google Scholar] [CrossRef]
  357. Collins, L.M.; Toulouse, A.; Connor, T.J.; Nolan, Y.M. Contributions of central and systemic inflammation to the pathophysiology of Parkinson’s disease. Neuropharmacology 2012, 62, 2154–2168. [Google Scholar] [CrossRef] [Green Version]
  358. Hare, D.J.; Ayton, S.; Bush, A.I.; Lei, P. A delicate balance: Iron metabolism and diseases of the brain. Front. Aging Neurosci. 2013, 5, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  359. Urrutia, P.; Aguirre, P.; Esparza, A.; Tapia, V.; Mena, N.P.; Arredondo, M.; González-Billault, C.; Núñez, M.T. Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells. J. Neurochem. 2013, 126, 541–549. [Google Scholar] [CrossRef] [PubMed]
  360. Niranjan, R. The Role of Inflammatory and Oxidative Stress Mechanisms in the Pathogenesis of Parkinson’s Disease: Focus on Astrocytes. Mol. Neurobiol. 2013, 49, 28–38. [Google Scholar] [CrossRef]
  361. Lian, T.-H.; Guo, P.; Zuo, L.-J.; Hu, Y.; Yu, S.-Y.; Yu, Q.-J.; Jin, Z.; Wang, R.-D.; Li, L.-X.; Zhang, W. Tremor-Dominant in Parkinson Disease: The Relevance to Iron Metabolism and Inflammation. Front. Neurosci. 2019, 13, 255. [Google Scholar] [CrossRef]
  362. Kortekaas, R.; Leenders, K.L.; Van Oostrom, J.C.H.; Vaalburg, W.; Bart, J.; Willemsen, A.T.M.; Hendrikse, N.H. Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann. Neurol. 2005, 57, 176–179. [Google Scholar] [CrossRef] [PubMed]
  363. Liu, J.T.; Dong, M.H.; Zhang, J.Q.; Bai, Y.; Kuang, F.; Chen, L.W. Microglia and astroglia: The role of neuroinflammation in lead toxicity and neuronal injury in the brain. Neuroimmunol. Neuroinflamm. 2015, 2, 131–137. [Google Scholar] [CrossRef] [Green Version]
  364. Liu, X.; Su, P.; Meng, S.; Aschner, M.; Cao, Y.; Luo, W.; Zheng, G.; Liu, M. Role of matrix metalloproteinase-2/9 (MMP2/9) in lead-induced changes in an in vitro blood-brain barrier model. Int. J. Biol. Sci. 2017, 13, 1351–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  365. Mangia, S.; Simpson, I.A.; Vannucci, S.J.; Carruthers, A. The in vivo neuron-to-astrocyte lactate shuttle in human brain: Evidence from modeling of measured lactate levels during visual stimulation. J. Neurochem. 2009, 109, 55–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  366. Jozefczak, M.; Remans, T.; Vangronsveld, J.; Cuypers, A. Glutathione Is a Key Player in Metal-Induced Oxidative Stress Defenses. Int. J. Mol. Sci. 2012, 13, 3145–3175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  367. Cabezas, R.; Fidel, M.; Torrente, D.; El-Bach, R.S.; Morales, L.; Gonzalez, J.; Barreto, G.E. Astrocytes Role in Parkinson: A Double-Edged Sword. Neurodegener. Dis. 2013. [Google Scholar] [CrossRef] [Green Version]
  368. Kiela, P.R.; Ghishan, F.K. Physiology of Intestinal Absorption and Secretion. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 145–159. [Google Scholar] [CrossRef] [Green Version]
  369. Onakpa, M.M.; Njan, A.A.; Kalu, O.C. A Review of Heavy Metal Contamination of Food Crops in Nigeria. Ann. Glob. Health 2018, 84, 488–494. [Google Scholar] [CrossRef] [Green Version]
  370. Chi, L.; Bian, X.; Gao, B.; Tu, P.; Ru, H.; Lu, K. The Effects of an Environmentally Relevant Level of Arsenic on the Gut Microbiome and Its Functional Metagenome. Toxicol. Sci. 2017, 160, 193–204. [Google Scholar] [CrossRef]
  371. Sears, M.E. Chelation: Harnessing and Enhancing Heavy Metal Detoxification—A Review. Sci. World J. 2013, 2013, 219840. [Google Scholar] [CrossRef] [Green Version]
  372. Bisanz, J.E.; Enos, M.K.; Mwanga, J.R.; Changalucha, J.P.; Burton, J.; Gloor, G.B.; Reid, G. Randomized Open-Label Pilot Study of the Influence of Probiotics and the Gut Microbiome on Toxic Metal Levels in Tanzanian Pregnant Women and School Children. mBio 2014, 5, e01580-14. [Google Scholar] [CrossRef] [Green Version]
  373. Astolfi, M.L.; Protano, C.; Schiavi, E.; Marconi, E.; Capobianco, D.; Massimi, L.; Ristorini, M.; Baldassarre, M.E.; Laforgia, N.; Vitali, M.; et al. A prophylactic multi-strain probiotic treatment to reduce the absorption of toxic elements: In-vitro study and biomonitoring of breast milk and infant stools. Environ. Int. 2019, 130, 104818. [Google Scholar] [CrossRef] [PubMed]
  374. Raghuvanshi, R.; Chaudhari, A.; Kumar, G.N. Amelioration of cadmium- and mercury-induced liver and kidney damage in rats by genetically engineered probiotic Escherichia coli Nissle 1917 producing pyrroloquinoline quinone with oral supplementation of citric acid. Nutrition 2016, 32, 1285–1294. [Google Scholar] [CrossRef] [PubMed]
  375. Zeitoun, M.; Al-Damegh, M.; Abdel-Salam, A. The Role of Fermented Milk Containing Probiotic, Dandelion as Prebiotic or their Combination on Serum Metabolites, Enzymes, Testosterone and Testicular Histopathology of Arsenic-Intoxicated Male Rats. J. Basic Appl. Sci. 2014, 10, 492–503. [Google Scholar] [CrossRef]
  376. Gerbino, E.; Carasi, P.; Tymczyszyn, E.E.; Gómez-Zavaglia, A. Removal of cadmium by Lactobacillus kefir as a protective tool against toxicity. J. Dairy Res. 2014, 81, 280–287. [Google Scholar] [CrossRef] [PubMed]
  377. Jama, A.M.; Ćulafić, D.M.; Kolarevic, S.; Djurasevic, S.; Knezevic-Vukcevic, J. Protective effect of probiotic bacteria against cadmium-induced genotoxicity in rat hepatocytes in vivo and in vitro. Arch. Biol. Sci. 2012, 64, 1197–1206. [Google Scholar] [CrossRef]
  378. Djurasevic, S.; Jama, A.; Jasnic, N.; Vujovic, P.; Jovanovic, M.; Ćulafić, D.M.; Knežević-Vukčević, J.; Cakic-Milosevic, M.; Ilijevic, K.; Djordjevic, J. The Protective Effects of Probiotic Bacteria on Cadmium Toxicity in Rats. J. Med. Food 2017, 20, 189–196. [Google Scholar] [CrossRef]
  379. Song, S.; Oh, S.; Lim, K.-T. Lactobacillus plantarum L67 glycoprotein protects against cadmium chloride toxicity in RAW 264.7 clls. J. Dairy Sci. 2016, 99, 1812–1821. [Google Scholar] [CrossRef] [Green Version]
  380. Zhai, Q.; Tian, F.; Zhao, J.; Zhang, H.; Narbad, A.; Chen, W. Oral Administration of Probiotics Inhibits Absorption of the Heavy Metal Cadmium by Protecting the Intestinal Barrier. Appl. Environ. Microbiol. 2016, 82, 4429–4440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  381. Abd-El-Moneim, O.M.; Abd El-Kader, H.A.; Abd El-Rahim, A.H.; Radwan, H.A.; Fadel, M.; Farag, I.M. Modulatory role of Saccharomyces cerevisiae against cadmium-induced genotoxicity in mice. J. Arab Soc. Med. Res. 2017, 12, 27–38. [Google Scholar] [CrossRef]
  382. Jafarpour, D.; Shekarforoush, S.S.; Ghaisari, H.R.; Nazifi, S.; Sajedianfard, J.; Eskandari, M.H. Protective effects of synbiotic diets of Bacillus coagulans, Lactobacillus plantarum and inulin against acute cadmium toxicity in rats. BMC Complement. Altern. Med. 2017, 17, 291. [Google Scholar] [CrossRef] [PubMed]
  383. Kadry, M.O.; Megeed, R.A. Probiotics as a Complementary Therapy in the Model of Cadmium Chloride Toxicity: Crosstalk of β-Catenin, BDNF, and StAR Signaling Pathways. Biol. Trace Elem. Res. 2018, 185, 404–413. [Google Scholar] [CrossRef] [PubMed]
  384. Allam, N.; Ali, E.; Shabanna, S.; Abd-Elrahman, E. Protective efficacy of Streptococcus thermophilus against acute cadmium toxicity in mice. Iran. J. Pharm. Res. 2018, 17, 695–707. [Google Scholar] [CrossRef]
  385. Wang, N.; Jiang, M.; Zhang, P.; Shu, H.; Li, Y.; Guo, Z.; Li, Y. Amelioration of Cd-induced bioaccumulation, oxidative stress and intestinal microbiota by Bacillus cereus in Carassius auratus gibelio. Chemosphere 2020, 245, 125613. [Google Scholar] [CrossRef]
  386. Zhai, Q.; Yu, L.; Li, T.; Zhu, J.; Zhang, C.; Zhao, J.; Zhang, H.; Chen, W. Effect of dietary probiotic supplementation on intestinal microbiota and physiological conditions of Nile tilapia (Oreochromis niloticus) under waterborne cadmium exposure. Antonie van Leeuwenhoek Int. J. Gen. Mol. Microbiol. 2017, 110, 501–513. [Google Scholar] [CrossRef] [PubMed]
  387. Abu-Braka, A.Z.; Zaki, M.S.; Abbas, H.H.; Ismail, N.E.D.A.; Khalil, R.; Tanekhy, M.; Saad, T. Filed studies on some probiotics to minimize hazard effects of prevailing heavy metals contamination for improving immunity and growth performance of Oreochromis niloticus. Electron. Physician 2017, 9, 4138. [Google Scholar] [CrossRef] [Green Version]
  388. Feng, S.; Liu, Y.; Huang, Y.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Influence of oral administration of Akkermansia muciniphila on the tissue distribution and gut microbiota composition of acute and chronic cadmium exposure mice. FEMS Microbiol. Lett. 2019, 366, fnz160. [Google Scholar] [CrossRef]
  389. Tian, F.; Zhai, Q.; Zhao, J.; Liu, X.; Wang, G.; Zhang, H.; Zhang, H.; Chen, W. Lactobacillus plantarum CCFM8661 Alleviates Lead Toxicity in Mice. Biol. Trace Elem. Res. 2012, 150, 264–271. [Google Scholar] [CrossRef]
  390. Zhai, Q.; Liu, Y.; Wang, C.; Qu, D.; Zhao, J.; Zhang, H.; Tian, F.; Chen, W. Lactobacillus plantarum CCFM8661 modulates bile acid enterohepatic circulation and increases lead excretion in mice. Food Funct. 2019, 10, 1455–1464. [Google Scholar] [CrossRef]
  391. Zhai, Q.; Qu, D.; Feng, S.; Yu, Y.; Yu, L.; Tian, F.; Zhao, J.; Zhang, H.; Chen, W. Oral Supplementation of Lead-Intolerant Intestinal Microbes Protects Against Lead (Pb) Toxicity in Mice. Front. Microbiol. 2020, 10, 3161. [Google Scholar] [CrossRef]
  392. Zhai, Q.; Wang, G.; Zhao, J.; Liu, X.; Tian, F.; Zhang, H.; Chen, W. Protective Effects of Lactobacillus plantarum CCFM8610 against Acute Cadmium Toxicity in Mice. Appl. Environ. Microbiol. 2013, 79, 1508–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  393. Giri, S.S.; Yun, S.; Jun, J.W.; Kim, H.J.; Kim, S.G.; Kang, J.W.; Kim, S.W.; Han, S.J.; Sukumaran, V.; Park, S.C. Therapeutic Effect of Intestinal Autochthonous Lactobacillus reuteri P16 Against Waterborne Lead Toxicity in Cyprinus carpio. Front. Immunol. 2018, 9, 1824. [Google Scholar] [CrossRef]
  394. Ghenioa, A.; Okle, O.; Nazem, A.; Ashry, K.M. Protective Effect of Probiotic Bactosac Against Induced Sub Chronic Lead Toxicity in Broiler Chicks. Alex. J. Vet. Sci. 2015, 47, 53–64. [Google Scholar] [CrossRef]
  395. Li, B.; Jin, D.; Yu, S.; Evivie, S.E.; Muhammad, Z.; Huo, G.; Liu, F. In Vitro and In Vivo Evaluation of Lactobacillus delbrueckii subsp. bulgaricus KLDS1.0207 for the Alleviative Effect on Lead Toxicity. Nutrients 2017, 9, 845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  396. Jiang, X.; Gu, S.; Liu, D.; Zhao, L.; Xia, S.; He, X.; Chen, H.; Ge, J. Lactobacillus brevis 23017 Relieves Mercury Toxicity in the Colon by Modulation of Oxidative Stress and Inflammation Through the Interplay of MAPK and NF-κB Signaling Cascades. Front. Microbiol. 2018, 9, 2425. [Google Scholar] [CrossRef] [PubMed]
  397. Majlesi, M.; Shekarforoush, S.S.; Ghaisari, H.R.; Nazifi, S.; Sajedianfard, J.; Eskandari, M.H. Effect of Probiotic Bacillus Coagulans and Lactobacillus Plantarum on Alleviation of Mercury Toxicity in Rat. Probiotics Antimicrob. Proteins 2017, 9, 300–309. [Google Scholar] [CrossRef] [PubMed]
  398. Abdel-Salam, A.M.; Al-Dekheil, A.; Babkr, A.; Farahna, M.; Mousa, H.M. High fiber probiotic fermented mare’s milk reduces the toxic effects of mercury in rats. N. Am. J. Med Sci. 2010, 2, 569–575. [Google Scholar] [CrossRef] [PubMed]
  399. Shao, M.; Zhu, Y. Long-term metal exposure changes gut microbiota of residents surrounding a mining and smelting area. Sci. Rep. 2020, 10, 4453. [Google Scholar] [CrossRef] [PubMed]
  400. Hattori, Y.; Naito, M.; Satoh, M.; Nakatochi, M.; Naito, H.; Kato, M.; Takagi, S.; Matsunaga, T.; Seiki, T.; Sasakabe, T.; et al. MetallothioneinMT2AA-5G Polymorphism as a Risk Factor for Chronic Kidney Disease and Diabetes: Cross-Sectional and Cohort Studies. Toxicol. Sci. 2016, 152, 181–193. [Google Scholar] [CrossRef] [Green Version]
  401. Nair, A.R.; Lee, W.-K.; Smeets, K.; Swennen, Q.; Sanchez, A.; Thévenod, F.; Cuypers, A. Glutathione and mitochondria determine acute defense responses and adaptive processes in cadmium-induced oxidative stress and toxicity of the kidney. Arch. Toxicol. 2014, 89, 2273–2289. [Google Scholar] [CrossRef]
  402. Mozzi, F.; Raya, R.R.; Vignolo, G.M. Biotechnology of Lactic Acid Bacteria: Novel Applications; John Wiley & Sons: Hoboken, NJ, USA, 2010; ISBN 9780813815831. [Google Scholar]
  403. Wedajo, B. Lactic Acid Bacteria: Benefits, Selection Criteria and Probiotic Potential in Fermented Food. J. Probiotics Health 2015, 3, 1–9. [Google Scholar] [CrossRef]
  404. Ameen, F.A.; Hamdan, A.M.; El-Naggar, M.Y. Assessment of the heavy metal bioremediation efficiency of the novel marine lactic acid bacterium, Lactobacillus plantarum MF042018. Sci. Rep. 2020, 10, 314. [Google Scholar] [CrossRef] [PubMed]
  405. Wu, G.; Xiao, X.; Feng, P.; Xie, F.; Yu, Z.; Yuan, W.; Liu, P.; Li, X. Gut remediation: A potential approach to reducing chromium accumulation using Lactobacillus plantarum TW1-1. Sci. Rep. 2017, 7, 15000. [Google Scholar] [CrossRef] [Green Version]
  406. Chandran, H.; Meena, M.; Sharma, K. Microbial Biodiversity and Bioremediation Assessment through Omics Approaches. Front. Environ. Chem. 2020, 1, 9. [Google Scholar] [CrossRef]
  407. Cassani, E.; Privitera, G.; Pezzoli, G.; Pusani, C.; Madio, C.; Iorio, L.; Barichella, M. Use of probiotics for the treatment of constipation in Parkinson’s disease patients. Minerva Gastroenterol. Dietol. 2011, 57, 117–121. [Google Scholar] [PubMed]
  408. Tamtaji, O.R.; Taghizadeh, M.; Kakhaki, R.D.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 1031–1035. [Google Scholar] [CrossRef]
  409. Tamtaji, O.R.; Kouchaki, E.; Salami, M.; Aghadavod, E.; Akbari, E.; Tajabadi-Ebrahimi, M.; Asemi, Z. The Effects of Probiotic Supplementation on Gene Expression Related to Inflammation, Insulin, and Lipids in Patients With Multiple Sclerosis: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Am. Coll. Nutr. 2017, 36, 660–665. [Google Scholar] [CrossRef]
  410. Georgescu, D.; Ancusa, O.E.; Georgescu, L.A.; Ionita, I.; Reisz, D. Nonmotor gastrointestinal disorders in older patients with Parkinson’s disease: Is there hope? Clin. Interv. Aging 2016, 11, 1601–1608. [Google Scholar] [CrossRef] [Green Version]
  411. Barichella, M.; Pacchetti, C.; Bolliri, C.; Cassani, E.; Iorio, L.; Pusani, C.; Pinelli, G.; Privitera, G.; Cesari, I.; Faierman, S.A.; et al. Probiotics and prebiotic fiber for constipation associated with Parkinson disease. Neurology 2016, 87, 1274–1280. [Google Scholar] [CrossRef]
  412. Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello, R.; Pane, M.; Comi, C. Probiotics May Have Beneficial Effects in Parkinson’s Disease: In vitro Evidence. Front. Immunol. 2019, 10, 969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  413. Srivastav, S.; Neupane, S.; Bhurtel, S.; Katila, N.; Maharjan, S.; Choi, H.; Hong, J.T.; Choi, D.-Y. Probiotics mixture increases butyrate, and subsequently rescues the nigral dopaminergic neurons from MPTP and rotenone-induced neurotoxicity. J. Nutr. Biochem. 2019, 69, 73–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  414. Hsieh, T.-H.; Kuo, C.-W.; Hsieh, K.-H.; Shieh, M.-J.; Peng, C.-W.; Chen, Y.-C.; Chang, Y.-L.; Huang, Y.-Z.; Chen, C.-C.; Chang, P.-K.; et al. Probiotics Alleviate the Progressive Deterioration of Motor Functions in a Mouse Model of Parkinson’s Disease. Brain Sci. 2020, 10, 206. [Google Scholar] [CrossRef] [Green Version]
  415. Sjögren, Y.M.; Tomicic, S.; Lundberg, A.; Böttcher, M.F.; Björkstén, B.; Sverremark-Ekström, E.; Jenmalm, M.C. Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses. Gut microbiota and immune responses. Clin. Exp. Allergy 2009, 39, 1842–1851. [Google Scholar] [CrossRef] [Green Version]
  416. Fujimura, K.E.; Lynch, S.V. Microbiota in Allergy and Asthma and the Emerging Relationship with the Gut Microbiome. Cell Host Microbe 2015, 17, 592–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  417. Zoghi, A.; Khosravi-Darani, K.; Sohrabvandi, S. Surface Binding of Toxins and Heavy Metals by Probiotics. Mini-Rev. Med. Chem. 2014, 14, 84–98. [Google Scholar] [CrossRef] [PubMed]
  418. Pieper, D.H.; Reineke, W. Engineering bacteria for bioremediation. Curr. Opin. Biotechnol. 2000, 11, 262–270. [Google Scholar] [CrossRef]
  419. Bhakta, J.N.; Ohnishi, K.; Munekage, Y.; Iwasaki, K.; Wei, M.Q. Characterization of lactic acid bacteria-based probiotics as potential heavy metal sorbents. J. Appl. Microbiol. 2012, 112, 1193–1206. [Google Scholar] [CrossRef]
  420. Patel, A.; SV, A.; Shah, N.; Verma, D.K. Lactic Acid Bacteria As Metal Quenchers To Improve Food Safety and Quality. AgroLife 2017.
  421. Halttunen, T.; Collado, M.C.; El-Nezami, H.; Meriluoto, J.; Salminen, S. Combining strains of lactic acid bacteria may reduce their toxin and heavy metal removal efficiency from aqueous solution. Lett. Appl. Microbiol. 2008, 46, 160–165. [Google Scholar] [CrossRef]
  422. Monachese, M.; Burton, J.P.; Reid, G. Bioremediation and tolerance of humans to heavy metals through microbial processes: A potential role for probiotics? Appl. Environ. Microbiol. 2012, 78, 6397–6404. [Google Scholar] [CrossRef] [Green Version]
  423. Beeler, E.; Singh, O.V. Extremophiles as sources of inorganic bio-nanoparticles. World J. Microbiol. Biotechnol. 2016, 32, 156. [Google Scholar] [CrossRef]
  424. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [Green Version]
  425. Liu, Y.; Li, Y.; Liu, K.; Shen, J. Exposing to Cadmium Stress Cause Profound Toxic Effect on Microbiota of the Mice Intestinal Tract. PLoS ONE 2014, 9, e85323. [Google Scholar] [CrossRef]
  426. Roberfroid, M.; Gibson, G.R.; Hoyles, L.; McCartney, A.L.; Rastall, R.; Rowland, I.; Wolvers, D.; Watzl, B.; Szajewska, H.; Stahl, B.; et al. Prebiotic effects: Metabolic and health benefits. Br. J. Nutr. 2010, 104, S1–S63. [Google Scholar] [CrossRef] [Green Version]
  427. Tian, F.; Xiao, Y.; Li, X.; Zhai, Q.; Wang, G.; Zhang, Q.; Zhang, H.; Chen, W. Protective Effects of Lactobacillus plantarum CCFM8246 against Copper Toxicity in Mice. PLoS ONE 2015, 10, e0143318. [Google Scholar] [CrossRef]
  428. Zhai, Q.; Wang, G.; Zhao, J.; Liu, X.; Narbad, A.; Chen, Y.Q.; Zhang, H.; Tian, F.; Chen, W. Protective Effects of Lactobacillus plantarum CCFM8610 against Chronic Cadmium Toxicity in Mice Indicate Routes of Protection besides Intestinal Sequestration. Appl. Environ. Microbiol. 2014, 80, 4063–4071. [Google Scholar] [CrossRef] [Green Version]
  429. Walter, J. Ecological Role of Lactobacilli in the Gastrointestinal Tract: Implications for Fundamental and Biomedical Research. Appl. Environ. Microbiol. 2008, 74, 4985–4996. [Google Scholar] [CrossRef] [Green Version]
  430. D’Antona, G.; Ragni, M.; Cardile, A.; Tedesco, L.; Dossena, M.; Bruttini, F.; Caliaro, F.; Corsetti, G.; Bottinelli, R.; Carruba, M.O.; et al. Branched-Chain Amino Acid Supplementation Promotes Survival and Supports Cardiac and Skeletal Muscle Mitochondrial Biogenesis in Middle-Aged Mice. Cell Metab. 2010, 12, 362–372. [Google Scholar] [CrossRef] [Green Version]
  431. Valerio, A.; D’Antona, G.; Nisoli, E. Branched-chain amino acids, mitochondrial biogenesis, and healthspan: An evolutionary perspective. Aging 2011, 3, 464–478. [Google Scholar] [CrossRef] [Green Version]
  432. Sridharan, G.V.; Choi, K.; Klemashevich, C.; Wu, C.; Prabakaran, D.; Bin Pan, L.; Steinmeyer, S.; Mueller, C.; Yousofshahi, M.; Alaniz, R.C.; et al. Prediction and quantification of bioactive microbiota metabolites in the mouse gut. Nat. Commun. 2014, 5, 5492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  433. Oliphant, K.; Allen-Vercoe, E. Macronutrient metabolism by the human gut microbiome: Major fermentation by-products and their impact on host health. Microbiome 2019, 7, 91. [Google Scholar] [CrossRef] [PubMed]
  434. Novarino, G.; El-Fishawy, P.; Kayserili, H.; Meguid, N.A.; Scott, E.M.; Schroth, J.; Silhavy, J.L.; Kara, M.; Khalil, R.O.; Ben-Omran, T.; et al. Mutations in BCKD-kinase Lead to a Potentially Treatable Form of Autism with Epilepsy. Science 2012, 338, 394–397. [Google Scholar] [CrossRef] [Green Version]
  435. Mally, J.; Szalai, G.; Stone, T.W. Changes in the concentration of amino acids in serum and cerebrospinal fluid of patients with Parkinson’s disease. J. Neurol. Sci. 1997, 151, 159–162. [Google Scholar] [CrossRef]
  436. Molina, J.A.; Jiménez-Jiménez, F.J.; Gómez, P.; Vargas, C.; Navarro, J.A.; Ortí-Pareja, M.; Gasalla, T.; Benito-León, J.; Bermejo, F.; Arenas, J. Decreased cerebrospinal fluid levels of neutral and basic amino acids in patients with Parkinson’s disease. J. Neurol. Sci. 1997, 150, 123–127. [Google Scholar] [CrossRef]
  437. Yan, Z.; Yang, F.; Cao, J.; Ding, W.; Yan, S.; Shi, W.; Wen, S.; Yao, L. Alterations of gut microbiota and metabolome with Parkinson’s disease. Microb. Pathog. 2021, 160, 105187. [Google Scholar] [CrossRef] [PubMed]
  438. Wuolikainen, A.; Jonsson, P.; Ahnlund, M.; Antti, H.; Marklund, S.L.; Moritz, T.; Forsgren, L.; Andersen, P.M.; Trupp, M. Multi-platform mass spectrometry analysis of the CSF and plasma metabolomes of rigorously matched amyotrophic lateral sclerosis, Parkinson’s disease and control subjects. Mol. BioSyst. 2016, 12, 1287–1298. [Google Scholar] [CrossRef]
  439. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [Green Version]
  440. Zheng, H.; Xu, P.; Jiang, Q.; Xu, Q.; Zheng, Y.; Yan, J.; Ji, H.; Ning, J.; Zhang, X.; Li, C.; et al. Correction to: Depletion of acetate-producing bacteria from the gut microbiota facilitates cognitive impairment through the gut-brain neural mechanism in diabetic mice. Microbiome 2021, 9, 164. [Google Scholar] [CrossRef]
  441. Shehata, M.G.; El Sohaimy, S.A.; El-Sahn, M.A.; Youssef, M.M. Screening of isolated potential probiotic lactic acid bacteria for cholesterol lowering property and bile salt hydrolase activity. Ann. Agric. Sci. 2016, 61, 65–75. [Google Scholar] [CrossRef] [Green Version]
  442. Liu, S.; Marcelin, G.; Blouet, C.; Jeong, J.H.; Jo, Y.-H.; Schwartz, G.J.; Chua, S. A gut–brain axis regulating glucose metabolism mediated by bile acids and competitive fibroblast growth factor actions at the hypothalamus. Mol. Metab. 2017, 8, 37–50. [Google Scholar] [CrossRef]
  443. Lyte, M. Host-microbiota neuroendocrine interactions influencing brain and behavior. Gut Microbes 2014, 5, 381–389. [Google Scholar] [CrossRef] [Green Version]
  444. Erikson, K.M.; Aschner, M. Manganese neurotoxicity and glutamate-GABA interaction. Neurochem. Int. 2003, 43, 475–480. [Google Scholar] [CrossRef]
  445. Nyarko-Danquah, I.; Pajarillo, E.; Digman, A.; Soliman, K.F.A.; Aschner, M.; Lee, E. Manganese Accumulation in the Brain via Various Transporters and Its Neurotoxicity Mechanisms. Molecules 2020, 25, 5880. [Google Scholar] [CrossRef]
  446. Gros, D.F.; Antony, M.M.; McCabe, R.E.; Swinson, R.P. Frequency and severity of the symptoms of irritable bowel syndrome across the anxiety disorders and depression. J. Anxiety Disord. 2009, 23, 290–296. [Google Scholar] [CrossRef]
  447. Kim, S.-H.; Field, K.G.; Morrissey, M.T.; Price, R.J.; Wei, C.-I.; An, H. Source and Identification of Histamine-Producing Bacteria from Fresh and Temperature-Abused Albacore. J. Food Prot. 2001, 64, 1035–1044. [Google Scholar] [CrossRef]
  448. Smolinska, S.; Jutel, M.; Crameri, R.; O’Mahony, L. Histamine and gut mucosal immune regulation. Allergy 2013, 69, 273–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  449. Ferstl, R.; Frei, R.; Schiavi, E.; Konieczna, P.; Barcik, W.; Ziegler, M.; Lauener, R.P.; Chassard, C.; Lacroix, C.; Akdis, C.A.; et al. Histamine receptor 2 is a key influence in immune responses to intestinal histamine-secreting microbes. J. Allergy Clin. Immunol. 2014, 134, 744–746. [Google Scholar] [CrossRef] [PubMed]
  450. Karri, V.; Schuhmacher, M.; Kumar, V. A systems toxicology approach to compare the heavy metal mixtures (Pb, As, MeHg) impact in neurodegenerative diseases. Food Chem. Toxicol. 2020, 139, 111257. [Google Scholar] [CrossRef]
  451. Stilling, R.M.; Moloney, G.M.; Ryan, F.J.; Hoban, A.E.; Bastiaanssen, T.F.S.; Shanahan, F.; Clarke, G.; Claesson, M.J.; Dinan, T.G.; Cryan, J.F. Social interaction-induced activation of RNA splicing in the amygdala of microbiome-deficient mice. eLife 2018, 7, e33070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  452. Tilocca, B.; Pieroni, L.; Soggiu, A.; Britti, D.; Bonizzi, L.; Roncada, P.; Greco, V. Gut–Brain Axis and Neurodegeneration: State-of-the-Art of Meta-Omics Sciences for Microbiota Characterization. Int. J. Mol. Sci. 2020, 21, 4045. [Google Scholar] [CrossRef]
  453. Wanichthanarak, K.; Fahrmann, J.F.; Grapov, D. Genomic, Proteomic, and Metabolomic Data Integration Strategies. Biomark. Insights 2015, 10s4, BMI.S29511–BMI.S29516. [Google Scholar] [CrossRef] [Green Version]
  454. Zhang, L.; Shimoji, M.; Thomas, B.; Moore, D.J.; Yu, S.-W.; Marupudi, N.I.; Torp, R.; Torgner, I.A.; Ottersen, O.P.; Dawson, T.M.; et al. Mitochondrial localization of the Parkinson’s disease related protein DJ-1: Implications for pathogenesis. Hum. Mol. Genet. 2005, 14, 2063–2073. [Google Scholar] [CrossRef] [Green Version]
  455. Schellenberger, J.; Que, R.; Fleming, R.M.T.; Thiele, I.; Orth, J.D.; Feist, A.M.; Zielinski, D.C.; Bordbar, A.; Lewis, N.E.; Rahmanian, S.; et al. Quantitative prediction of cellular metabolism with constraint-based models: The COBRA Toolbox v2.0. Nat. Protoc. 2011, 6, 1290–1307. [Google Scholar] [CrossRef] [Green Version]
  456. Mo, M.L.; Jamshidi, N.; Palsson, B. A genome-scale, constraint-based approach to systems biology of human metabolism. Mol. BioSyst. 2007, 3, 598–603. [Google Scholar] [CrossRef]
  457. Orth, J.D.; Thiele, I.; Palsson, B.Ø. What is flux balance analysis? Nat. Biotechnol. 2010, 28, 245–248. [Google Scholar] [CrossRef]
  458. Thiele, I.; Sahoo, S.; Heinken, A.; Hertel, J.; Heirendt, L.; Aurich, M.K.; Fleming, R.M. Personalized whole-body models integrate metabolism, physiology, and the gut microbiome. Mol. Syst. Biol. 2020, 16, e8982. [Google Scholar] [CrossRef]
  459. Lu, K.; Abo, R.P.; Schlieper, K.A.; Graffam, M.E.; Levine, S.; Wishnok, J.S.; Swenberg, J.A.; Tannenbaum, S.R.; Fox, J.G. Arsenic Exposure Perturbs the Gut Microbiome and Its Metabolic Profile in Mice: An Integrated Metagenomics and Metabolomics Analysis. Environ. Health Perspect. 2014, 122, 284–291. [Google Scholar] [CrossRef]
  460. Richardson, J.B.; Dancy, B.C.R.; Horton, C.L.; Lee, Y.S.; Madejczyk, M.S.; Xu, Z.Z.; Ackermann, G.; Humphrey, G.; Palacios, G.; Knight, R.; et al. Exposure to toxic metals triggers unique responses from the rat gut microbiota. Sci. Rep. 2018, 8, 6578. [Google Scholar] [CrossRef]
  461. Latha, S.; Vinothini, G.; Dhanasekaran, D. Chromium [Cr(VI)] biosorption property of the newly isolated actinobacterial probiont Streptomyces werraensis LD22. 3 Biotech 2014, 5, 423–432. [Google Scholar] [CrossRef] [Green Version]
  462. Chi, L.; Gao, B.; Bian, X.; Tu, P.; Ru, H.; Lu, K. Manganese-induced sex-specific gut microbiome perturbations in C57BL/6 mice. Toxicol. Appl. Pharmacol. 2017, 331, 142–153. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Bacterial metabolites signaling in the intestine. (A) Secondary bile acids, SCFAs and tryptophan metabolites are microbially derived metabolites. Secondary bile acids activate FXR and TGR5, which stimulate different types of cells. Enterochromaffin cells are stimulated promoting 5-HT release and motility in the colon. Enteroendocrine L-cells promote glucose tolerance through release of the incretin GLP-1. Enteric neurons with TGR5 stimulate or inhibit motility [128,129,130]. Bacterial fermentation of dietary fibers leads to the reduction in luminal pH and production of SCFAs [131]. The bacterial-produced SCFAs activate G-protein-coupled receptors (e.g., GPR41, GPR43 and GPR109) on enteroendocrine cells, enterochromaffin cells, and enteric neurons, leading to the increased production of GLP-1 and 5-HT, thus directing changes in gut motility [132]. With regards to colonocytes, anionic-SCFAs enter the cell through a carrier-mediated transport (similar to MCT1), promoting of mitochondrial fatty acid beta-oxidation and reduction in luminal availability of oxygen through PPARγ activation [131,132]. Finally, the reduction in oxygen availability upregulates the expression of tight junction proteins which are important for the maintenance of the gut barrier [132,133]. (B) Under dysbiosis, there is a limited availability of SCFAs, leading to a decreased amount of available substrate for the colonocytes. This leads to a decreased activation of PPARγ and less oxygen uptake in colonocytes causing dysregulation of cellular growth and differentiation, which can be described as a metabolic switch towards anaerobic glycolysis and lactate production. When the concentration of oxygen increases, decreases in the expression of HIF1 lowers tight junction expression, thus further degenerating the gut barrier [131,132,133,134,135,136]. Furthermore, pathogenic interaction with the impaired cells increases their translocation to the lamina propria, thus promoting oxidative stress events and chronic inflammation [133,134,135,136]. Abbreviations: 5-HT, 5-hydroxytryptamine; FXR, farnesoid X receptor; GI, gastrointestinal; GLP-1, glucagon-like peptide-1; HIF1, hypoxia-inducible factor 1; PPARγ, proliferator-activated receptor gamma; TGR5, Takeda G protein-coupled receptor 5.
Figure 1. Bacterial metabolites signaling in the intestine. (A) Secondary bile acids, SCFAs and tryptophan metabolites are microbially derived metabolites. Secondary bile acids activate FXR and TGR5, which stimulate different types of cells. Enterochromaffin cells are stimulated promoting 5-HT release and motility in the colon. Enteroendocrine L-cells promote glucose tolerance through release of the incretin GLP-1. Enteric neurons with TGR5 stimulate or inhibit motility [128,129,130]. Bacterial fermentation of dietary fibers leads to the reduction in luminal pH and production of SCFAs [131]. The bacterial-produced SCFAs activate G-protein-coupled receptors (e.g., GPR41, GPR43 and GPR109) on enteroendocrine cells, enterochromaffin cells, and enteric neurons, leading to the increased production of GLP-1 and 5-HT, thus directing changes in gut motility [132]. With regards to colonocytes, anionic-SCFAs enter the cell through a carrier-mediated transport (similar to MCT1), promoting of mitochondrial fatty acid beta-oxidation and reduction in luminal availability of oxygen through PPARγ activation [131,132]. Finally, the reduction in oxygen availability upregulates the expression of tight junction proteins which are important for the maintenance of the gut barrier [132,133]. (B) Under dysbiosis, there is a limited availability of SCFAs, leading to a decreased amount of available substrate for the colonocytes. This leads to a decreased activation of PPARγ and less oxygen uptake in colonocytes causing dysregulation of cellular growth and differentiation, which can be described as a metabolic switch towards anaerobic glycolysis and lactate production. When the concentration of oxygen increases, decreases in the expression of HIF1 lowers tight junction expression, thus further degenerating the gut barrier [131,132,133,134,135,136]. Furthermore, pathogenic interaction with the impaired cells increases their translocation to the lamina propria, thus promoting oxidative stress events and chronic inflammation [133,134,135,136]. Abbreviations: 5-HT, 5-hydroxytryptamine; FXR, farnesoid X receptor; GI, gastrointestinal; GLP-1, glucagon-like peptide-1; HIF1, hypoxia-inducible factor 1; PPARγ, proliferator-activated receptor gamma; TGR5, Takeda G protein-coupled receptor 5.
Antioxidants 11 00071 g001
Figure 2. Neuronal and immunity crosstalk. (A) Under homeostasis, macrophages mature within the intestinal mucosa, where they are utilized to capture invading pathogens, recognize antigens, and clear old apoptotic cells within the area. Macrophages then transfer the captured antigens to the dendritic cells, which are responsible for entering the mesenteric lymph nodes to induce the differentiation of peripheral T regulatory (Treg) cells from naïve T cells[161]. These cells constitutively produce IL-10, which promotes the secondary expansion of regulatory T cells in the mucosa and maintains the homeostasis [161,162]. Butyrate is important in the modulation of intestinal macrophages via the inhibition of histone deacetylases and NF-kB, and thus downregulates proinflammatory responses such as IL-6, IL-12 and NO [163,164]. On the other hand, neuron enteric cells are essential for gastrointestinal motility. The Toll-like receptors (TLRs) expressed by enteric neurons can recognize gut microbe-derived signals and influence the gut motility[165]. These cells promote crosstalk with the Muscularis Macrophages (MMs) in longitudinal and circular smooth muscles and incite the production of the growth factor bone morphogenetic protein 2 (BMP2) to stimulate motility that is directed by the neurons. In turn, the enteric neurons through the production of macrophage survival factor CSF1 promote the maintenance of the MM [152,165,166]. (B) Under dysbiotic conditions, the epithelial barrier is impaired; this in turn leads to the translocation of microbiota, toxins, and deleterious exposures, as suggested by Braak’s Hypothesis, thereby leading to the activation of the host immune response. Macrophages then produce proinflammatory cytokines and present antigens to dendritic cells; together, both cells induce the expansion of TH1 and TH17, thereby recruiting other innate effector cells such as neutrophils and eosinophils [162,163,164]. Additionally, excess TLR activation by the microbiota or toxins, activates cytokine release, proinflammatory responses, and apoptosis in enteric neurons. Thus, MMs decrease the expression of BMP2, and enteric neurons decrease the expression of CSF1 and MM motility[152,166]. Enteric bacteria can further influence the development and differentiation of CD4+and CD8+ T cells, as well as B cell activity and IgA production [167,168]. Abbreviations: BMP2, bone morphogenetic protein 2; CSF1, colony stimulating factor 1; Interferon γ (IFN-γ); TLRs, Toll-like receptors; TNF-α, tumor necrosis factor-alpha.
Figure 2. Neuronal and immunity crosstalk. (A) Under homeostasis, macrophages mature within the intestinal mucosa, where they are utilized to capture invading pathogens, recognize antigens, and clear old apoptotic cells within the area. Macrophages then transfer the captured antigens to the dendritic cells, which are responsible for entering the mesenteric lymph nodes to induce the differentiation of peripheral T regulatory (Treg) cells from naïve T cells[161]. These cells constitutively produce IL-10, which promotes the secondary expansion of regulatory T cells in the mucosa and maintains the homeostasis [161,162]. Butyrate is important in the modulation of intestinal macrophages via the inhibition of histone deacetylases and NF-kB, and thus downregulates proinflammatory responses such as IL-6, IL-12 and NO [163,164]. On the other hand, neuron enteric cells are essential for gastrointestinal motility. The Toll-like receptors (TLRs) expressed by enteric neurons can recognize gut microbe-derived signals and influence the gut motility[165]. These cells promote crosstalk with the Muscularis Macrophages (MMs) in longitudinal and circular smooth muscles and incite the production of the growth factor bone morphogenetic protein 2 (BMP2) to stimulate motility that is directed by the neurons. In turn, the enteric neurons through the production of macrophage survival factor CSF1 promote the maintenance of the MM [152,165,166]. (B) Under dysbiotic conditions, the epithelial barrier is impaired; this in turn leads to the translocation of microbiota, toxins, and deleterious exposures, as suggested by Braak’s Hypothesis, thereby leading to the activation of the host immune response. Macrophages then produce proinflammatory cytokines and present antigens to dendritic cells; together, both cells induce the expansion of TH1 and TH17, thereby recruiting other innate effector cells such as neutrophils and eosinophils [162,163,164]. Additionally, excess TLR activation by the microbiota or toxins, activates cytokine release, proinflammatory responses, and apoptosis in enteric neurons. Thus, MMs decrease the expression of BMP2, and enteric neurons decrease the expression of CSF1 and MM motility[152,166]. Enteric bacteria can further influence the development and differentiation of CD4+and CD8+ T cells, as well as B cell activity and IgA production [167,168]. Abbreviations: BMP2, bone morphogenetic protein 2; CSF1, colony stimulating factor 1; Interferon γ (IFN-γ); TLRs, Toll-like receptors; TNF-α, tumor necrosis factor-alpha.
Antioxidants 11 00071 g002
Figure 3. Human exposure to heavy metals in the diet underlies neurodegenerative diseases. Multiple levels of environmental exposure to heavy metals, which are derived from pollution, lead to adverse effects on the gut and brain in humans.
Figure 3. Human exposure to heavy metals in the diet underlies neurodegenerative diseases. Multiple levels of environmental exposure to heavy metals, which are derived from pollution, lead to adverse effects on the gut and brain in humans.
Antioxidants 11 00071 g003
Figure 4. Gut–microbiome metal uptake. (A) Mechanisms of microbial heavy metal bioremediation in the gut: 1. Biosorption 2. Bioaccumulation, 3. Biotransformation [216]. (B) Schematic of intestinal metal transport: The primary mechanisms by which iron and other divalent metals (Pb2+, Hg2+, Fe2+, Mn2+) are taken up by the enterocyte is through DMT-1 from the luminal membrane[217]. In the case of Fe3+, this metal is reduced to Fe2+ by DYCTB [217]. Other receptors for Zn2+ (ZIP, ZnT) [201,218,219,220,221,222], Ca2+ and amino acids transporters (b0,+, PepT1) [201,223,224] are utilized to uptake MeHg or other metal ions. Iron was also suggested to be transported with heme with HCP1 and as ferritin [225]. Once entering the intracellular space, metals are readily stored as ferritin (Fe2+) with metallothioneins and exported into the body circulation through ferroportin or other transporters (LAT, ZIP, ZnT) [201,218,219,220,221,222,226,227,228]. Ferritin is converted to Fe3+ by ferroxidase, which is then released for use by transferrin, other metals are transported by albumin or bound to other cell-derived proteins [201,229]. Abbreviations: HCP-1, Heme carrier protein; Znt, Zinc transporter; ZIP, RT, IRT-like protein; LAT, L-type/large neutral amino acid transporter; MRPs, Multidrug Resistance-Related Protein; DMT-1, Divalent metal transporter 1; DYCTB, Duodenal cytochrome B.
Figure 4. Gut–microbiome metal uptake. (A) Mechanisms of microbial heavy metal bioremediation in the gut: 1. Biosorption 2. Bioaccumulation, 3. Biotransformation [216]. (B) Schematic of intestinal metal transport: The primary mechanisms by which iron and other divalent metals (Pb2+, Hg2+, Fe2+, Mn2+) are taken up by the enterocyte is through DMT-1 from the luminal membrane[217]. In the case of Fe3+, this metal is reduced to Fe2+ by DYCTB [217]. Other receptors for Zn2+ (ZIP, ZnT) [201,218,219,220,221,222], Ca2+ and amino acids transporters (b0,+, PepT1) [201,223,224] are utilized to uptake MeHg or other metal ions. Iron was also suggested to be transported with heme with HCP1 and as ferritin [225]. Once entering the intracellular space, metals are readily stored as ferritin (Fe2+) with metallothioneins and exported into the body circulation through ferroportin or other transporters (LAT, ZIP, ZnT) [201,218,219,220,221,222,226,227,228]. Ferritin is converted to Fe3+ by ferroxidase, which is then released for use by transferrin, other metals are transported by albumin or bound to other cell-derived proteins [201,229]. Abbreviations: HCP-1, Heme carrier protein; Znt, Zinc transporter; ZIP, RT, IRT-like protein; LAT, L-type/large neutral amino acid transporter; MRPs, Multidrug Resistance-Related Protein; DMT-1, Divalent metal transporter 1; DYCTB, Duodenal cytochrome B.
Antioxidants 11 00071 g004
Figure 5. Heavy metals and oxidative stress in neuronal cells: the disruption of BBB through metal neurotoxicity and molecular mimicry. (A) Mn, Fe and other toxic metals accumulate within the mitochondria by utilizing the MCU. An increase in oxidative stress insults leads to a dysfunction of the mitochondrial electron transport chain. Metals that are able to bind to ADP via ATP-synthase uniquely uncouples oxidative phosphorylation and ATP formation within the mitochondria. Pb2 has an inhibitory effect on the activity of calmodulin and, consequently, leads to the avoidance of synaptic vesicles release [311,312,313,314,319,320,321,322]. (B) In the neuronal synapses, PB, Cd, and Mn impair Ca2+ channels (voltage-gated Ca2+ and Ca2+ ATPase) leading to Ca2+ ion avoidance and alterations in ion signaling. Pb2 has an inhibitory effect on the activity of calmodulin and consequently leads to the avoidance of synaptic vesicle release [323,324]. In the binding sites of postsynaptic cells at the NMDA-receptor/channel, and AMPA disturbances of the postsynaptic potential, leads to the synaptic plasticity and induction of LTP[323,324,325]. (C) Glial cells further increase the production of ROS by mitochondrial damage. Specifically, alterations in antioxidant levels and metal reactions further promote the activation of the NF-kB pathway by the release of proinflammatory cytokines (TNF-α, IL-1, IL-6). Increased cellular damage associated with the accumulation of metals promotes α -synuclein formation in astrocytes leads to the suppression of protective functions and a decrease in glutamate uptake, leading to excitotoxicity in neurons [326,327,328,329,330,331,332,333]. (D) In endothelial cells, the increase in ROS production promotes the release of metalloproteinase-9 and PGE-2 by endothelial cells. This factor then stimulates both pericytes and glial cells to further promote pro-inflammatory signals and, as a consequence, degrades the extracellular matrix of both tight junctions and BBB permeability [41,273,283]. (E) The interaction of glial cells with ROS leads to mitochondrial damage and the release of proinflammatory cytokines, which are further bolstered by the additional burden of heavy metals. This impairs the essential roles of neuron maintenance by disrupting glutamate/GABA-glutamine shuttling. The expression of EAAT1 and EAAT2 and the activity of glutamine synthetase can be downregulated by the excess of intra-astroglial heavy metals [334,335,336,337]. Thus, glutamine catabolism and elevated extracellular glutamate further induces excitotoxicity and neuronal damage, finally leading to neurodegeneration[334,338,339]. Abbreviations: AMPA: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, DMT1: Divalent metal transporter 1, DAT: dopamine active transporter, EAAT: excitatory amino acid transporter, IL-1: Interleukin-1, IL-6: In-terleukin-6, IL-12: Interleukin-12, JAM: junctional adhesion molecule, LTP: Long term potentiation, MCU: mitochondrial calcium uniporter, MMP-9/-3: metalloproteinase-9/-3, MRP: Multidrug resistance-associated proteins, MTs: metallothioneins, NMDA: N-Methyl-D-Aspartic acid, NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase, NF-κB: nuclear factor kappa, PGE-2: Prostaglandin E2, PECAM: Platelet/endothelial cell adhesion mole-cule-1, ROS: Reactive oxygen species, TfR: Transferrin receptor, TNF: Tumor Necrosis Factor, VE-CADHERINE: vascular endothelial cadherin, ZIP: Zinc-imidazolate polymers, ZnT1: zinc transporter protein-1.
Figure 5. Heavy metals and oxidative stress in neuronal cells: the disruption of BBB through metal neurotoxicity and molecular mimicry. (A) Mn, Fe and other toxic metals accumulate within the mitochondria by utilizing the MCU. An increase in oxidative stress insults leads to a dysfunction of the mitochondrial electron transport chain. Metals that are able to bind to ADP via ATP-synthase uniquely uncouples oxidative phosphorylation and ATP formation within the mitochondria. Pb2 has an inhibitory effect on the activity of calmodulin and, consequently, leads to the avoidance of synaptic vesicles release [311,312,313,314,319,320,321,322]. (B) In the neuronal synapses, PB, Cd, and Mn impair Ca2+ channels (voltage-gated Ca2+ and Ca2+ ATPase) leading to Ca2+ ion avoidance and alterations in ion signaling. Pb2 has an inhibitory effect on the activity of calmodulin and consequently leads to the avoidance of synaptic vesicle release [323,324]. In the binding sites of postsynaptic cells at the NMDA-receptor/channel, and AMPA disturbances of the postsynaptic potential, leads to the synaptic plasticity and induction of LTP[323,324,325]. (C) Glial cells further increase the production of ROS by mitochondrial damage. Specifically, alterations in antioxidant levels and metal reactions further promote the activation of the NF-kB pathway by the release of proinflammatory cytokines (TNF-α, IL-1, IL-6). Increased cellular damage associated with the accumulation of metals promotes α -synuclein formation in astrocytes leads to the suppression of protective functions and a decrease in glutamate uptake, leading to excitotoxicity in neurons [326,327,328,329,330,331,332,333]. (D) In endothelial cells, the increase in ROS production promotes the release of metalloproteinase-9 and PGE-2 by endothelial cells. This factor then stimulates both pericytes and glial cells to further promote pro-inflammatory signals and, as a consequence, degrades the extracellular matrix of both tight junctions and BBB permeability [41,273,283]. (E) The interaction of glial cells with ROS leads to mitochondrial damage and the release of proinflammatory cytokines, which are further bolstered by the additional burden of heavy metals. This impairs the essential roles of neuron maintenance by disrupting glutamate/GABA-glutamine shuttling. The expression of EAAT1 and EAAT2 and the activity of glutamine synthetase can be downregulated by the excess of intra-astroglial heavy metals [334,335,336,337]. Thus, glutamine catabolism and elevated extracellular glutamate further induces excitotoxicity and neuronal damage, finally leading to neurodegeneration[334,338,339]. Abbreviations: AMPA: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, DMT1: Divalent metal transporter 1, DAT: dopamine active transporter, EAAT: excitatory amino acid transporter, IL-1: Interleukin-1, IL-6: In-terleukin-6, IL-12: Interleukin-12, JAM: junctional adhesion molecule, LTP: Long term potentiation, MCU: mitochondrial calcium uniporter, MMP-9/-3: metalloproteinase-9/-3, MRP: Multidrug resistance-associated proteins, MTs: metallothioneins, NMDA: N-Methyl-D-Aspartic acid, NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase, NF-κB: nuclear factor kappa, PGE-2: Prostaglandin E2, PECAM: Platelet/endothelial cell adhesion mole-cule-1, ROS: Reactive oxygen species, TfR: Transferrin receptor, TNF: Tumor Necrosis Factor, VE-CADHERINE: vascular endothelial cadherin, ZIP: Zinc-imidazolate polymers, ZnT1: zinc transporter protein-1.
Antioxidants 11 00071 g005
Table 1. Molecular mimicry of metals throughout the gut–brain axis.
Table 1. Molecular mimicry of metals throughout the gut–brain axis.
TransportersMolecule/Ion Metal Being MimickedMetal ReplacementCells Containing the Transporters of the TransporterCitation
Organic anion transporters: CH3Hg+Endothelial cells/Glial cells/Enterocytes[201,230,231]
OAT1GSH
CysteineGS-Cd-S-G
OAT3
CH3Hg-S-Cys
Zinc-regulated zinc transporter 1 (hZTL1)Zn 2+Cd2+Enterocytes/Neurons[218,219,220]
Ca2+ channelsCa2+Cd2+Neurons/Endothelial cells/glial cells[201,223,224]
Pb2+
Mn2+
Divalent metal transporter 1Fe2+Pb2+Enterocytes/Endothelial cells/Neurons/Glial cells[217]
DMT-1Mn2+
Cd2+
Zinc-imidazolate polymers (ZIPs)Zn 2+Pb2+Enterocytes/Endothelial/Astrocytes[201,221,222]
1.2Mn2+
8, 14Cd2+
Hg2+
Transferrin receptorFe2+Mn2+Neurons/Endothelial cells/Glial cells[229]
TfR
Amino acid transporters (system b0,+, system L)CysteineCH3Hg-S-CysEnterocytes/Endothelial cells/Glial cells[201,226,227,228]
MethionineCys-S-Hg-S-Cys
Cys-S-Cd-S-Cys
CH3Hg-S-CysGly
Multidrug resistance-associated proteins CH3Hg+Enterocytes/Endothelial cells/glial cells[201,232]
MRP 1, 2, 3,4GSSGG-S-Cd-S-G
GSHCH3Hg-S-G
As (III)
As–GSH
FerroportinFe2+Cd2+Enterocytes/Endothelial cells/Neurons/Oligodendrocytes/Astrocytes[201,233,234,235]
Mn2+
Glucose permeasesGlucoseAs (III)Enterocytes/Endothelial cells/Astrocytes[210]
GLUT 1, 2, 5
Sodium-dependent phosphate transportersPhosphateAs(V)Enterocytes[218,219,220,236,237]
NaPiIIb
AquaporinsGlycerolAs(III) [218,219,220,238,239,240]
AQP 3, 10WaterHg2+Enterocytes/Enteric neurons/Endothelial cells/Astrocytes
AQP 4 Pb2+
Fe2+
Mn2+
Organic anion transporting polypeptidesAmphipathic organic compoundsAs(III)Enterocytes/Astrocytes/Endothelial[218,219,220]
OATPB
Table 2. Metal ions and ROS/NOS adversely oxidize neuroproteins and lipid metabolism.
Table 2. Metal ions and ROS/NOS adversely oxidize neuroproteins and lipid metabolism.
Metal IonReactive SpeciesOxidized MoleculesAdverse OutcomesCite
LeadPb2+δ-aminolevulinic acid dehydratase (ALAD)Reduces the antioxidant (glutathione) levels[275,276]
Hydrogen peroxide Superoxide radicalSuperoxide dismutase (SOD)Oxidative stress
Hydroxyl radicalsCatalaseAlteration in Ca2+ influx
Glucose-6-phosphate dehydrogenase (G6PD)Apoptosis
GSH
Glutathione reductase
Glutathione peroxidase
Glutathione s-transferase
Voltage-gated calcium (Ca2+) channels
N-methyl-d-aspartate (NMDA)
CadmiumCd2+ThioredoxinDepresses antioxidants (glutathione) levels[258,274,277,278,279]
Superoxide anionCysteineOxidative stress
Hydrogen peroxideUbiquitin enzymesDamage in the electron transport chain
Hydroxyl radicalsMitochondrial Complex II IIIApoptosis
Topoisomerase IILipid peroxidation
DNA methyltransferasesAlteration in maintaining genomic integrity
GSH
Glutathione reductase
Glutathione peroxidase
Glutathione s-transferase
MercuryMeHgDNADepresses antioxidants (glutathione) levels[280,281,282,283,284,285]
Hg2+Thioredoxin reductaseOxidative stress
Hydrogen peroxideNitric oxide synthaseLipid peroxidation
Nitric oxideMonoamine oxidaseMitochondrial function
Glutathione reductaseDecreases GABA signaling
Glutathione peroxidaseNeurotransmitter metabolism
Astrocytic glutamine transporterGlutamine uptake
Choline acetyltransferaseAcetylcholine synthesis
Creatine kinaseDecrease ATP production
Cytosolic phospholipase A2Membrane damage in Endothelial cells
Enolase
Glutamate transporters
Ca2+ ATP
MnMn2+DopamineImpairment of oxidative phosphorylation[276,281,286,287,288,289]
Mn3+SODDecrease ATP synthesis
DA-o-quinoneComplex I, IIDisruption of mitochondrial energy production
AminochromeAconitaseAlteration in Ca2+ influx
Adenylate cyclase
Succinate
Malate
Glutamate
N-methyl-d-aspartate (NMDA)
ATP synthase
IronFe2+Complex I IIILipid peroxidation[281,290,291,292]
Fe3+SODDopamine metabolism
Hydrogen peroxide Superoxide radicalα-synucleinMitochondrial functions disruption in ATP synthesis
Hydroxyl radicalsTyrosine hydroxylaseApoptosis
3,4- dihydroxyphenylacetaldehyde DA-o-quinoneHydrogen peroxideDNA/protein degradation
6-hydroxydopamineLipid peroxide
AminochromeDNA/RNA
Creatine kinase BB
Cytochrome c oxidase
Ketoglutarate dehydrogenase
ArseniciAsVATP synthaseUncouples oxidative phosphorylation[250,293,294]
iAsIIIβ-tubulinDecrease ATP formation in the mitochondria
Monomethyl arsonous acid (MMAIII),Glucose 6-arsenateInhibition of the hexokinase
Monomethylarsonic acid (MMAV)Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)Decrease in mitochondrial biogenesis
Dimethylarsinous acid (DMAIII), andMitochondrial transcription factor A (TFAM)
Dimethylarsinic acid (DMAV)Pyruvate dehydrogenase
Arsenic triglutathione
Table 3. Gut microbial interactions with metals, both antagonistic and synergistic.
Table 3. Gut microbial interactions with metals, both antagonistic and synergistic.
Probiotic MicroorganismToxic ElementModel/Cell LineTreatment EffectsCitation
Lactobacillus rhamnosus GR-1Arsenic, Cadmium, Mercury, LeadHumansReduction in toxic levels in blood and microbiota changes (Succinivibrionaceae and Gammaproteobacteria families)[372]
Mix of microorganisms: L. plantarum DSM 24730, L. acidophilus DSM 24735, L. paracasei DSM 24733, B. infantis DSM 24737, B. longum DSM 24736, L. delbrueckii subsp. bulgaricus DSM 24734, B. breve DSM 24732 and Streptococcus thermophilus DSM 24731.Arsenic, Cadmium, Mercury, LeadHumansConcentration of Cd, Hg, and Pb in breast milk, lower concentration of Cd in stools from newborns treated with the probiotics[373]
Lactobacillus rhamnosus GR-1Cadmium, LeadCaco-2 cellsReduce translocation of toxics[24]
Escherichia coli Nissle 1917 (EcN-20) and (EcN-21)Cadmium, MercuryRatsIncreasing GSH levels, SOD activity and catalase, decrease lipid peroxidation in liver and kidney and decrease ALT, AST and ALP activities, bilirubin, creatinine and urea levels in serum[374]
Streptococcus thermophilus, Lactobacillus acidophilus and Bifidobacterium bifidumArsenicRatsAmeliorating the toxic effects on testis and blood metabolites, increase antioxidant enzyme activity (glutathione-s-transferase)[375]
Lactobacillus kefir CIDCA 8348 and JCM 5818CadmiumHuman hepatoma cell lineIncrease cell viability by binding to the toxic metal[376]
Lactobacillus rhamnosus Rosell-11, Lactobacillus acidophilus Rosell-52 and Bifidobacterium longum Rosell-175CadmiumRat hepatocytes, RatsDecrease genotoxicity through binding to toxic metal[377]
Lactobacillus rhamnosus Rosell-11, Lactobacillus acidophilus Rosell-52 and Bifidobacterium longum Rosell-175CadmiumRatsIncrease in toxic metal secretion by feces and decrease in concentration in the blood. Normalized ALT and AST activities[378]
Lactobacillus plantarum L67CadmiumMurine RAW 264.7 cellsInhibits cytotoxicity and intracellular Ca2+ mobilization; suppressed the expression of AP-1 and MAPK protecting against inflammation[379]
Lactobacillus plantarum CCFM8610CadmiumHuman intestinal cell line HT-29 and miceProtection against cell damage, reversed the disruption of tight junctions, protected against inflammation, and decreased the intestinal permeability[380]
Saccharomyces cerevisiaeCadmiumMiceProtected against genotoxic and spermatotoxic effects[381]
Lactobacillus plantarum and Bacillus coagulansCadmiumRatsDecreased AST, ALT, BUN, bilirubin (increased by toxic exposition) and metal accumulation in the liver and kidney[382]
Lactobacillus delbrueckii, Lactobacillus fermentum, Lactobacillus acidophilus, Bifidobacterium and Lactobacillus bulgaricusCadmiumMiceIncreased toxic excretion in feces, and increasing β-catenin and BDNF in brain tissue[383]
Streptococcus thermophilusCadmiumMiceThrough toxic metal binding, decreased levels of toxic metal in blood and attenuation levels of MDA and GSH[384]
Pediococcus pentosaceus GS4CadmiumMiceReduced tissue deposition, increased fecal secretion of toxic, and increased enzymatic activity[208]
Bacillus cereusCadmiumFishReduced Cd accumulation in organs, modulate antioxidant activity and intestinal microbial composition[385]
Lactobacillus plantarum CCFM8610CadmiumHT-29 cells, miceInhibition of metal absorption, alleviated cytotoxicity, reversal of the disruption of tight junctions and inhibition of inflammation[380]
Lactobacillus plantarum CCFM8610CadmiumFishMitigation of oxidative stress in tissues and reversed alterations in hematological and biochemical parameters[386]
Lactobacillus acidophilusCadmiumFishDecreased number of micronucleus formation in erythrocytes and improvement of animal survival rate.[387]
Akkermansia muciniphilaCadmiumMiceModulation of gut microbiota composition[388]
Lactobacillus plantarum CCFM8661LeadMiceDecreased toxic levels in blood and tissues, recover blood δ-aminolevulinic acid dehydratase activity, avoidance of alterations in glutathione, glutathione peroxidase, superoxide dismutase, malondialdehyde, and reactive oxygen species[389]
Lactobacillus plantarum CCFM8662LeadMiceInduced fecal metal excretion through hepatic bile acids synthesis, enhanced biliary glutathione and increased fecal bile acid excretion.[390]
Faecalibacterium prausnitzii and Oscillibacter ruminantiumLeadMiceIncrease in the expression of TJ proteins (ZO-1, occludin and claudin-1), increased fecal Pb excretion, increase SCFAs, pH and oxidative reduction in the intestinal lumen[391]
Lactobacillus plantarum CCFM8610CadmiumMiceDecrease intestinal metal absorption, tissue accumulation, oxidative stress and ameliorate hepatic histopathological changes[392]
Lactobacillus reuteriLeadFishDecreased oxidative stress, reversed alterations in hemato-biochemical parameters, and restored intestinal enzymatic activities[393]
Lactobacillus plantarum, Lactobacillus acidophilus, Bacillus subtilis, Pediococcus pentosaceus, Bacillus licheniformis and Saccharomyces cerevisiaeLeadBroiler ChicksImproved antioxidant parameters, liver transaminases, decreased accumulation of metals and morphological alterations[394]
Lactobacillus delbrueckii subsp. bulgaricus KLDS1.0207LeadMiceIncreased fecal excretion, decreased oxidative stress, lipid peroxide by decreasing MDA concentration, and improved antioxidant production[395]
Lactobacillus pentosus ITA23 and Lactobacillus acidipiscis ITA44LeadBroiler ChicksReduced metal tissue accumulation, decreased lipid peroxidation, and normalized antioxidant activity[179]
Lactobacillus brevis 23017MercuryMiceReduced intestinal inflammation and decreased oxidative stress[396]
and Lactobacillus plantarumMercuryRatsDecreased tissue accumulation of toxic metal, avoided antioxidant alterations, normalized creatinine, bilirubin, urea AST, and ALT levels[397]
Streptococcus thermophilus, Lactobacillus acidophilus and Bifidobacterium bifidumMercuryRatsProtection against the adverse effects in the brain and kidney. Increased activity of glutathione-S-transferase, lactate dehydrogenase, normalized creatinine, triglycerides levels and modulate histopathological changes in the brain.[398]
Table 4. Interactions of probiotic microbiota and their effects on the gut–brain axis.
Table 4. Interactions of probiotic microbiota and their effects on the gut–brain axis.
Probiotic MicroorganismModelOutcomeCitation
Lactobacillus casei ShirotaHumansImproves stool consistency and bowel habits[407]
Lactobacillus acidophilus, Lactobacillus reuteri, Lactobacillus fermentum and Bifidobacterium bifidumHumansDecreases The Movement Disorders Society-Unified Parkinson’s Disease Rating Scale (MDS-UPDRS), biomarkers of inflammation and oxidative stress (high-sensitivity C-reactive protein (hs-CRP), Malondialdehyde (MDA), Glutathione) and insulin metabolism.[408]
Lactobacillus acidophilus, Bifidobacterium bifidum, L. reuteri and Lactobacillus fermentumHumansDownregulates gene expression levels of IL-1, IL-8 and TNF-α; increases the gene expression of TGF-β and PPAR-γ[409]
Lactobacillus acidophilus and Bifidobacterium infantisHumansImproves abdominal pain, bloating and constipation with incomplete evacuation[410]
Streptococcus salivarius subsp thermophilus, Enterococcus faecium, Lactobacillus rhamnosus GG, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus delbrueckii subsp bulgaricus, and Bifidobacterium (breve and animalis subsp lactis)HumansImproves constipation in PD patients[411]
Lactobacillus salivarius LS01 and Lactobacillus acidophilusPeripheral blood mononuclear cells from PD patients and Caco-2 cellsReduces proinflammatory cytokines, oxidative stress and increased the anti-inflammatory response and protection of the epithelium from gut permeability[412]
Lactobacillus acidophilus, Lactobacillus rhamnosus GG and Bifidobacterium animalis lactisMiceIncreases levels of BDNF, GDNF and dopamine, and decreases levels of MAO-B in the brain.[413]
Bifidobacterium bifidum, Bifidobacterium longum, Lactobacillus rhamnosus, Lactobacillus rhamnosus GG, Lactobacillus plantarum LP28 and Lactococcus lactis subsp. LactisMicePreserves dopamine neurons, reduces the motor impairments, and maintains tyrosine hydroxylase in neurons[414]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Forero-Rodríguez, L.J.; Josephs-Spaulding, J.; Flor, S.; Pinzón, A.; Kaleta, C. Parkinson’s Disease and the Metal–Microbiome–Gut–Brain Axis: A Systems Toxicology Approach. Antioxidants 2022, 11, 71. https://doi.org/10.3390/antiox11010071

AMA Style

Forero-Rodríguez LJ, Josephs-Spaulding J, Flor S, Pinzón A, Kaleta C. Parkinson’s Disease and the Metal–Microbiome–Gut–Brain Axis: A Systems Toxicology Approach. Antioxidants. 2022; 11(1):71. https://doi.org/10.3390/antiox11010071

Chicago/Turabian Style

Forero-Rodríguez, Lady Johanna, Jonathan Josephs-Spaulding, Stefano Flor, Andrés Pinzón, and Christoph Kaleta. 2022. "Parkinson’s Disease and the Metal–Microbiome–Gut–Brain Axis: A Systems Toxicology Approach" Antioxidants 11, no. 1: 71. https://doi.org/10.3390/antiox11010071

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