Next Article in Journal
Pharmacological Targeting of Cell Cycle, Apoptotic and Cell Adhesion Signaling Pathways Implicated in Chemoresistance of Cancer Cells
Next Article in Special Issue
Molecular Mechanisms of Oligodendrocyte Regeneration in White Matter-Related Diseases
Previous Article in Journal
Physiological and Proteomics Analyses Reveal Low-Phosphorus Stress Affected the Regulation of Photosynthesis in Soybean
Previous Article in Special Issue
Fewer Functional Deficits and Reduced Cell Death after Ranibizumab Treatment in a Retinal Ischemia Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease

1
Pharmacology Building, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
2
APC Microbiome Ireland, University College Cork, T12YT20 Cork, Ireland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(6), 1689; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19061689
Submission received: 3 May 2018 / Revised: 31 May 2018 / Accepted: 3 June 2018 / Published: 6 June 2018
(This article belongs to the Special Issue Molecular Research on Neurodegenerative Diseases)

Abstract

:
Parkinson’s disease (PD) is a progressively debilitating neurodegenerative disease characterized by α-synucleinopathy, which involves all districts of the brain-gut axis, including the central, autonomic and enteric nervous systems. The highly bidirectional communication between the brain and the gut is markedly influenced by the microbiome through integrated immunological, neuroendocrine and neurological processes. The gut microbiota and its relevant metabolites interact with the host via a series of biochemical and functional inputs, thereby affecting host homeostasis and health. Indeed, a dysregulated microbiota-gut-brain axis in PD might lie at the basis of gastrointestinal dysfunctions which predominantly emerge many years prior to the diagnosis, corroborating the theory that the pathological process is spread from the gut to the brain. Toll-like receptors (TLRs) play a crucial role in innate immunity by recognizing conserved motifs primarily found in microorganisms and a dysregulation in their signaling may be implicated in α-synucleinopathy, such as PD. An overstimulation of the innate immune system due to gut dysbiosis and/or small intestinal bacterial overgrowth, together with higher intestinal barrier permeability, may provoke local and systemic inflammation as well as enteric neuroglial activation, ultimately triggering the development of alpha-synuclein pathology. In this review, we provide the current knowledge regarding the relationship between the microbiota-gut–brain axis and TLRs in PD. A better understanding of the dialogue sustained by the microbiota-gut-brain axis and innate immunity via TLR signaling should bring interesting insights in the pathophysiology of PD and provide novel dietary and/or therapeutic measures aimed at shaping the gut microbiota composition, improving the intestinal epithelial barrier function and balancing the innate immune response in PD patients, in order to influence the early phases of the following neurodegenerative cascade.

Graphical Abstract

1. Introduction

Parkinson’s disease (PD) is acknowledged as the second most common neurodegenerative disorder, estimated to affect 1–2 per 1000 of the population worldwide [1]. About seven to ten million people in the world suffer from PD [2]. This figure is expected to double in the near future, circa 2030, due to aging of the population [3]. The etiology of PD still remains unclear. However, the slow progression of the disease evolves years before the diagnosis is ascertained, involving various neuroanatomical areas, arising from an assortment of genetic and environmental factors, and exhibiting a large array of debilitating symptoms. From a histopathological perspective, PD is hallmarked by a distinctive depauperation of dopaminergic neurons in the substantia nigra pars compacta (SNc) with consequent dopamine deficiency within the striatum and by the manifestation of intracellular eosinophilic inclusions, the so called Lewy bodies and Lewy neurites in the remaining neurons. Lewy pathology is characterized by intracellular insoluble aggregates of misfolded α-synuclein and implicates not only the brain but is also widespread in the spinal cord and peripheral nervous system, including sympathetic ganglia, enteric nervous system (ENS), salivary glands, adrenal medulla, vagus nerve, cutaneous nerves and the sciatic nerve [4,5,6]. The progressive dopamine deficit in the basal ganglia determines the characteristic parkinsonian triad of motor symptoms—rigidity, bradykinesia and tremor. However, other significant non-motor symptoms have been receiving increasing attention for the more negative impact in the quality of life of PD patients in comparison to motor symptoms. These non-motor symptoms involve neuropsychiatric disorders (e.g., cognitive impairment, depression, apathy, psychosis), sleep disturbances, sensory alterations (pain, olfactory impairment), and the common gastrointestinal (GI) dysfunction reported by more than 80% of PD patients [7]. The first-line intervention in PD management is the administration of dopamine modulators even if they can exert serious side effects, produce limited benefits on alleviating non-motor disturbances and often fail to be effective in the later stages of PD [6,7,8].
In the last decade, emerging evidence has revealed the presence of an intense dialogue between the brain and the GI system, the so-called brain-gut axis. Disruption of this complex relationship has been shown to be associated to the pathogenesis of several disorders, ranging from irritable bowel syndrome (IBS), liver disease and chronic abdominal pain syndromes, to depression, anxiety, autism spectrum disorders, dementia and PD. The brain-gut (or gut-brain axis) crosstalk can occur in a bidirectional fashion: firstly, through a gut quiver on central nervous system (CNS) activities (e.g., changes in mood, cognition or perception due to functional GI disorders [9,10], or subsequent to the release of gut hormones [11], or following serious GI inflammatory diseases such as acute pancreatitis [12,13,14]); secondly, through a central quiver on gut activities (e.g., stress-induced GI dysfunction [15]). Indeed, a common alignment of both brain and gut can be identified in neurobiological disorders (e.g., neurodegeneration or gliosis) [16].
Traditionally, the brain-gut axis was viewed as a flux of information, mediated by neurohormones and inflammatory factors, travelling between the central, autonomic and enteric nervous systems (CNS, ANS and ENS, respectively) with the concurrent participation of the neuroendocrine and neuroimmune systems.
Several studies have focused on the role of the CNS in modulating the intestinal inflammation through both parts of the ANS, the sympathetic and parasympathetic nervous system [17,18]. The inflammatory state of peripheral tissues is conveyed to the brain through afferent nerves which in turn suppress cytokine production, improve intestinal barrier integrity and limit gut inflammation [18]. Recent studies performed in animal models of acute and chronic pancreatitis have shown that the stimulation of primary afferent capsaicin-sensitive neurons or treatment with peptides (e.g., calcitonin gene-related peptide) before the exposure to harmful factors, can activate an adaptive mechanism called “preconditioning” which is able to reduce pancreatitis development [19,20,21]. Sensory neurons are involved in gastroprotection and regulation of visceral blood flow and their stimulation by capsaicin can potentially inhibit the progression of inflammation, by improving the endogenous release of nitric oxide (NO) and thus the pancreatic blood flow [22,23,24]. In recent years, several preclinical studies highlighted that certain psychoactive molecules can modulate the endocannabinoid system in the gut and possibly impact the pathogenesis of inflammatory bowel disease, as well as its extra intestinal manifestations such as pancreatitis [25,26]. It has been demonstrated by Warzecha et al. [25] that anandamide reduces mucosal oxidative stress, inhibits the inflammatory process and preserves the integrity of gastric mucosa in stress-induced gastric ulcers [25]. These effects are partly mediated by capsaicin-sensitive sensory nerves [25] and, in the case of acute pancreatitis, the protective action of anandamide depends on the phase of the inflammation [27,28]. Thus, the modulation of the endocannabinoid system may be useful to treat gut-brain motor dysfunction in PD.
Changes in the ANS occurred in conjunction with intestinal inflammation, however, disorders such as IBS are also linked to inflammatory abnormalities of the ENS [17]. The ENS is the largest nervous system outside the CNS, which autonomously regulates numerous functions of the GI tract, either independently through neuro-glial circuits in the myenteric and submucosal plexus, or by input of sympathetic and parasympathetic pathways to/from the brain. The enteric neurons and glial cells form a vast communication network in close relationship with the gut microbiota and thus the ENS can easily be affected by microbiome alterations and be involved in GI disorders as well as in neurodegenerative diseases. Therefore, the ENS could represent an entry point for pathogens or—conversely—for therapeutic interventions based on diet and/or commensal microbes-derived molecules [29].
Lately, it is becoming increasingly clear that a third player, such as the gut microbiota, can significantly influence the gut-brain crosstalk, having a marked impact on digestive processes, immune responses, emotional status, perception and cognitive functions [16]. The microbiota-gut-brain axis has attracted much attention regarding the pathogenesis of PD, in which GI dysfunction appears about twenty years before motor impairments. Although PD patients manifest both gut dysmotility and altered microbial composition, it is still unclear which condition comes first and what role the gut and the gut microbiota have in PD progression.
In addition to maintaining gut homeostasis and several essential host physiological functions, the gut microbiota is a producer of an assortment of Toll-like receptor (TLR) ligands, which can exert proinflammatory effects under certain conditions. Despite microbial-derived components being potent TLR ligands, the gut has a high tolerance to TLR ligands because epithelial cells express minimal TLRs under physiological conditions [30]. In contrast, altered gut microbiota and disrupted gut epithelial barrier activate TLRs which in turn trigger downstream signaling pathways, promoting inflammation and oxidative stress in both the gut and brain of PD patients. Thus, gut microbiota and TLRs could represent potential targets for PD treatment.
The exact mechanisms by which gut microbiota contribute to PD are still poorly understood, despite the role of gut microbiota in the development of PD being well documented. Here, we first describe the functional aspects of gut microbiota observed in PD. Then, we review the role of TLRs associated with PD and their potential as a new target of dietary and/or therapeutic interventions.

2. Microbiota-Gut-Brain Axis and Host Health

Over the last decade, an increasing amount of literature has focused on the codevelopment of the gut microbiota with the human host since birth and on their mutual shaping clearly relying on the host genome, nutrition and lifestyle [31]. While the association of neuropsychiatric disorders with GI disturbances dates back to Hippocrates, a clear demonstration of the essential cooperation between brain and microbes was first described by the impressive amelioration of symptoms in patients affected by hepatic encephalopathy following treatment with nonadsorbable oral antibiotics [32]. The gut microbiota is now being referred to as a new organ or an emergent system, which comprises a number of microorganisms (bacteria, archaea, fungi, and viruses) comparable to the number of cells residing in the human body [33]. In particular, the enteric microbiota, distributed along the human GI tract, displays similar results in terms of relative abundance and distribution between healthy adults, although the microbe profile is quite stable and unique for each individual, and can be considered a personal microbic fingerprint or enterotype [34]. Firmicutes and Bacteriodetes are the most dominant phyla (about 51% and 48%, respectively), with Actinobacteria (including the Bifidobacteria genera), Cyanobacteria, Lentisphaerae, Fusobacteria, Spirochaetes, Proteobacteria, and Verrucomicrobia phyla existing in relatively low abundance [35]. Although a relative consistency in microbial composition in healthy people is usually maintained over time, small daily variations are found in each individual unless exposed to disrupting agents or conditions, such as antibiotics, colonization by foreign commensal microbes, marked changes in diet or lifestyle, or infectious or noninfectious disease [36,37,38]. The magnitude and the length of the disruption might affect the capability of microbiota to recover and return to the original composition once the dysfunction is resolved, however repeated harmful stimuli will weaken its recovery with potential downstream outcomes on host physiology [38].
Aging is a critical window for not only the gut and brain function but also for the composition of enteric microbiota that in turn may have serious consequences on health integrity in these latter stages of life [38].
The bidirectional dialogue between the gut and the brain involves different mechanisms, including the enteric and central neural network, neuroendocrine-hypothalamic-pituitary-adrenal axis, immune system, several neurotransmitters and neural regulators directly produced by gut bacteria, and barrier paths such as intestinal mucosal barrier and blood-brain barrier. The enteric microbiota is implicated in the upregulation of the local and systemic inflammatory response induced by lipopolysaccharides (LPS) derived from pathogens and the related production of proinflammatory mediators. The dysregulation of immune response to environmental and/or microbial agents is associated with the onset of inflammatory bowel disease in genetically susceptible individuals [39]. On the other hand, exposition to low amounts of LPS in early life can affect the ability of the immune cells to produce the cytokines, increasing the resistance of the organism to systemic diseases such as pancreatic inflammation [40].
Gut dysbiosis and/or small intestinal overgrowth (SIBO) increase intestinal permeability and bacterial translocation, determining an immune system’s overresponse and consequent systemic and/or central nervous system (CNS) inflammation. Enteric bacterial cells possess the capacity to produce numerous neuroactive molecules, such as serotonin, catecholamines, glutamate, γ-amminobutyric acid (GABA) and short-chain-fatty acids (SCFAs) [41,42,43]. It has been proposed that the variety of neurotransmitters, neuromodulators and neurohormones produced by microorganisms are the “words” of a common language that enables a sophisticated synergic communication [44]. However, considering the extreme complexity of this communication network, it remains to be determined whether microbial neurochemicals are generated at an adequate level in respect to host production to exert any kind of effect, or can be delivered to central neurocircuits through systemic circulation [41,42]. Although some reports indicate the ability of bacteria to modulate the level of neurotransmitters through TLRs and heat-shock proteins [45], this form of interaction may occur directly via neuroactive molecules.
Most of the functions of the GI tract are regulated by enteric neurons, hormones produced by enteroendocrine cells, and cytokines synthesized in somatic cells [46]. Of note, more than 90% of all serotonin in the body is synthesized in the gut by the enterochromaffin cells, an enteroendocrine cell subtype, known to be involved in controlling gut motility, emesis, visceral hypersensitivity and secretion [47]. Most enteric serotonin is tuned by gut microbiome whereas the circulating serotonin is generally metabolized by the liver [48]. This neuroamine regulates a variety of physiological processes at the periphery but, even if it cannot cross the brain blood barrier (BBB), can affect central neurocircuits by interfering with vagal nerve activity and BBB permeability, thus indirectly influencing brain functions [49]. Therefore, although the brain and the gut are the two major sources of serotonin synthesis, physically separated by the BBB, from a biological point of view these pathways are potentially not always distinct. In this respect, enteric bacteria can also produce neurotoxic molecules (e.g., d-lactic, ammonia) and neurotoxins, (e.g., those produced by Clostridium perfringens, Clostridium botulinum, Clostridium butyricum, Clostridium baratii, among others) and reach CNS being transported via systemic circulation or through extrinsic afferent nerve fibers (i.e., vagal nerve projections from the gut to the brainstem and spinal nerve projections from the gut to the spinal cord), provoking neuronal injury [50,51]. In turn, the CNS can control the enteric microbiota through adrenergic neurotransmission, primarily modulating intestinal motility and neuroimmune crosstalk [52].
Most of the effects mediated by the gut microbiota or potential food-based therapeutic interventions (e.g., probiotics, prebiotics, synbiotics) on CNS functionality have been shown to be exerted through the modulation of vagal neurotransmission [53]. Likewise, microbial colonization of the gut after birth and during infancy is markedly involved in the postnatal development and maturation of the host nervous, immune, and endocrine systems. Altogether these pieces of evidence highlight the key role of microbiota-gut-brain axis in ensuring and protecting host health and its involvement in a plethora of diseases ranging from stress-induced disorders to neurologic and psychiatric disorders [53].

3. The Gastrointestinal System in Parkinson’s Disease

GI dysfunctions are generally experienced by most patients with PD and usually include hypersalivation, dysphagia, delayed gastric emptying, nausea, constipation and altered bowel habits. Disturbances in oral and pharyngeal swallowing have been shown in other neurological diseases, such as amyotrophic lateral sclerosis, and manometric endoscopy of the upper GI tract was a useful procedure for the assessment of the severity of deglutition disorders among these patients [54].
A higher prevalence of peptic ulcer and Helicobacter pylory (Hp) infection has been revealed in PD patients. Hp infection is considered to be the most important factor responsible for the development of gastroduodenal diseases, including active chronic gastritis, peptic ulcer disease and gastric adenocarcinoma [55]. However, there is increasing evidence that Hp gastric infections are potentially associated with several systemic extra-GI diseases such as cardiovascular, immunological and inflammatory disorders (e.g., acute pancreatitis) [55].
Constipation is a major enteric dysfunction of PD and predates motor symptoms years before the manifestation of the disease, making it one of the earliest biomarkers of the pathological process that will ultimately emerge into PD [6]. The anomalies in gut functionality in PD may result from both central and peripheral altered neurotransmission pathways. Even if until now the findings of an impaired enteric neurotransmission are contradictory [56], a pathological hallmark of PD is the distribution of α-synuclein pathology in olfactory bulbs and in both submucosal and mucosal plexuses of gut ENS from esophagus to the rectal end. Braak et al. [57] have proposed that α-synuclein deposition might begin in the olfactory bulbs or/and in the ENS by an unknown environmental toxin and/or microbial pathogen and then proceed towards SNc and further sites in the CNS. Full truncal vagotomy in PD patients resulted in decreased risk of PD progression compared to partial or no vagotomy, suggesting an involvement of the vagus nerve as a conduit for the spreading of Lewy bodies from ENS to CNS [58]. Moreover, it has been shown that toxins like pesticides (e.g., rotenone), orally administered to mice, replicate the staging of PD-like pathology from ENS to CNS [35]. In this preclinical model, the resection of sympathetic and parasympathetic nerves (i.e., hemivagotomy and partial sympathectomy) prior to the oral exposure to rotenone prevented the spreading of PD-like pathology from ENS to the previously connected central neurocircuits and delay the manifestation of gut motor symptoms [59]. Considering that gut mucosa is in contact with environmental factors, these findings support the concept that external triggers, including diet-derived molecules, toxins or microbes, might have a major role in eliciting and spreading PD pathology, potentially in a milieu of genetic vulnerability.
However, recent studies have suggested that the initiation of α-synuclein pathology in the GI tract does not necessarily need a pathogenic or environmental trigger since it can be instigated by enteric microbiota [60,61].

4. The Gut Microbiota in Parkinson’s Disease

Gut constipation, SIBO, increased intestinal permeability of the mucosal barrier and GI inflammation are all symptoms closely interrelated to gut microbiota composition and microbial-derived metabolites. In the early stages of PD, higher intestinal permeability, a condition known as leaky gut syndrome, has been observed to be associated with enteric α-synuclein pathology [60]. Increased intestinal permeability, due to a defective gut barrier function, facilitates the translocation of microorganisms (e.g., bacteria) and microbial products (e.g., LPS), which, in turn, might initiate inflammation and oxidative stress, thereby leading to α-synucleinopathy in the ENS [60]. Moreover, higher levels of gut-derived LPS can disrupt the integrity of the BBB, promoting neuroinflammation and damage in SNc [62].
Current evidence from different clinical studies has revealed that gut microbiota composition is altered in PD patients and related to the clinical phenotype of the disease. Analysis of fecal samples of PD subjects disclosed higher levels of Enterobacteriaceae, which were positively associated with the degree of gait and postural instability [63], as well as a reduced abundance of the Prevotellaceae bacterial family. Prevotellaceae are commensals bacteria, implicated in the production of mucin in the gut mucosal layer, in the synthesis of neuroactive SCFAs (e.g., acetate, propionate, and butyrate) through fiber fermentation, and in the release of thiamine and folate. Hence, the decreased levels of Prevotellaceae could be related to reduced mucin synthesis and increased intestinal permeability, with consequent systemic exposure to bacterial antigens and endotoxins [60], and potential development of α-synucleinopathy by the disruption of clearance mechanisms of the intra- and extraneuronal protein by SCFA-dependent modulation of gene expression [64].
Furthermore, the lower abundance of Prevotellaceae associated with an increase of Lactobacillaceae has been linked to a reduction in the levels of gut hormone ghrelin, known to be involved in ensuring physiological nigrostriatal dopamine activity [65]. Indeed, in PD patients an impaired postprandial response to ghrelin has been reported [66].
A subsequent study demonstrated changes in both mucosal and fecal microbial compositions of PD, such as a higher abundance in the putative, proinflammatory Proteobacteria of the genus Ralstonia and a reduced level of bacteria from the genera Blautia, Coprococcus, and Roseburia, involved in producing butyrate, a SCFA associated with anti-inflammatory properties [61]. Furthermore, decreased concentrations in SCFA have also been evidenced in PD patients and could be related to the observed impaired peristaltic regulation by the ENS and consequent gut dysmotility. At a genomic level, PD microbiota owns more genes implicated in LPS biosynthesis and type III bacterial secretion systems and a lower number of genes related to metabolism [61]. Although the role of gut microbiota in PD is still at its infancy, a recent experimental work demonstrated that fecal microbiota transplant from PD-donor accelerated the disease in genetically predisposed animals [67]. These findings advocate that PD-associated factors in the host alter enteric microbiome, which in turn worsens PD pathology and symptoms. Indeed, the enteric microbiota is implicated in the regulation of the local and systemic inflammatory response induced by LPS and the production of proinflammatory mediators.

5. Toll-Like Receptors

Being the first line of defense against invading microbes, the innate immune system is vital in early recognition of infection [68]. Innate immunity senses the presence of microorganisms through a number of patter-recognition receptors (PRRs) [69], which specifically recognize evolutionarily conserved molecular structures, denominated pathogen-associated molecular patterns (PAMPs), widely expressed by a variety of infectious microbes, which are essential for their own survival [70]. Among the different PRRs, situated in both extracellular and intracellular milieu, TLRs are the most interesting family of mammalian receptors, orthologs of the Toll receptors discovered in Drosophila Melanogaster in 1988 [71]. Until now, thirteen TLRs have been identified in mammals, of which a total of ten have been characterized in a variety of human tissue and cells, not only belonging to the immune system and epithelial tissue but also to neurons and glial cells of both the peripheral nervous system (PNS) and the CNS. The distribution of these immune sensors in the nervous system highlights the ability of neural cells to mediate immune responses as well as their vital dependency for development and homeostasis on TLRs signaling [72,73,74,75,76]. Indeed, TLRs are not only engaged by PAMPs but also by a set of endogenous molecules generated during tissue damage (damage-associated molecular patterns, DAMPs), activating firstly an acute immune response and, secondarily, tuning the subsequent adaptive immune reaction. TLRs are further classified into subfamilies according to the types of PAMPs or DAMPs that they recognize. The plasma membrane bound TLRs interact with molecules composing the bacterial cell walls and membranes, such as lipopeptides, peptidoglycans and glycolipids (TLR1/TLR2, TLR2/TLR6, TLR2/TLR10), fibronectin, lipopolysaccharides, and heat shock proteins (TLR4), and flagellin (TLR5). On the contrary, the intracellular TLRs, expressed in cell endosomes, detect microbial nucleic acids, specifically viral double-strand RNAs (TLR3), single-strand RNAs (TLR7 and TLR8), unmethylated CpG DNA (TLR9) [77]. TLR engagement activates distinctive adaptor proteins, such as Myeloid differentiation primary response gene 88 (MyD88) or the TIR- domain-containing adapter-inducing interferon-β (TRIF) pathway, to deliver signals to several downstream pathways, comprising the nuclear factor-kappa B (NF-κB), mitogen-activated protein kinases (MAPK), and/or interferon-regulatory factor (IRF) signaling pathways, which are pivotal for the expression of a variety of gene products involved in inflammatory responses [77,78].
More and more studies have focused their attention on the role that TLRs may play in neurodegeneration, considering that they are extensively expressed in immune and non-immune cells and their expression can change not only during microbial infections but also in the presence of sterile inflammation when the pathogens are absent.

6. TLRs in Parkinson’s Disease

Even if the exact etiology of sporadic PD is still to be found, increasing evidence suggest that misfolded α-synuclein activates microglial cells in the SNc, promoting inflammation and oxidative stress, leading to neurodegeneration. Extracellular misfolded, fibrillar α-synuclein, released from neural cells or oligodendrocytes is recognized as PAMP or DAMP by microglial TLR2 (as heterodimer with TLR1), which in turn activates downstream pathways involving MyD88 and NF-κB, triggering the production of TNF and IL-1β [79,80] and increasing selective expression of TLRs in a localization- and time-dependent manner [81]. In in vitro studies TLR4 appears also to interact with α-synuclein, triggering microglial responses, including α-synuclein uptake, proinflammatory cytokine release, and oxidative stress promotion [82]. These findings were corroborated in an MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin)–induced murine model of PD where the genetic absence of TLR4 signaling protected the mice from neurodegeneration, highlighting the primary role of TLR4 in PD development [83]. Recent evidence in experimental models suggests a potential involvement of the NLRP3 inflammasome, which can be modulated by TLRs signaling. Reduced caspase-1 activation and IL-1β release together with decreased loss of dopaminergic neurons in SNc was found in NLRP3-deficient mice following MPTP insult [84]. Fibrillar α-synuclein has been reported to engage TLR2 on human monocytes to prime the NLRP3 inflammasome [85]. Intriguingly, NLRP3 inflammasome inhibition was shown to be mediated by dopamine itself, although it is still unclear if involvement of the inflammasome is antecedent to the degeneration of dopaminergic neurons or is the result of neural depauperation [86]. The most convincing evidence for a role of TLRs in PD has been found so far for TLR2 and TLR4. The contribution of TLR2 and TLR4 in PD might be a double-edged sword: their activation in microglia can trigger neurotoxicity but on the other hand they might be essential for clearing misfolded α-synuclein, hence being neuroprotective [87].
There is an increasing recognition of the involvement of TLRs in neuronal degeneration based on the following pieces of evidence: (i) relevant cells of the nervous system express TLRs; (ii) TLRs are activated by α-synuclein; (iii) their activation induces an inflammatory response that precedes neuronal loss; (iv) halting TLRs engagement delays PD progression [88].

7. Microbiota-Gut-Brain-Axis and Toll-Like Receptor Signaling: Potential Implications in Parkinson’s Disease

Aging determines alterations in responsivity of microglia, which may acquire a hyperreactive phenotype (e.g., increased release of proinflammatory mediators and overexpression of cell surface receptors) or an impaired condition with defective phagocytosis and clearance of misfolded protein aggregates [89]. The acquired activated state of microglia in response to accumulation of abnormally folded proteins and neurodegeneration is a process referred to as microglial priming, similar to the newly emerging theory of “trained immunity” or “innate immune memory”, consisting in the epigenetic and metabolic reprogramming of peripheral innate immune cells following an initial insult. Indeed, macrophages derived from human hematopoietic stem and progenitor cells challenged with a TLR2 agonist during their differentiation and then inoculated in irradiated mice evidenced a tolerant phenotype by releasing a reduced amount of inflammatory mediators and reactive oxygen species in response to the inflammatory stimulus [90]. A growing body of evidence suggests that microglial priming could be induced by epigenetic reprogramming as shown in trained immune cells [86]. Intestinal barrier function is impaired in patients with PD placing them at higher risk to be exposed to microbial products [91], therefore the translocation of bacteria or bacterial-derived products such as LPS (a TLR4 ligand) can induce a systemic inflammation, producing a more severe neurodegeneration. Extracellular α-synuclein is recognized as a DAMP and TLR4 ablation has been shown to impair the phagocytic response of microglia to α-synuclein with consequent accumulation of α-synuclein and increased dopaminergic neurodegeneration in the SNc [92], highlighting the neuroprotective effect of TLR4 signaling mediated through the clearance of α-synuclein, analogously to the suggested protective role of TLR2 towards β-amyloid and α-synuclein.
It has been demonstrated that bacteria residing in the enteric microbiota produce extracellular amyloid proteins. Curli, an amyloid protein synthesized by E. coli and S. typhimurium, has been shown to enhance colonization and biofilm formation. Even if few studies have focused on the presence of gut bacteria-derived amyloid proteins in the GI tract, the effect of bacterial amyloids on α-synuclein accumulation or other molecular mechanisms associated to neurodegeneration is still unknown [93]. The enteric microbiota is a huge antigenic load resident in the gut and confers marked potential danger if not kept under continuous surveillance, such as under TLRs sensing. However, the enteric commensal microbiota is required for the constant stimulation of the immune system and TLR-mediated sensing of these microorganisms may play a dual role in disease development as a source of both inflammatory and regulatory signals. In this respect, it is important to take into account that the microbiota is also a source of biological active signaling molecules, immune mediators and gut hormones. Some of those, including serotonin, purines, GABA and neurotrophic factors, among others, have been shown to be involved in TLRs signaling [72,73,94,95,96,97]. Recent evidence has also pointed out that microbiome-derived SCFAs finely tune microglial function, further suggesting that microbiome-derived factors and nutrition are involved in controlling innate immune function in neurodegeneration [98]. Intriguingly, the SCFA butyrate has been shown to increase TLR-dependent responses by increasing their expression in a cellular model of human enteroendocrine L-cells [99].
The emerging evidence described above suggests the existence of a multifaceted TLR signaling network that influences neural circuits and immune-mediated processes both in the gut and in the brain. Further studies focused on discovering the enteric microbiota-derived factors responsible of TLRs engagement and the consequent signaling outcomes of TLR activation in both the ENS and the CNS will provide novel insights into the complex dialogue between the host and the microbiota in PD and other relevant neurodegenerative disorders.

8. Potential Therapeutic Strategies

Recent research has gathered solid scientific evidence on the involvement of the GI tract in PD, highlighting three important features. First, in animal models and human studies a clear association has been put forward between the GI tract, gut inflammation, increased permeability, and PD. Second, some dietary interventions appear to exert beneficial effects by modulating microbiota dysbiosis toward a healthy state, reducing gut permeability, and/or decreasing oxidative stress and intestinal inflammation. Third, since TLRs are dynamic guardians laying squarely between the environment and the host, especially in the gut, any changes in TLR activity and expression might reduce or prevent the incidence of developing diseases associated with an inflammatory status such as PD. Deciphering the interactive dialogue that occurs between microorganisms in the gut and the activity of the immune system regulated by TLRs is crucial for the discovery and development of compounds such as pharma- and nutraceuticals (including syn-, pre- and probiotics), critical for preserving or restoring homeostasis in the gut as well as in other host tissues.
None of the current medicines for PD show a beneficial influence on disease progression and do not exert any effects on the microbiota-gut-brain axis to reduce the spread of Lewy pathology or to alleviate motor and/or non-motor symptoms. However, traditional treatments for PD could be combined with modulators of TLRs activity and/or food-based interventions to alleviate gut dysfunction and symptoms, as well as positively influence the microbiota-gut-brain axis, thus reducing GI dysbiosis and protecting the complex neuroglia network in both the ENS and CNS, [41,100,101,102,103].

8.1. Dietary Supplements

High levels of polyunsaturated fatty acids (PUFA), such as the omega-3 fatty acid docosahexaenoic (DHA), have been shown to induce anti-inflammatory effects and reduce mitochondrial dysfunction-mediated motor symptoms together with decreasing alpha-synuclein accumulation and inflammation in PD animal models [102,104,105,106,107]. DHA inhibits, whereas saturated fatty acids can activate, certain TLR-mediated pro-inflammatory signaling pathways. DHA blocks the activation of TLR4 and TLR2/1 or TLR2/6 and other TLRs in an indirect manner, targeting TLRs downstream pathways during the receptor dimerization process (e.g., lipid rafts) [108]. Overall these findings highlight the involvement of diet, such as the intake of saturated fatty acids and DHA, in the modulation of TLR signaling pathways and related involvement in chronic inflammation and subsequent risk of chronic diseases [108,109]. An in-vitro study has shown that an extract of Panax notoginseng was able to suppress microglial activation and decrease the release of inflammatory factors (IL-6 and TNF-α), suggesting the potential therapeutic utility in slowing down PD progression [110]. The flavonoid silymarin, extracted from the seeds and fruit of Silybum marianum, was found to exert neuroprotective effects in 6-OHDA-induced hemi-parkinsonian rats, through the alleviation of nigral injury, the increase of anti-oxidant defenses and suppression of TLR4 activation [111].

8.2. Probiotics

Probiotics are defined by the Food and Agriculture Organization of the United Nations and by the World Health Organization (FAO/WHO) as “live microorganisms which, when administered in adequate amounts, confer a health benefit on the host” [112]. The common benefit of probiotics on gut microbiota originates from creating a more favorable GI environment and supporting a healthy immune system [112]. Common probiotic functions are dependent on key core mechanisms found in the shared architecture of the cell surface structures of most Gram-positive probiotics, such as peptidoglycan, cell wall teichoic and lipoteichoic acid (LTA), and common but varying components including exopolysaccharides, surface layer associated proteins (SLAPS), mucin-binding proteins (MUBs), fibronectin binding proteins, and pili. These cell surface macromolecules in bacteria are important factors in the beneficial microbiota-host dialogue, as they can interact directly with the intestinal epithelium, mucus, and TLRs of the GI mucosa. The traditionally-used probiotics are from the genera Lactobacillus and Bifidobacterium, which have been shown to enhance intestinal epithelial integrity, protect from gut barrier disruption, regulate mucosal immune system and inhibit pathogenic bacterial growth [112,113].
The most common probiotics belonging to the genera of Lactobacillus and Bifidobacterium are Gram-positive bacteria and present a well-conserved core molecular architecture of peptidoglycan and LTA, which are documented MAMPs interacting with TLR2/6 heterodimers. Modifications in LTA structure or removal of LTA have elicited significant anti-inflammatory consequences observed in mouse models of both colitis and colon cancer, suggesting that Gram-positive probiotics with decreased expression of LTA are more prone to modulate the anti-inflammatory immunological consequences in degenerative disorders [114].
The administration of probiotics has been shown to positively modulate brain function, including stabilizing anxiety-like [115] and depression-like behavior [116], through the microbiota-gut-brain axis [117]. Chronic administration of the Lactobacillus strain in healthy mice has been shown to reduce anxiety, depression and stress responses, and modulate the expression of central GABA receptors, but only when the vagus nerve function was maintained intact, thereby highlighting the critical role of autonomic transmission in the gut-brain axis [118]. Preclinical or clinical evidence on the beneficial effects of probiotics in PD are still very limited. Ingestion of a fermented milk, supplemented with multiple probiotic strains and prebiotic fiber, for four weeks was found to be superior to placebo (a pasteurized, fermented, fiber-free milk) in improving constipation in patients with PD [119]. Another study showed that fermented milk containing Lactobacillus casei strain Shirota for five weeks decreased abdominal pain and bloating, and improved stool consistency [120].
A recent well-designed double-blinded randomized clinical trial (RCT) by Ojetti [121] showed that Lactobacillus reuteri improved bowel movement frequency compared to placebo in patients with functional constipation.
As mentioned above, gut dysfunction, including constipation and gut dysbiosis, contributes to PD morbidity and participates in the pathological process of PD. Hence, the use of probiotics might offer relief from the complications as well as decrease gut permeability, microbial translocation and neuroinflammation in the ENS. Restoring gut function by administration of probiotics might lead to better levodopa adsorption and improve behavioral and cognitive performance, which are common symptoms in PD patients.

8.3. Prebiotics

Prebiotics are defined as “a selectively fermented ingredient that results in specific changes, in the composition and/or activity of the gut microbiota, thus conferring benefit(s) upon host health” [122]. Examples of prebiotics are inulin, galacto-oligisaccharides (GOS), fructo-oligosaccharides (FOS) and SCFAs. Considering that human enzymes are not able to digest complex carbohydrates, specific gut microbes are involved in fermenting carbohydrates to SCFAs (i.e., acetate, proprionate and butyrate), lactose, hydrogen, methane, and carbon dioxide, all metabolic products that, by lowering the pH of the intestinal milieu, inhibit the survival and proliferation of pathogenic bacteria [103]. Prebiotic fibers are well known for their beneficial effects on gut motility and immune function that are compromised in PD patients [123]. The chronic administration of GOS and FOS for five weeks increased the levels of brain-derived neurotrophic factor (BDNF) in the hippocampus of healthy rats, suggesting that prebiotics can be implicated in ensuring CNS neuroprotection [124]. Although several studies have shown the beneficial effect of prebiotics on GI function, immune response and neuroprotection, to date, no clinical study has evaluated their use in PD which could be highly interesting in view of the fact that PD patients have a lower abundance of SCFA-butyrate producing bacteria [61].

8.4. Antibiotics

Treating SIBO with antibiotics is a potential therapeutic strategy to improve the motor symptoms, reduce intestinal bacterial contents as well as gas production. However, the therapy for SIBO is challenging due to the absence of well-designed clinical trials in patients with or without PD and SIBO. Moreover, there is still an absence of consensus on the exact definition of SIBO together with specific diagnostic criteria that render the treatment complicated. Antibiotic therapy is usually a good option for overcoming SIBO; however, considering that SIBO is caused by Gram-positive and -negative anaerobes and aerobes, the choice of most appropriate antibiotic poses a challenge for gastroenterologists. Rifaximin, a broad-spectrum nonadsorbable antibiotic, might be effective in the treatment of SIBO for its action on Gram-negative and -positive aerobic and anaerobic bacteria [125]. While rifaximin is the only antibiotic FDA-approved for treatment of diarrhea-predominant IBS and the most studied antibiotic for the treatment of SIBO, no data on the use of rifaximin for treatment of SIBO in patients with PD is currently available. A variety of other antibiotics (e.g., metronidazole, ciprofloxacin, norfloxacin, amoxicillin-clavulanate, tetracycline, doxycycline and neomycin etc.) have been evaluated for curing SIBO but the lower quality and sample power of most RCT have yielded no clear consensus on the most appropriate type of antibiotic and related posology to be used. A growing body of evidence has suggested that minocycline elicits neuroprotective effects in PD, owing to its ability to restore gut microbiota balance (reduction of Firmicutes/Bacteroidetes). Clinically, Phase II trials have assessed its efficacy in PD patients and it is now under consideration for undertaking a Phase III trial [126].

8.5. TLRs Modulators

As mentioned above, TLRs appear to be associated with the neurodegenerative processes characteristic of PD. TLR2 and TLR4 are potentially the most involved receptors in the progression of PD. However, it was not clear until now whether the most beneficial outcome would be achieved by stimulating TLR2 and/or TLR4 (e.g., favoring alpha-syn clearance) or to repress their signaling to delimitate Lewy pathology as well as augment the levels of toxic factors such as proinflammatory cytokines and reactive oxygen and nitrogen species. Considering that chronic administration of TLR2 and TLR4 ligands elicits microglial activation and a related marked release of toxic factors, current knowledge in other pathologies advocates for their inhibition to prevent further tissue damage. Several negative regulatory mechanisms have been developed to mitigate TLR signaling and ensure a balanced immune response. To date, six levels of negative regulation have been revealed: (i) degradation of TLR protein; (ii) down regulation of transcription mechanisms of TLR and related genes; (iii) post transcriptional repression by microRNAs (miRNAs); (iv) release of soluble TLRs acting as soluble decoy; (v) intracellular inhibitors; and (vi) membrane-bound blockers of TLR signaling pathways after TLR engagement by its ligands [127]. Preclinical and clinical studies have evaluated Eritoran tetrasodium, a compound that interferes with TLR4/MD-2/LPS, inhibiting the LPS-induced release of TNF-α, IL-β and IL-6, thus limiting excessive inflammation associated with TLR4 activation, as a potential therapeutic for sepsis. However, as far as we know, the efficacy of Eritoran has never been tested in PD [128]. Ibudilast is a cyclic nucleotide phosphodiesterase inhibitor as well as a TLR4 antagonist which is currently used as a bronchodilator, vasodilator and anti-inflammatory agent for the treatment of asthma, post-stroke dizziness and ocular allergies in Japan and other Asian countries [129]. Considering its documented ability on up-regulating the anti-inflammatory cytokines (i.e., IL-10, IL-4) and promoting the production of neurotrophic factors (i.e., GDNF, NGF, NT-4), it has been recently tested in an animal model of PD. Ibudilast showed an anti-inflammatory response via the modulation of glial cell activity, attenuation of TNF-α expression and induction of GDNF. However, these ibudilast-mediated beneficial events were not a sufficient protection in the acute phase of injury [129]. Anti-TLR2 antibodies have been tested in clinical studies for treating severe sepsis and inflammation. It is conceivable that these TLRs signaling inhibitors could modulate the progression of the severity of PD.
MicroRNAs (miRNAs) are recognized to be involved in the control of disease progression. Very recently it has been shown that the overexpression of miR-22 was induced by microbiota-produced butyrate [130]. The study of gut microbiota–miRNA interplay has disclosed that the gut microbiota may affect the host by producing miRNAs, and on the other side the gut microbiota might be regulated by host-secreted miRNAs [131]. An interesting study has highlighted that gut microbiota modulates miRNA-associated mRNA expression patterns in the hippocampus of germ-free mice. In addition, these transcriptional changes are sex-dependent, pointing towards a divergence between molecular pathways that control the gut-brain axis [132]. Some new findings advocate that miRNAs control the TLR-signaling pathway at several stages, including the regulation of TLR mRNA expression, direct activation of the receptor, binding to TLR or TLR-specific signaling pathway components and TLR-induced transcription factors and functional cytokines [133]. The research of the possible relationships between exosomes, miRNAs and TLRs in the nervous systems is still in its infancy. However, we can hypothesize that miRNAs entering the cells via exosomes may finely tune the activation of TLRs. Furthermore, TLR tolerance, a hyporesponsive state of the receptor, characterized by reprogramming of TLR-mediated signal transduction, may be achieved by intracellular delivery of miRNA using exosomes.
Potential pharma- and/or nutraceutical modulators of the microbiota-gut-brain axis as well as TLRs signaling in the progression of pathology in PD are summarized in Table 1.

9. Conclusions

Currently, no treatment for curing PD is available. Levodopa is the primary anti-parkinsonian medicine, which exerts a symptomatic effect but does not stop neurodegeneration and is ineffective on non-motor dysfunctions. Therefore a better understanding of the interaction between TLRs and the enteric microbiota-gut-brain axis might help to generate novel insights into PD pathology, as well as lead to new therapeutic strategies, such as pharmacological or dietary approaches. There is now mounting evidence for the beneficial effects of probiotics on ameliorating intestinal epithelial barrier function, stimulating host homeostasis of the mucosal immune system and preventing pathogenic microbial growth and colonization. Considering that TLR ligands derived from probiotics could suppress inflammation partially through the production of anti-inflammatory cytokines, the use of probiotics, or prebiotics or synbiotics, appears to be an interesting strategy, given their huge potential as medications or prophylactic agents against neurodegeneration. This potential stems from the fact that they exert a beneficial effect on the composition and function of the gut microbiota, restoring the complex dialogue between enteric microbes and the host, and ultimately reestablishing a balanced gut-brain axis.

Author Contributions

V.C. and M.C.G. conceived and wrote the review manuscript.

Funding

The present work was funded by University of Padova (Italy) PRID Grant 2017, University of Padova (Italy) Assegno di Ricerca 2016, San Camillo Hospital, Treviso (Italy) Grant 2015 to M.C.G.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tysnes, O.B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural. Transm. (Vienna) 2017, 124, 901–905. [Google Scholar] [PubMed]
  2. Nair, A.T.; Ramachandran, V.; Joghee, N.M.; Antony, S.; Ramalingam, G. Gut microbiota dysfunction as reliable non-invasive early diagnostic biomarkers in the pathophysiology of Parkinson’s disease: A critical review. J. Neurogastroenterol. Motil. 2018, 24, 30–42. [Google Scholar] [CrossRef] [PubMed]
  3. Dorsey, E.R.; Constantinescu, R.; Thompson, J.P.; Biglan, K.M.; Holloway, R.G.; Kieburtz, K.; Marshall, F.J.; Ravina, B.M.; Schifitto, G.; Siderowf, A.; et al. Projected number of people with parkinson disease in the most populous nations, 2005 through 2030. Neurology 2007, 68, 384–386. [Google Scholar] [CrossRef] [PubMed]
  4. Beach, T.G.; Adler, C.H.; Sue, L.I.; Vedders, L.; Lue, L.; White Iii, C.L.; Akiyama, H.; Caviness, J.N.; Shill, H.A.; Sabbagh, M.N.; et al. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with lewy body disorders. Acta Neuropathol. 2010, 119, 689–702. [Google Scholar] [CrossRef] [PubMed]
  5. Braak, H.; Sastre, M.; Bohl, J.R.; de Vos, R.A.; Del Tredici, K. Parkinson’s disease: Lesions in dorsal horn layer i, involvement of parasympathetic and sympathetic pre- and postganglionic neurons. Acta Neuropathol. 2007, 113, 421–429. [Google Scholar] [CrossRef] [PubMed]
  6. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
  7. Fasano, A.; Visanji, N.P.; Liu, L.W.; Lang, A.E.; Pfeiffer, R.F. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 2015, 14, 625–639. [Google Scholar] [CrossRef]
  8. Poewe, W.; Antonini, A.; Zijlmans, J.C.; Burkhard, P.R.; Vingerhoets, F. Levodopa in the treatment of Parkinson’s disease: An old drug still going strong. Clin. Interv. Aging 2010, 5, 229–238. [Google Scholar] [PubMed]
  9. Shah, E.; Rezaie, A.; Riddle, M.; Pimentel, M. Psychological disorders in gastrointestinal disease: Epiphenomenon, cause or consequence? Ann. Gastroenterol. 2014, 27, 224–230. [Google Scholar] [PubMed]
  10. Fadgyas-Stanculete, M.; Buga, A.M.; Popa-Wagner, A.; Dumitrascu, D.L. The relationship between irritable bowel syndrome and psychiatric disorders: From molecular changes to clinical manifestations. J. Mol. Psychiatry 2014, 2, 4. [Google Scholar] [CrossRef] [PubMed]
  11. Dembinski, A.; Warzecha, Z.; Ceranowicz, P.; Pawlik, M.; Dembinski, M.; Kabat, K.; Konturek, S.J.; Kownacki, P.; Hladki, W.; Pawlik, W.W. Influence of central and peripheral administration of pancreatic polypeptide on gastric mucosa growth. J. Physiol. Pharmacol. 2004, 55, 223–237. [Google Scholar] [PubMed]
  12. Ceranowicz, P.; Cieszkowski, J.; Warzecha, Z.; Dembinski, A. Experimental models of acute pancreatitis. Postepy Hig. Med. Dosw. (Online) 2015, 69, 264–269. [Google Scholar] [CrossRef] [PubMed]
  13. Ceranowicz, P.; Cieszkowski, J.; Warzecha, Z.; Kusnierz-Cabala, B.; Dembinski, A. The beginnings of pancreatology as a field of experimental and clinical medicine. Biomed. Res. Int. 2015, 2015, 128095. [Google Scholar] [CrossRef] [PubMed]
  14. Dumnicka, P.; Maduzia, D.; Ceranowicz, P.; Olszanecki, R.; Drozdz, R.; Kusnierz-Cabala, B. The interplay between inflammation, coagulation and endothelial injury in the early phase of acute pancreatitis: Clinical implications. Int. J. Mol. Sci. 2017, 18, 354. [Google Scholar] [CrossRef] [PubMed]
  15. Moloney, R.D.; O’Mahony, S.M.; Dinan, T.G.; Cryan, J.F. Stress-induced visceral pain: Toward animal models of irritable-bowel syndrome and associated comorbidities. Front. Psychiatry 2015, 6, 15. [Google Scholar] [CrossRef] [PubMed]
  16. Felice, V.D.; Quigley, E.M.; Sullivan, A.M.; O’Keeffe, G.W.; O’Mahony, S.M. Microbiota-gut-brain signalling in Parkinson’s disease: Implications for non-motor symptoms. Parkinsonism Relat. Disord. 2016, 27, 1–8. [Google Scholar] [CrossRef] [PubMed]
  17. Straub, R.H.; Wiest, R.; Strauch, U.G.; Harle, P.; Scholmerich, J. The role of the sympathetic nervous system in intestinal inflammation. Gut 2006, 55, 1640–1649. [Google Scholar] [CrossRef] [PubMed]
  18. Costantini, T.W.; Baird, A. Lost your nerve? Modulating the parasympathetic nervous system to treat inflammatory bowel disease. J. Physiol. 2016, 594, 4097–4098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Warzecha, Z.; Dembinski, A.; Ceranowicz, P.; Konturek, P.C.; Stachura, J.; Tomaszewska, R.; Konturek, S.J. Calcitonin gene-related peptide can attenuate or augment pancreatic damage in caerulein-induced pancreatitis in rats. J. Physiol. Pharmacol. 1999, 50, 49–62. [Google Scholar] [PubMed]
  20. Warzecha, Z.; Dembinski, A.; Ceranowicz, P.; Stachura, J.; Tomaszewska, R.; Konturek, S.J. Effect of sensory nerves and cgrp on the development of caerulein-induced pancreatitis and pancreatic recovery. J. Physiol. Pharmacol. 2001, 52, 679–704. [Google Scholar] [PubMed]
  21. Dembinski, A.; Warzecha, Z.; Ceranowicz, P.; Jaworek, J.; Sendur, R.; Knafel, A.; Dembinski, M.; Bilski, J.; Pawlik, W.W.; Tomaszewska, R.; et al. Stimulation of sensory nerves and cgrp attenuate pancreatic damage in ischemia/reperfusion induced pancreatitis. Med. Sci. Monit. 2003, 9, BR418–BR425. [Google Scholar] [PubMed]
  22. Dembinski, A.; Warzecha, Z.; Ceranowicz, P.; Konturek, S.J. The role of capsaicin-sensitive sensory neurons and nitric oxide in regulation of gastric mucosal growth. J. Physiol. Pharmacol. 1995, 46, 351–362. [Google Scholar] [PubMed]
  23. Dembinski, A.; Warzecha, Z.; Konturek, P.J.; Ceranowicz, P.; Konturek, S.J. Influence of capsaicin-sensitive afferent neurons and nitric oxide (no) on cerulein-induced pancreatitis in rats. Int. J. Pancreatol. 1996, 19, 179–189. [Google Scholar] [PubMed]
  24. Warzecha, Z.; Dembinski, A.; Jaworek, J.; Ceranowicz, P.; Szlachcic, A.; Walocha, J.; Konturek, S.J. Role of sensory nerves in pancreatic secretion and caerulein-induced pancreatitis. J. Physiol. Pharmacol. 1997, 48, 43–58. [Google Scholar] [PubMed]
  25. Warzecha, Z.; Dembinski, A.; Ceranowicz, P.; Dembinski, M.; Cieszkowski, J.; Kownacki, P.; Konturek, P.C. Role of sensory nerves in gastroprotective effect of anandamide in rats. J. Physiol. Pharmacol. 2011, 62, 207–217. [Google Scholar] [PubMed]
  26. Ramos, L.R.; Sachar, D.B.; DiMaio, C.J.; Colombel, J.F.; Torres, J. Inflammatory bowel disease and pancreatitis: A review. J. Crohn’s Colitis 2016, 10, 95–104. [Google Scholar] [CrossRef] [PubMed]
  27. Dembinski, A.; Warzecha, Z.; Ceranowicz, P.; Dembinski, M.; Cieszkowski, J.; Pawlik, W.W.; Konturek, S.J.; Tomaszewska, R.; Hladki, W.; Konturek, P.C. Cannabinoids in acute gastric damage and pancreatitis. J. Physiol. Pharmacol. 2006, 57 (Suppl. S5), 137–154. [Google Scholar] [PubMed]
  28. Dembinski, A.; Warzecha, Z.; Ceranowicz, P.; Warzecha, A.M.; Pawlik, W.W.; Dembinski, M.; Rembiasz, K.; Sendur, P.; Kusnierz-Cabala, B.; Tomaszewska, R.; et al. Dual, time-dependent deleterious and protective effect of anandamide on the course of cerulein-induced acute pancreatitis. Role of sensory nerves. Eur. J. Pharmacol. 2008, 591, 284–292. [Google Scholar] [CrossRef] [PubMed]
  29. Endres, K.; Schafer, K.H. Influence of commensal microbiota on the enteric nervous system and its role in neurodegenerative diseases. J. Innate Immun. 2018, 1–9. [Google Scholar] [CrossRef] [PubMed]
  30. Kubinak, J.L.; Round, J.L. Toll-like receptors promote mutually beneficial commensal-host interactions. PLoS Pathog. 2012, 8, e1002785. [Google Scholar] [CrossRef] [PubMed]
  31. Cani, P.D. Gut microbiota—At the intersection of everything? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 321–322. [Google Scholar] [CrossRef] [PubMed]
  32. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
  33. Sender, R.; Fuchs, S.; Milo, R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef] [PubMed]
  34. Costea, P.I.; Hildebrand, F.; Arumugam, M.; Backhed, F.; Blaser, M.J.; Bushman, F.D.; de Vos, W.M.; Ehrlich, S.D.; Fraser, C.M.; Hattori, M.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [PubMed]
  35. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed]
  36. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ubeda, C.; Pamer, E.G. Antibiotics, microbiota, and immune defense. Trends Immunol. 2012, 33, 459–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the immune system. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef] [PubMed]
  39. Dembinski, A.; Warzecha, Z.; Ceranowicz, P.; Dembinski, M.; Cieszkowski, J.; Gosiewski, T.; Bulanda, M.; Kusnierz-Cabala, B.; Galazka, K.; Konturek, P.C. Synergic interaction of rifaximin and mutaflor (escherichia coli nissle 1917) in the treatment of acetic acid-induced colitis in rats. Gastroenterol. Res. Pract. 2016, 2016, 3126280. [Google Scholar] [CrossRef] [PubMed]
  40. Jaworek, J.; Tudek, B.; Kowalczyk, P.; Kot, M.; Szklarczyk, J.; Leja-Szpak, A.; Pierzchalski, P.; Bonior, J.; Dembinski, A.; Ceranowicz, P.; et al. Effect of endotoxemia in suckling rats on pancreatic integrity and exocrine function in adults: A review report. Gastroenterol. Res. Pract. 2018, 2018, 6915059. [Google Scholar] [CrossRef] [PubMed]
  41. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef] [PubMed]
  42. Foster, J.A.; McVey Neufeld, K.A. Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef] [PubMed]
  43. Mazzoli, R.; Pessione, E. The neuro-endocrinological role of microbial glutamate and gaba signaling. Front. Microbiol. 2016, 7, 1934. [Google Scholar] [CrossRef] [PubMed]
  44. Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. Bioessays 2011, 33, 574–581. [Google Scholar] [CrossRef] [PubMed]
  45. Sivamaruthi, B.S.; Madhumita, R.; Balamurugan, K.; Rajan, K.E. Cronobacter sakazakii infection alters serotonin transporter and improved fear memory retention in the rat. Front. Pharmacol. 2015, 6, 188. [Google Scholar] [CrossRef] [PubMed]
  46. Ceranowicz, P.; Warzecha, Z.; Dembinski, A. Peptidyl hormones of endocrine cells origin in the gut—Their discovery and physiological relevance. J. Physiol. Pharmacol. 2015, 66, 11–27. [Google Scholar] [PubMed]
  47. Bellono, N.W.; Bayrer, J.R.; Leitch, D.B.; Castro, J.; Zhang, C.; O’Donnell, T.A.; Brierley, S.M.; Ingraham, H.A.; Julius, D. Enterochromaffin cells are gut chemosensors that couple to sensory neural pathways. Cell 2017, 170, 185–198.e116. [Google Scholar] [CrossRef] [PubMed]
  48. O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef] [PubMed]
  49. Alam, R.; Abdolmaleky, H.M.; Zhou, J.R. Microbiome, inflammation, epigenetic alterations, and mental diseases. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2017, 174, 651–660. [Google Scholar] [CrossRef] [PubMed]
  50. Galland, L. The gut microbiome and the brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef] [PubMed]
  51. Yang, N.J.; Chiu, I.M. Bacterial signaling to the nervous system through toxins and metabolites. J. Mol. Biol. 2017, 429, 587–605. [Google Scholar] [CrossRef] [PubMed]
  52. Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef] [PubMed]
  53. Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609–10620. [Google Scholar] [CrossRef] [PubMed]
  54. Tomik, J.; Tomik, B.; Gajec, S.; Ceranowicz, P.; Pihut, M.; Olszanecki, R.; Strek, P.; Skladzien, J. The balloon-based manometry evaluation of swallowing in patients with amyotrophic lateral sclerosis. Int. J. Mol. Sci. 2017, 18, 707. [Google Scholar] [CrossRef] [PubMed]
  55. Warzecha, Z.; Dembinski, A.; Ceranowicz, P.; Dembinski, M.; Sendur, R.; Pawlik, W.W.; Konturek, S.J. Deleterious effect of helicobacter pylori infection on the course of acute pancreatitis in rats. Pancreatology 2002, 2, 386–395. [Google Scholar] [CrossRef] [PubMed]
  56. Pellegrini, C.; Colucci, R.; Antonioli, L.; Barocelli, E.; Ballabeni, V.; Bernardini, N.; Blandizzi, C.; de Jonge, W.J.; Fornai, M. Intestinal dysfunction in Parkinson’s disease: Lessons learned from translational studies and experimental models. Neurogastroenterol. Motil. 2016, 28, 1781–1791. [Google Scholar] [CrossRef] [PubMed]
  57. Braak, H.; de Vos, R.A.; Bohl, J.; Del Tredici, K. Gastric alpha-synuclein immunoreactive inclusions in meissner’s and auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci. Lett. 2006, 396, 67–72. [Google Scholar] [CrossRef] [PubMed]
  58. Svensson, E.; Horvath-Puho, E.; Thomsen, R.W.; Djurhuus, J.C.; Pedersen, L.; Borghammer, P.; Sorensen, H.T. Vagotomy and subsequent risk of Parkinson’s disease. Ann. Neurol. 2015, 78, 522–529. [Google Scholar] [CrossRef] [PubMed]
  59. Pan-Montojo, F.; Schwarz, M.; Winkler, C.; Arnhold, M.; O’Sullivan, G.A.; Pal, A.; Said, J.; Marsico, G.; Verbavatz, J.M.; Rodrigo-Angulo, M.; et al. Environmental toxins trigger pd-like progression via increased alpha-synuclein release from enteric neurons in mice. Sci. Rep. 2012, 2, 898. [Google Scholar] [CrossRef] [PubMed]
  60. Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS ONE 2011, 6, e28032. [Google Scholar] [CrossRef] [PubMed]
  61. Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef] [PubMed]
  62. Banks, W.A.; Erickson, M.A. The blood-brain barrier and immune function and dysfunction. Neurobiol. Dis. 2010, 37, 26–32. [Google Scholar] [CrossRef] [PubMed]
  63. 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] [PubMed]
  64. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Backhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [PubMed]
  65. Andrews, Z.B.; Erion, D.; Beiler, R.; Liu, Z.W.; Abizaid, A.; Zigman, J.; Elsworth, J.D.; Savitt, J.M.; DiMarchi, R.; Tschoep, M.; et al. Ghrelin promotes and protects nigrostriatal dopamine function via a ucp2-dependent mitochondrial mechanism. J. Neurosci. 2009, 29, 14057–14065. [Google Scholar] [CrossRef] [PubMed]
  66. Unger, M.M.; Moller, J.C.; Mankel, K.; Eggert, K.M.; Bohne, K.; Bodden, M.; Stiasny-Kolster, K.; Kann, P.H.; Mayer, G.; Tebbe, J.J.; et al. Postprandial ghrelin response is reduced in patients with Parkinson’s disease and idiopathic rem sleep behaviour disorder: A peripheral biomarker for early Parkinson’s disease? J. Neurol. 2011, 258, 982–990. [Google Scholar] [CrossRef] [PubMed]
  67. 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.e1412. [Google Scholar] [CrossRef] [PubMed]
  68. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [PubMed]
  69. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [PubMed]
  70. Medzhitov, R.; Janeway, C., Jr. Innate immunity. N. Engl. J. Med. 2000, 343, 338–344. [Google Scholar] [CrossRef] [PubMed]
  71. Hashimoto, C.; Hudson, K.L.; Anderson, K.V. The toll gene of drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell 1988, 52, 269–279. [Google Scholar] [CrossRef]
  72. Brun, P.; Giron, M.C.; Qesari, M.; Porzionato, A.; Caputi, V.; Zoppellaro, C.; Banzato, S.; Grillo, A.R.; Spagnol, L.; De Caro, R.; et al. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology 2013, 145, 1323–1333. [Google Scholar] [CrossRef] [PubMed]
  73. Brun, P.; Gobbo, S.; Caputi, V.; Spagnol, L.; Schirato, G.; Pasqualin, M.; Levorato, E.; Palu, G.; Giron, M.C.; Castagliuolo, I. Toll like receptor-2 regulates production of glial-derived neurotrophic factors in murine intestinal smooth muscle cells. Mol. Cell. Neurosci. 2015, 68, 24–35. [Google Scholar] [CrossRef] [PubMed]
  74. Okun, E.; Griffioen, K.J.; Mattson, M.P. Toll-like receptor signaling in neural plasticity and disease. Trends Neurosci. 2011, 34, 269–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Prehaud, C.; Megret, F.; Lafage, M.; Lafon, M. Virus infection switches tlr-3-positive human neurons to become strong producers of beta interferon. J. Virol. 2005, 79, 12893–12904. [Google Scholar] [CrossRef] [PubMed]
  76. Barajon, I.; Serrao, G.; Arnaboldi, F.; Opizzi, E.; Ripamonti, G.; Balsari, A.; Rumio, C. Toll-like receptors 3, 4, and 7 are expressed in the enteric nervous system and dorsal root ganglia. J. Histochem. Cytochem. 2009, 57, 1013–1023. [Google Scholar] [CrossRef] [PubMed]
  77. Kawasaki, T.; Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 2014, 5, 461. [Google Scholar] [CrossRef] [PubMed]
  78. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed]
  79. Kim, C.; Ho, D.H.; Suk, J.E.; You, S.; Michael, S.; Kang, J.; Joong Lee, S.; Masliah, E.; Hwang, D.; Lee, H.J.; et al. Neuron-released oligomeric alpha-synuclein is an endogenous agonist of tlr2 for paracrine activation of microglia. Nat. Commun. 2013, 4, 1562. [Google Scholar] [CrossRef] [PubMed]
  80. Daniele, S.G.; Beraud, D.; Davenport, C.; Cheng, K.; Yin, H.; Maguire-Zeiss, K.A. Activation of myd88-dependent tlr1/2 signaling by misfolded alpha-synuclein, a protein linked to neurodegenerative disorders. Sci. Signal. 2015, 8, ra45. [Google Scholar] [CrossRef] [PubMed]
  81. Beraud, D.; Maguire-Zeiss, K.A. Misfolded alpha-synuclein and toll-like receptors: Therapeutic targets for Parkinson’s disease. Parkinsonism. Relat. Disord. 2012, 18 (Suppl. S1), S17–S20. [Google Scholar] [CrossRef]
  82. Fellner, L.; Irschick, R.; Schanda, K.; Reindl, M.; Klimaschewski, L.; Poewe, W.; Wenning, G.K.; Stefanova, N. Toll-like receptor 4 is required for alpha-synuclein dependent activation of microglia and astroglia. Glia 2013, 61, 349–360. [Google Scholar] [CrossRef] [PubMed]
  83. Noelker, C.; Morel, L.; Lescot, T.; Osterloh, A.; Alvarez-Fischer, D.; Breloer, M.; Henze, C.; Depboylu, C.; Skrzydelski, D.; Michel, P.P.; et al. Toll like receptor 4 mediates cell death in a mouse mptp model of parkinson disease. Sci. Rep. 2013, 3, 1393. [Google Scholar] [CrossRef] [PubMed]
  84. Yan, Y.; Jiang, W.; Liu, L.; Wang, X.; Ding, C.; Tian, Z.; Zhou, R. Dopamine controls systemic inflammation through inhibition of nlrp3 inflammasome. Cell 2015, 160, 62–73. [Google Scholar] [CrossRef] [PubMed]
  85. Codolo, G.; Plotegher, N.; Pozzobon, T.; Brucale, M.; Tessari, I.; Bubacco, L.; de Bernard, M. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS ONE 2013, 8, e55375. [Google Scholar] [CrossRef] [PubMed]
  86. Labzin, L.I.; Heneka, M.T.; Latz, E. Innate immunity and neurodegeneration. Annu. Rev. Med. 2018, 69, 437–449. [Google Scholar] [CrossRef] [PubMed]
  87. Rietdijk, C.D.; van Wezel, R.J.; Garssen, J.; Kraneveld, A.D. Neuronal toll-like receptors and neuro-immunity in Parkinson’s disease, Alzheimer’s disease and stroke. Neuroimmunol. Neuroinflamm. 2016, 3, 27–37. [Google Scholar] [CrossRef]
  88. Drouin-Ouellet, J.; Cicchetti, F. Inflammation and neurodegeneration: The story ‘retolled’. Trends Pharmacol. Sci. 2012, 33, 542–551. [Google Scholar] [CrossRef] [PubMed]
  89. Mosher, K.I.; Wyss-Coray, T. Microglial dysfunction in brain aging and Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 594–604. [Google Scholar] [CrossRef] [PubMed]
  90. Yanez, A.; Hassanzadeh-Kiabi, N.; Ng, M.Y.; Megias, J.; Subramanian, A.; Liu, G.Y.; Underhill, D.M.; Gil, M.L.; Goodridge, H.S. Detection of a tlr2 agonist by hematopoietic stem and progenitor cells impacts the function of the macrophages they produce. Eur. J. Immunol. 2013, 43, 2114–2125. [Google Scholar] [CrossRef] [PubMed]
  91. Houser, M.C.; Tansey, M.G. The gut-brain axis: Is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinsons Dis. 2017, 3, 3. [Google Scholar] [CrossRef] [PubMed]
  92. Stefanova, N.; Fellner, L.; Reindl, M.; Masliah, E.; Poewe, W.; Wenning, G.K. Toll-like receptor 4 promotes alpha-synuclein clearance and survival of nigral dopaminergic neurons. Am. J. Pathol. 2011, 179, 954–963. [Google Scholar] [CrossRef] [PubMed]
  93. Friedland, R.P. Mechanisms of molecular mimicry involving the microbiota in neurodegeneration. J. Alzheimers Dis. 2015, 45, 349–362. [Google Scholar] [CrossRef] [PubMed]
  94. Caputi, V.; Marsilio, I.; Filpa, V.; Cerantola, S.; Orso, G.; Bistoletti, M.; Paccagnella, N.; De Martin, S.; Montopoli, M.; Dall’Acqua, S.; et al. Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice. Br. J. Pharmacol. 2017, 174, 3623–3639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Latorre, E.; Layunta, E.; Grasa, L.; Castro, M.; Pardo, J.; Gomollon, F.; Alcalde, A.I.; Mesonero, J.E. Intestinal serotonin transporter inhibition by toll-like receptor 2 activation. A feedback modulation. PLoS ONE 2016, 11, e0169303. [Google Scholar] [CrossRef] [PubMed]
  96. Caputi, V.; Marsilio, I.; Cerantola, S.; Roozfarakh, M.; Lante, I.; Galuppini, F.; Rugge, M.; Napoli, E.; Giulivi, C.; Orso, G.; et al. Toll-like receptor 4 modulates small intestine neuromuscular function through nitrergic and purinergic pathways. Front. Pharmacol. 2017, 8, 350. [Google Scholar] [CrossRef] [PubMed]
  97. Yan, X.; Jiang, E.; Weng, H.R. Activation of toll like receptor 4 attenuates gaba synthesis and postsynaptic gaba receptor activities in the spinal dorsal horn via releasing interleukin-1 beta. J. Neuroinflamm. 2015, 12, 222. [Google Scholar] [CrossRef] [PubMed]
  98. Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the cns. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
  99. Larraufie, P.; Dore, J.; Lapaque, N.; Blottiere, H.M. Tlr ligands and butyrate increase pyy expression through two distinct but inter-regulated pathways. Cell. Microbiol. 2017, 19. [Google Scholar] [CrossRef] [PubMed]
  100. Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The impact of the gut microbiota on human health: An integrative view. Cell 2012, 148, 1258–1270. [Google Scholar] [CrossRef] [PubMed]
  101. Maslowski, K.M.; Mackay, C.R. Diet, gut microbiota and immune responses. Nat. Immunol. 2011, 12, 5–9. [Google Scholar] [CrossRef] [PubMed]
  102. Perez-Pardo, P.; Dodiya, H.B.; Broersen, L.M.; Douna, H.; van Wijk, N.; Lopes da Silva, S.; Garssen, J.; Keshavarzian, A.; Kraneveld, A.D. Gut-brain and brain-gut axis in Parkinson’s disease models: Effects of a uridine and fish oil diet. Nutr. Neurosci. 2017, 1–12. [Google Scholar] [CrossRef] [PubMed]
  103. Perez-Pardo, P.; Kliest, T.; Dodiya, H.B.; Broersen, L.M.; Garssen, J.; Keshavarzian, A.; Kraneveld, A.D. The gut-brain axis in Parkinson’s disease: Possibilities for food-based therapies. Eur. J. Pharmacol. 2017, 817, 86–95. [Google Scholar] [CrossRef] [PubMed]
  104. Miller, R.L.; James-Kracke, M.; Sun, G.Y.; Sun, A.Y. Oxidative and inflammatory pathways in Parkinson’s disease. Neurochem. Res. 2009, 34, 55–65. [Google Scholar] [CrossRef] [PubMed]
  105. Afshordel, S.; Hagl, S.; Werner, D.; Rohner, N.; Kogel, D.; Bazan, N.G.; Eckert, G.P. Omega-3 polyunsaturated fatty acids improve mitochondrial dysfunction in brain aging—Impact of bcl-2 and npd-1 like metabolites. Prostaglandins Leukot. Essent. Fatty Acids 2015, 92, 23–31. [Google Scholar] [CrossRef] [PubMed]
  106. Bazan, N.G.; Molina, M.F.; Gordon, W.C. Docosahexaenoic acid signalolipidomics in nutrition: Significance in aging, neuroinflammation, macular degeneration, alzheimer’s, and other neurodegenerative diseases. Ann. Rev. Nutr. 2011, 31, 321–351. [Google Scholar] [CrossRef] [PubMed]
  107. Coulombe, K.; Kerdiles, O.; Tremblay, C.; Emond, V.; Lebel, M.; Boulianne, A.S.; Plourde, M.; Cicchetti, F.; Calon, F. Impact of dha intake in a mouse model of synucleinopathy. Exp. Neurol. 2018, 301, 39–49. [Google Scholar] [CrossRef] [PubMed]
  108. Hwang, D.H.; Kim, J.A.; Lee, J.Y. Mechanisms for the activation of toll-like receptor 2/4 by saturated fatty acids and inhibition by docosahexaenoic acid. Eur. J. Pharmacol. 2016, 785, 24–35. [Google Scholar] [CrossRef] [PubMed]
  109. Lee, J.Y.; Zhao, L.; Hwang, D.H. Modulation of pattern recognition receptor-mediated inflammation and risk of chronic diseases by dietary fatty acids. Nutr. Rev. 2010, 68, 38–61. [Google Scholar] [CrossRef] [PubMed]
  110. Beamer, C.A.; Shepherd, D.M. Inhibition of tlr ligand- and interferon gamma-induced murine microglial activation by panax notoginseng. J. Neuroimmune Pharmacol. 2012, 7, 465–476. [Google Scholar] [CrossRef] [PubMed]
  111. Haddadi, R.; Nayebi, A.M.; Eyvari Brooshghalan, S. Silymarin prevents apoptosis through inhibiting the bax/caspase-3 expression and suppresses toll like receptor-4 pathway in the snc of 6-ohda intoxicated rats. Biomed. Pharmacother. 2018, 104, 127–136. [Google Scholar] [CrossRef] [PubMed]
  112. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  113. Derrien, M.; van Hylckama Vlieg, J.E. Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol. 2015, 23, 354–366. [Google Scholar] [CrossRef] [PubMed]
  114. Sanders, M.E.; Benson, A.; Lebeer, S.; Merenstein, D.J.; Klaenhammer, T.R. Shared mechanisms among probiotic taxa: Implications for general probiotic claims. Curr. Opin. Biotechnol. 2018, 49, 207–216. [Google Scholar] [CrossRef] [PubMed]
  115. Wallace, C.J.K.; Milev, R. The effects of probiotics on depressive symptoms in humans: A systematic review. Ann. Gen. Psychiatry 2017, 16, 14. [Google Scholar] [CrossRef] [PubMed]
  116. Abildgaard, A.; Elfving, B.; Hokland, M.; Wegener, G.; Lund, S. Probiotic treatment reduces depressive-like behaviour in rats independently of diet. Psychoneuroendocrinology 2017, 79, 40–48. [Google Scholar] [CrossRef] [PubMed]
  117. Sun, M.F.; Shen, Y.Q. Dysbiosis of gut microbiota and microbial metabolites in Parkinson’s disease. Ageing Res. Rev. 2018, 45, 53–61. [Google Scholar] [CrossRef] [PubMed]
  118. 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] [PubMed]
  119. 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: An rct. Neurology 2016, 87, 1274–1280. [Google Scholar] [CrossRef] [PubMed]
  120. 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]
  121. Ojetti, V.; Ianiro, G.; Tortora, A.; D’Angelo, G.; Di Rienzo, T.A.; Bibbo, S.; Migneco, A.; Gasbarrini, A. The effect of lactobacillus reuteri supplementation in adults with chronic functional constipation: A randomized, double-blind, placebo-controlled trial. J. Gastrointest. Liver Dis. JGLD 2014, 23, 387–391. [Google Scholar]
  122. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The international scientific association for probiotics and prebiotics (isapp) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  123. Rasmussen, H.E.; Piazza, B.R.; Forsyth, C.B.; Keshavarzian, A. Nutrition and gastrointestinal health as modulators of Parkinson’s disease. In Pharma-Nutrition: An Overview; Folkerts, G., Garssen, J., Eds.; Springer International Publishing: Cham, Germany, 2014; pp. 213–242. [Google Scholar]
  124. Savignac, H.M.; Corona, G.; Mills, H.; Chen, L.; Spencer, J.P.; Tzortzis, G.; Burnet, P.W. Prebiotic feeding elevates central brain derived neurotrophic factor, n-methyl-d-aspartate receptor subunits and d-serine. Neurochem. Int. 2013, 63, 756–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Barboza, J.L.; Okun, M.S.; Moshiree, B. The treatment of gastroparesis, constipation and small intestinal bacterial overgrowth syndrome in patients with Parkinson’s disease. Expert Opin. Pharmacother. 2015, 16, 2449–2464. [Google Scholar] [CrossRef] [PubMed]
  126. Parashar, A.; Udayabanu, M. Gut microbiota: Implications in Parkinson’s disease. Parkinsonism Relat. Disord. 2017, 38, 1–7. [Google Scholar] [CrossRef] [PubMed]
  127. Villena, J.; Kitazawa, H. Modulation of intestinal tlr4-inflammatory signaling pathways by probiotic microorganisms: Lessons learned from lactobacillus jensenii tl2937. Front. Immunol. 2014, 4, 512. [Google Scholar] [CrossRef] [PubMed]
  128. Barochia, A.; Solomon, S.; Cui, X.; Natanson, C.; Eichacker, P.Q. Eritoran tetrasodium (e5564) treatment for sepsis: Review of preclinical and clinical studies. Expert Opin. Drug Metab. Toxicol. 2011, 7, 479–494. [Google Scholar] [CrossRef] [PubMed]
  129. Schwenkgrub, J.; Zaremba, M.; Joniec-Maciejak, I.; Cudna, A.; Mirowska-Guzel, D.; Kurkowska-Jastrzebska, I. The phosphodiesterase inhibitor, ibudilast, attenuates neuroinflammation in the mptp model of Parkinson’s disease. PLoS ONE 2017, 12, e0182019. [Google Scholar] [CrossRef] [PubMed]
  130. Pant, K.; Yadav, A.K.; Gupta, P.; Islam, R.; Saraya, A.; Venugopal, S.K. Butyrate induces ros-mediated apoptosis by modulating mir-22/sirt-1 pathway in hepatic cancer cells. Redox Biol. 2017, 12, 340–349. [Google Scholar] [CrossRef] [PubMed]
  131. Feng, Q.; Chen, W.D.; Wang, Y.D. Gut microbiota: An integral moderator in health and disease. Front. Microbiol. 2018, 9, 151. [Google Scholar] [CrossRef] [PubMed]
  132. Moloney, G.M.; O’Leary, O.F.; Salvo-Romero, E.; Desbonnet, L.; Shanahan, F.; Dinan, T.G.; Clarke, G.; Cryan, J.F. Microbial regulation of hippocampal mirna expression: Implications for transcription of kynurenine pathway enzymes. Behav. Brain Res. 2017, 334, 50–54. [Google Scholar] [CrossRef] [PubMed]
  133. Paschon, V.; Takada, S.H.; Ikebara, J.M.; Sousa, E.; Raeisossadati, R.; Ulrich, H.; Kihara, A.H. Interplay between exosomes, micrornas and toll-like receptors in brain disorders. Mol. Neurobiol. 2016, 53, 2016–2028. [Google Scholar] [CrossRef] [PubMed]
  134. Tang, R.; Lin, Y.M.; Liu, H.X.; Wang, E.S. Neuroprotective effect of docosahexaenoic acid in rat traumatic brain injury model via regulation of tlr4/nf-kappa b signaling pathway. Int. J. Biochem. Cell Biol. 2018, 99, 64–71. [Google Scholar] [CrossRef] [PubMed]
  135. Kumar, H.; Kawai, T.; Akira, S. Toll-like receptors and innate immunity. Biochem. Biophys. Res. Commun. 2009, 388, 621–625. [Google Scholar] [CrossRef] [PubMed]
  136. Alizadeh, A.; Akbari, P.; Difilippo, E.; Schols, H.A.; Ulfman, L.H.; Schoterman, M.H.; Garssen, J.; Fink-Gremmels, J.; Braber, S. The piglet as a model for studying dietary components in infant diets: Effects of galacto-oligosaccharides on intestinal functions. Br. J. Nutr. 2016, 115, 605–618. [Google Scholar] [CrossRef] [PubMed]
  137. Riviere, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and butyrate-producing colon bacteria: Importance and strategies for their stimulation in the human gut. Front. Microbiol. 2016, 7, 979. [Google Scholar] [CrossRef] [PubMed]
Table 1. Pharma- and/or nutraceuticals as potential modulators of the microbiota-gut-brain axis and Toll-like receptors (TLRs) signaling in the progression of pathology in Parkinson’s disease (PD).
Table 1. Pharma- and/or nutraceuticals as potential modulators of the microbiota-gut-brain axis and Toll-like receptors (TLRs) signaling in the progression of pathology in Parkinson’s disease (PD).
ModulatorInfluence on Microbiota-Gut-Brain Axis Potential TLRs TargetReferences
Dietary supplements
Docosahexaenoic acid (DHA)Reduction of oxidative stress by improving neuronal mitochondrial dysfunctionTLR2, TLR4[104,108,134]
Panax Notoginseng (NotoG)Suppression of microglial activation and reduction of IL-6 and TNF-α releaseTLR4[110]
SylimarinAntioxidant and neuroprotective effects (salvaging of free radicals)TLR4[111]
Probiotics
L. rhamnosus (JB-1)Modulation of GABAA and GABAB receptors in the brainTLR1, TLR2, TLR6[118,135]
Lactobacillus casei ShirotaDecrease of visceral pain and bloating; improvement of stool consistencyTLR1, TLR2, TLR6[120,135]
Lactobacillus reuteriImprovement of bowel movement and increase of the frequency of evacuationTLR1, TLR2, TLR6[121,135]
Prebiotics
Fructo oligosaccharides (FOS)Increase of BDNF expression in the hippocampusTLR2, TLR4[73,124,135]
Galacto oligosaccharides (GOS)Improvement of villus surface area in the small intestine TLR1-TLR13[135,136]
Short-chain fatty acids (SCFA)-ButyrateMaintenance of colonic epithelium integrityTLR1, TLR2, TLR6[135,137]
Antibiotics
RifaximinTreatment of small intestinal overgrowthTLR4[125]
MynociclineNeuroprotective effect on nigrostriatal dopaminergic neuronsTLR4[126]
TLRs modulators
Eritoran tetrasodiumInhibition of LPS-induced proinflammatory cytokine releaseTLR4[128]
IbudilastImprovement of anti-inflammatory cytokine release, modulation of glial cells activity and induction of GDNF expressionTLR4[129]
MiR-22Induced by butyrate-producing commensal bacteriaTLR1, TLR2, TLR6[130,135]

Share and Cite

MDPI and ACS Style

Caputi, V.; Giron, M.C. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease. Int. J. Mol. Sci. 2018, 19, 1689. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19061689

AMA Style

Caputi V, Giron MC. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease. International Journal of Molecular Sciences. 2018; 19(6):1689. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19061689

Chicago/Turabian Style

Caputi, Valentina, and Maria Cecilia Giron. 2018. "Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease" International Journal of Molecular Sciences 19, no. 6: 1689. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19061689

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