Next Article in Journal
The Associations of Single Nucleotide Polymorphisms with Risk and Symptoms of Irritable Bowel Syndrome
Next Article in Special Issue
Optimizing Treatment of Parkinson’s Disease
Previous Article in Journal
Locoregional Recurrence Prediction Using a Deep Neural Network of Radiological and Radiotherapy Images
Previous Article in Special Issue
Analyzing the Effects of Parameters for Tremor Modulation via Phase-Locked Electrical Stimulation on a Peripheral Nerve
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gastrointestinal Dysfunction in Parkinson’s Disease: Current and Potential Therapeutics

by
Myat Noe Han
1,2,3,
David I. Finkelstein
4,
Rachel M. McQuade
1,2,3,*,† and
Shanti Diwakarla
1,2,3,†
1
Gut-Axis Injury and Repair Laboratory, Department of Medicine Western Health, University of Melbourne, Melbourne, VIC 3021, Australia
2
The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
3
Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
4
Parkinson’s Disease Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Pers. Med. 2022, 12(2), 144; https://doi.org/10.3390/jpm12020144
Submission received: 24 December 2021 / Revised: 17 January 2022 / Accepted: 18 January 2022 / Published: 21 January 2022
(This article belongs to the Special Issue Optimizing Treatment of Parkinson’s Disease)

Abstract

:
Abnormalities in the gastrointestinal (GI) tract of Parkinson’s disease (PD) sufferers were first reported over 200 years ago; however, the extent and role of GI dysfunction in PD disease progression is still unknown. GI dysfunctions, including dysphagia, gastroparesis, and constipation, are amongst the most prevalent non-motor symptoms in PD. These symptoms not only impact patient quality of life, but also complicate disease management. Conventional treatment pathways for GI dysfunctions (i.e., constipation), such as increasing fibre and fluid intake, and the use of over-the-counter laxatives, are generally ineffective in PD patients, and approved compounds such as guanylate cyclase C agonists and selective 5-hyroxytryptamine 4 receptor agonists have demonstrated limited efficacy. Thus, identification of potential targets for novel therapies to alleviate PD-induced GI dysfunctions are essential to improve clinical outcomes and quality of life in people with PD. Unlike the central nervous system (CNS), where PD pathology and the mechanisms involved in CNS damage are relatively well characterised, the effect of PD at the cellular and tissue level in the enteric nervous system (ENS) remains unclear, making it difficult to alleviate or reverse GI symptoms. However, the resurgence of interest in understanding how the GI tract is involved in various disease states, such as PD, has resulted in the identification of novel therapeutic avenues. This review focuses on common PD-related GI symptoms, and summarizes the current treatments available and their limitations. We propose that by targeting the intestinal barrier, ENS, and/or the gut microbiome, may prove successful in alleviating PD-related GI symptoms, and discuss emerging therapies and potential drugs that could be repurposed to target these areas.

1. Introduction

Parkinson’s disease (PD) is the second most frequent neurological disease after Alzheimer’s disease, and affects 1% of the population above 60 years of age worldwide [1]. Clinical hallmarks of the disease include resting tremor, rigidity, bradykinesia, and postural instability, which is primarily attributed to the progressive degeneration of dopamine-containing neurons in the substantia nigra pars compacta (SNpc) [2,3,4]. In the majority of PD cases, the presence of Lewy bodies (LB) in the SNpc, which contain aggregates of the protein alpha-synuclein, serve as a pathological hallmark of the disease. However, LBs are also found in other regions of the central nervous system (CNS), such as the peripheral and enteric nervous system (ENS) [5,6], and may play a role in the onset of non-motor symptoms associated with the disease.
Most PD patients develop some form of autonomic disorder involving the cardiovascular, genitourinary, thermoregulatory, and gastrointestinal (GI) systems [7]. GI dysfunctions are a common occurrence in PD patients and can include symptoms such as dysphagia (difficulty swallowing), gastroparesis (slowed gastric emptying), and chronic constipation. Due to limitations associated with methods of detection, studies being performed in heterogenous patient populations, and a lack of patient reporting, the exact prevalence of GI dysfunctions in PD patients is difficult to determine. However, it is clear that some forms of GI dysfunction precede the onset of motor deficits by decades [8] and greatly impact on patient quality of life.
Gut dysfunction contributes directly to the morbidity of PD, and complicates clinical management. Weight loss has been found to precede PD diagnosis by years, and is associated with disease severity as well as duration [9], whilst low body mass index has a significant impact on the life prognosis of PD patients, particularly males [10]. Nausea, resulting from delayed stomach emptying, is thought to contribute to weight loss, whilst delayed colonic transit time due to poor motility causes harder stools and constipation by increasing the absorption of water.
Whilst motor symptoms can be managed using levodopa—a dopamine prodrug—GI symptoms often remain unaffected, suggesting that the loss of dopamine does not drive GI symptom onset [11,12]. The presence of LBs in the ENS has led to the suggestion that enteric neuronal loss and/or dysfunction may contribute to the development of GI symptoms. The ENS is a subdivision of the autonomic nervous system, and together with its CNS connections it forms the primary control centre for GI function [13]. Beginning in the esophagus and extending down to the anus, the ENS is embedded in the lining of the GI tract and interacts with the CNS through the vagus and pelvic nerves, as well as sympathetic (prevertebral ganglia) connections that regulate contraction, relaxation, secretion, and absorption throughout the GI tract [13].
Gut biopsies from PD patients show evidence of enteric neuronal loss and alteration in the expression of important neurotransmitters, such as vasoactive intestinal peptide (VIP) and dopamine [5,14,15,16,17,18]. Moreover, altered gut barrier function and reduced expression of tight junction (TJ) proteins that mediate paracellular transport (the passage of molecules between adjacent epithelial cells in the gut) have been implicated in PD-related gut dysfunction [19,20]. In addition, the gut microbiome has gained much attention in PD with accumulating evidence linking microbial dysbiosis to PD symptomology and pathophysiology [21]. Thus, PD-induced damage at the level the ENS may be a key contributor to the development of GI symptoms.
The conventional management of PD-induced GI complications, such as dietary interventions and exercise generally do not provide long-term efficacy for PD patients, and approved compounds, such as dopamine antagonists and selective 5-hydroxytryptamine/serotonin 4 (5-HT4) receptor agonists, have demonstrated limited efficacy. Thus, identification of potential targets for novel therapies to alleviate PD-induced GI dysfunctions are essential to improve clinical outcomes and quality of life amongst PD sufferers. This review describes the most common GI symptoms that affect individuals suffering from PD, and the impact they have on quality of life and disease management. Given the limitations of current treatments, we propose a novel approach to the alleviation of GI symptoms by highlighting the intestinal barrier, ENS, and gut microbiome as potential therapeutic avenues, and expand on current emerging therapies and propose several therapeutics that may prove effective through drug repurposing.

2. Gastrointestinal Dysfunction in Parkinson’s Disease

The clinical manifestations of GI dysfunctions in PD include oral and dental disorders, sialorrhea (drooling), dysphagia, gastroparesis, malabsorption, constipation, and defecatory dysfunction [22,23,24,25,26,27,28]. Unlike motor symptoms, which only become apparent on formal diagnosis in the established disease (when degeneration of 50–70% of dopaminergic neurons in the CNS has occurred) [29,30,31,32], GI symptoms have been reported to arise up to 10 years prior to clinical diagnosis. Although the time of onset and severity of GI complications can vary, almost every PD patient suffers from GI symptoms during the natural course of the disease [33,34,35,36,37]. Most PD-related GI symptoms, such as dysphagia and constipation, are a result of abnormal motility of the GI tract. These symptoms not only impact on patient quality of life, but some, particularly dysphagia and gastroparesis, can worsen motor symptoms through the inadequate absorption of oral anti-PD medications. Despite vigorous attempts to understand the pathophysiological underpinnings of these manifestations over the past two decades, GI dysfunction in PD patients is not well understood and lacks effective treatments [38].

2.1. Dysphagia

Dysphagia is primarily caused by one of the cardinal symptoms of PD, bradykinesia, which reduces control of the oral, pharyngeal, and esophageal cavities [26,39,40]; however, several cortical regions also contribute to swallowing difficulties [41,42,43]. The proportion of PD patients experiencing dysphagia ranges from 9–77% [27,44]. This wide range in prevalence is thought to result from the combined effect of patients being unaware that they have symptoms, and the lack of established diagnostic criteria to assess the presence and severity of dysphagia symptoms. Dysphagia is responsible for sialorrhea, one of the most common GI complications reported in PD patients, with up to 75% of patients indicating mild to severe symptoms [39,45]. Although drooling is commonly associated with the excessive production of saliva, a recent study has shown that patients with PD produce less saliva than healthy controls [26]. The accumulation of saliva and the subsequent manifestation of drooling in patients with PD is caused by a combination of disease features, which include dysphagia, a stooped posture, and a tendency towards an open mouth [39].
Whilst there is no correlation between the severity of dysphagia and the clinical progression of PD, there is a clear link between dysphagia and an increased risk of mortality. Dysphagia can cause additional complications such as malnutrition and subsequent weight loss and dehydration, and aspiration pneumonia (estimated to account for 70% of the mortality rates among PD patients) [46,47,48,49].

2.2. Gastroparesis

Gastroparesis is characterized by feeling full after only a small amount of food (early satiety), and the sensation of having food in the stomach (postprandial fullness), which can lead to weight loss, malnutrition, abdominal pain, nausea, vomiting, and bloating [50,51]. Based on scintigraphy studies, approximately 70–90% of PD patients suffer from gastroparesis [50,52]; however, this high prevalence most likely includes asymptomatic manifestation, as patient reporting indicates that observed features of gastroparesis are present in 25–45% of patients [53]. Gastroparesis can be present in both early and late stages of the disease, and unlike dysphagia, the severity of gastroparesis correlates with the severity of motor impairment [52].
The pathophysiological abnormalities of gastroparesis in PD include central and enteric components, with the dorsal motor nucleus of the vagus involved early in the course of disease [54]. The vagus nerve interacts with the ENS to cause neuromuscular dysfunction, which can result in antral hypomotility and/or dysfunction of the pyloric sphincter [55]. These changes in normal function result in delayed stomach emptying that can significantly impact on the control of motor symptoms by delaying the absorption of drugs, such as levodopa, in the small intestines [22,56]. Compounding this dilemma, levodopa treatment can lead to gastroparesis [50], making it difficult to directly attribute gastroparesis to PD. Nevertheless, it is obvious that treatment for this GI symptom is needed to both improve quality of life and control PD symptoms overall.

2.3. Constipation

Constipation, caused by delayed colon transit or the irregular contraction of the voluntary sphincters during defecation [57], is another common GI symptom in PD patients [27,58]. The estimated prevalence of constipation in PD ranges from 7% to 70%, depending on the specific definition [59,60]. According to the most frequently used definition for constipation (<three bowel movements per week) [61], the median prevalence is estimated to be 50% [62]. Similar to other PD-related GI symptoms, the prevalence of constipation within the PD community is difficult to determine due to the varying diagnostic criteria and methods used to assess the symptom. Moreover, drugs used to control motor symptoms, particularly anticholinergics, can both cause and exacerbate constipation [63].
In addition to the vagal connection between the CNS and ENS, the presence of synucleinopathy in the submucosal plexus and myenteric plexus of the colon may also directly affect the action of intestinal muscles that function to move material through the intestines [64]. Constipation is thought to occur early in the course of disease, and it has been reported that it could serve as a predictor, with individuals suffering from long-term constipation having a 2–5 times higher risk of suffering from PD later in life [65,66].

3. Current Treatments for Gastrointestinal Dysfunction in Parkinson’s Disease

Given the impact GI complications have on patient quality of life and drug efficacy, it is important to control symptom severity to enable clinical management of the disease overall. Currently, many treatments involve dietary interventions, physical therapies, and in some cases, drug interventions that target muscle relaxation and contraction of the GI tract (Figure 1). These treatment options have been somewhat successful in alleviating the symptoms, but many have unwanted side effects or offer only short-term relief.

3.1. Dysphagia

Initial treatments for dysphagia primarily revolve around managing food intake, such as requesting patients to chew food more thoroughly, eating smaller portions, and trying foods with softer textures to make it easier to swallow. However, these interventions may not be suitable for all patients, particularly those with more severe symptoms. Swallowing therapy as a rehabilitative treatment has shown promising results [24,67], with a randomised and blinded control study demonstrating that expiratory muscle strength training improved swallowing safety and hyolaryngeal function during swallowing [68]. In addition, video-assisted swallowing therapy, which allows patients to visualize their swallowing patterns, has proven to be effective [69]. This positive outcome is mainly due to PD patients responding better to visual information, and not having to rely on proprioceptive signals and remembered visual information [69,70].
Surgical treatments such as cricopharyngeal sphincterotomy have been reported to yield excellent results [71,72]. However, most studies have involved small samples sizes, and surgical intervention may not be appropriate for all forms of dysphagia (oropharyngeal dysphagia vs. oesophageal dysphagia). Neuromuscular electrical stimulation (NMES) has also been trialled and compared to traditional therapies for dysphagia (logopedic treatment by a speech therapist) in a randomized control trial where NMES was used to facilitate contraction of the suprahyoid muscles. Interestingly, both the traditional method and NMES significantly improved patient quality of life scores; however, there was no significant difference between the two [73].
The outcomes of pharmacological interventions with respect to dysphagia vary substantially between studies and patients. In general, dopaminergic drugs are helpful for dysphagia in PD [47,74,75]; however, some studies showed no improvement [76,77], while others showed equivocal results [78,79,80]. Dysphagia in patients with early stages of PD can be ameliorated by implementing dopaminergic treatment during the first few years after disease onset [22]. However, dysphagia is usually underdiagnosed in these patients due to poor self-awareness of the symptoms [81,82]. Therefore, early detection and individualised intervention are crucial for improving quality of life in patients.

3.2. Gastroparesis

In general, the therapeutic options for gastroparesis are limited and there is currently no gold standard for the treatment of gastroparesis in PD. Management of gastroparesis in PD predominantly involves changes in diet, antiemetics, prokinetics, and in severe cases, feeding tubes or parenteral nutrition [83,84]. Pharmacological treatment of gastroparesis is limited; however, several clinical trials focusing specifically on the management of PD-related gastroparesis have recently been conducted using 5-HT4 receptor agonists, erythromycin, and motolin receptor agonists, which indicate the importance of managing this symptom.
Dietary interventions can help reduce symptoms and improve nutritional deficiencies, and are used as the first line of management for gastroparesis with different aetiologies [85]. These interventions include eating small frequent meals consisting of a low-fibre and low-fat diet of, predominantly, liquids and protein supplements. In such cases, the identification and correction of nutritional deficiencies needs to be considered, such as zinc, iron, selenium, folate, B12, vitamin C and vitamins A, D, K and E. For more severe gastroparesis, a liquid diet, a jejunal feeding tube, gastric electrical stimulation, total parenteral nutrition, or other approaches, may be necessary [85].
With regards to pharmaceutical intervention, to date, only domperidone has proven to be both effective and safe in the treatment of gastroparesis [86]. Domperidone is a peripheral dopamine receptor 2 antagonist, that functions by blocking the inhibitory effect of dopamine, resulting in the stimulation of muscle contraction. In patients with gastroparesis, domperidone increases oesophageal peristalsis and facilitates gastric emptying by augmenting gastric peristalsis and improving antroduodenal co-ordination [87,88].
As well as its prokinetic properties, domperidone has the added benefit of being a potent antiemetic and may increase the bioavailability of levodopa [89,90]. Furthermore, domperidone does not readily cross the blood–brain barrier, and thus the risk of developing extra-pyramidal side-effects is minimal. However, domperidone may prolong the time taken for ventricular depolarisation and repolarisation (QT interval) in predisposed patients, and has been associated with arrhythmia, cardiac arrest, and sudden death at high intravenous doses. Recent case-control studies have also indicated concerns regarding cardiotoxicity following oral administration, particularly in patients over 60 years of age [91,92].
Whilst domperidone remains the most studied prokinetic in patients with PD, other dopaminergic blockers, such as metoclopramide, have been trialled clinically. Metoclopramide is the only Food and Drug Administration (FDA)-approved medication for gastroparesis [93] and is commonly used to prevent levodopa-induced nausea and vomiting. However, metaclopromide can readily cross the blood–brain barrier and has central dopaminergic activity [94,95]. As a result, metoclopramide can precipitate unwanted side effects of excess dopamine receptor stimulation, and its use is not recommended.
Several therapeutic interventions have shown promise for the treatment of gastropareisis, such as the selective 5-HT4 receptor agonists cisapride, tegaserod, and mosapride. Although these drugs were found to alleviate PD-induced gastroparesis, tegaserod and cisapride were found to have toxic cardiovascular effects [96,97], while mosapride can induce abdominal pain, nausea, diarrhea and sometimes constipation.

3.3. Constipation

Prolonged colon content transit time is the physiological basis of symptomatic and asymptomatic constipation [98]. Dysmotility may be related to poor fibre and fluid intake or reduced physical activity [66]. Management of constipation commonly involves interventions that are used to treat constipation in the general population, such as dietary changes, an increase in physical activity, and the use of laxatives and stool softeners.
Lifestyle modifications, especially dietary interventions, are predominantly used as a first-line treatment for constipation. Interventions include increasing dietary fibre through the use of fibre supplements, which have been found to increase stool frequency and output compared with placebo in the general population [98]. Fibre plays an important role in the management of constipation and adequate consumption, at least 30 to 35 g daily, together with appropriate fluid intake (at least 1500 mL daily), is often encouraged [65,99]. Given that stool water content is a crucial factor for stool softening, and consequently, for stool consistency, water-soluble fibres such as psyllium or β-glucan are also recommended. These categories of fibre absorb water to become a gelatinous, viscous substance, which are fermented by bacteria in the digestive tract, promoting peristalsis and increasing stool bulk, softness and intraluminal pressure [28]. Although there is conflicting evidence for the effectiveness of soluble fibres and insoluble fibres for constipation, psyllium intake has shown promise in PD patients where 8 weeks of supplementation increased stool frequency and weight [98]. In addition, psyllium was found to improve the absorption of levodopa, thus providing more stable and higher concentrations of circulating drug [100]. However, despite advice from the World Gastroenterology Organisation to include more fibre in the diet, there is contradictory evidence regarding the use of fibre as a dietary treatment for different types of constipation.
Physical therapies, such as abdominal massage, are known to relieve constipation by stimulating peristalsis, decreasing colonic transit time, and increasing the frequency of bowel movements [101,102,103]. Given that physical interventions are free of harmful side effects, they are often recommended as an adjunct intervention to PD patients [104]. However, qualitative research by McClurg et al. (2016) testing recruitment, retention, and the intervention’s appropriateness failed to demonstrate significant clinical improvement in stool frequency compared to lifestyle advice. Moreover, the Movement Disorder Society concluded that there was “insufficient evidence” for the beneficial effects of abdominal massage on relieving constipation and that the intervention was “investigational” [105]. Although the use of physical therapies such as abdominal massage in PD-associated constipation is contentious, the intervention is generally well tolerated, and side effects are scarce. Thus, there is room for further research.
Polyethylene glycol (PEG) and lubiprostone are first-line pharmacological interventions recommended by evidence-based medicine guidelines for the treatment of constipation due to slow colonic transit in PD [106]. PEG, also known as macrogol, is an osmotic laxative that passes through the GI tract without being absorbed and relieves constipation by drawing water in the bowel. As a result of excess water in the lumen, water content and volume of the stools in the bowel are increased, making them softer and easier to pass. Bowel movements, stool consistency and ease of defecation have been found to be significantly improved in PD patients administered twice daily with isosmotic macrogol electrolyte solution (PEG 7.3 g sachets dissolved in 250 mL of water) both in a double-blind randomized control trial [107] and in an open study [108]. Moreover, a Cochrane review found a statistically significant improvement in the number of bowel motions or successful bowel care routines per week following the use of PEG [109]. Unfortunately, abdominal bloating, vomiting, and cramps have been reported as the most common adverse effects of PEG in other investigations [110,111,112].
Lubiprostone is an intestinal chloride secretagogue that functions by selectively activating type 2 chloride channels in the apical membrane of the GI epithelium [113]. It was approved by the FDA in 2006 for the treatment of chronic idiopathic constipation in adults, and in 2008 for irritable bowel syndrome (IBS) with constipation in adult women [106]. A single placebo-controlled trial of lubiprostone for constipation associated with PD found a marked or very marked clinical global improvement in 16 of 25 (64.0%) patients receiving lubiprostrone vs. 5 of 27 (18.5%) patients receiving placebo (p < 0.001) alongside significant improvements in constipation rating and stools per day. Adverse events were mild, with the most common being intermittent loose stools [114]. Lubiprostone has also been found to have a prostaglandin-like action in the intestine, such that it stimulates mucin release, and has been shown to protect and repair the epithelial barrier and cell function in animal models of stress and disease [115,116].
Patients with PD commonly suffer from GI dysfunctions such as dysphagia, gastroparesis, and constipation. Many of these symptoms appear decades before the onset of motor symptoms. Current treatments often include physical therapies and dietary interventions. Adapted from “Adult female (anterior, lateral head, open mouth”, by BioRender.com (accessed on 17 January 2022) (2021). Retrieved from https://app.biorender.com/biorender-templates (accessed on 17 January 2022).

4. Potential Treatments for Gastrointestinal Dysfunction in Parkinson’s Disease

To date, there are no clinically effective neuroprotective or disease-modifying treatments that can halt PD progression. The current clinical approach for both motor and non-motor symptoms, including GI dysfunctions, focus on symptomatic management and do not aim to prevent or reverse disease progression. Given that GI dysfunction affects drug pharmacodynamics, potentially worsening motor fluctuations and further disability, it is of critical importance that therapeutics/interventions that improve GI health and function are identified. Current treatments for dysphagia, gastroparesis, and constipation result in additional side-effects, and in most cases are not recommended for long-term use, which is difficult when many PD patients require daily intervention [117]. In addition, most interventions have not been validated in the specific context of PD-induced GI dysfunction.

5. Targeting Enteric Neurons

Since the introduction of Braaks staging hypothesis, which states that LBs can initiate their spread from the ENS to unmyelinated ganglionic fibres of the dorsal motor nucleus of the vagus (DMV), multiple studies have shown that alpha-synuclein has prion-like properties that allow it to spread from the ENS to the CNS [118,119,120]. The idea that the GI tract may be involved in PD pathology and progression has sparked interest in whether protecting enteric neurons could prevent GI symptoms, as well as impeding the spread of the disease to the CNS [8].
Deficits at the level of the ENS have been reported in PD patients. Enteric neuronal loss has been found in the ascending and descending colon of patients with PD [17], and downregulation of VIP in colonic submucosal neurons has been reported in PD patients with chronic constipation [121]. Although the extent of neuronal damage in the GI tract in patients with PD varies between studies [122], alpha-synuclein inclusions have been found in dopaminergic enteric neurons [123] and are more common in PD patients than healthy controls [5,17,123,124,125]. However, the contribution of this finding to GI symptomology remains unclear.
Enteric neuropathy and changes in enteric neuron subpopulations have been linked to gut dysfunction in other pathological conditions, including inflammatory bowel disease (IBD), chronic intestinal pseudo-obstruction, and Hirschsprung’s disease [126,127,128]. Thus, it is conceivable that targeting the ENS could be beneficial in impeding gut symptoms and potentially the pathological progression of PD in the CNS.

5.1. 5-Hydroxytryptamine 4 (5-HT4) Receptor Agonists

5-HT is predominantly found in enteroendocrine cells located on the mucosal surface of the gut [129] where it is involved in a number of physiological roles including motility, secretion, and mucosal growth and maintenance [130]. Of the various 5-HT receptors, the 5-HT4 receptor plays a prominent role in releasing several neurotransmitters, most importantly, acetylcholine, the major excitatory neurotransmitter in the GI tract, to modulate propulsive patterns of gut motility [129,131,132].
Prucalopride is a highly selective 5-HT4 receptor agonist that is approved for the treatment of chronic constipation, primarily in elderly patients [133]. Prucalopride, as well as other 5-HT4 agonists such as velusetrag and naronapride, have been proven to be effective in various conditions associated with constipation with impressive safety and tolerance profiles [134,135]. In contrast to studies in healthy human subjects where the prokinetic effect of prucalopride was localized to the colon [136], in patients with constipation, prucalopride also aided with gastric emptying [137,138].
Prucalopride has additional properties that make it a novel therapeutic candidate for the treatment of GI dysfunctions that involve neuronal injury/loss. Prucalopride has been shown to protect primary human enteric neurospheres from oxidative stress injury in culture and protect both excitatory and inhibitory neuron populations [139] that are important for the contraction and relaxation of the GI tract (Figure 2, panel A). Although prucalopride has not been extensively trialled in PD patients for constipation, the small studies that have been performed are promising [140,141]. In addition, the neuroprotective role of prucalopride widens its role, and other 5-HT4 receptor agonists, as a potential therapeutic for GI dysfunctions that involve neuron damage/loss, such as that seen in PD.

5.2. Flavonoids

Flavanoids are group of plant metabolites that are found in a variety of fruits and vegetables. Flavanoids modulate several signalling pathways and are known to have antioxidative, anti-inflammatory, anti-mutagenic, and anti-carcinogenic properties. Depending on their structure, flavanoids can be subdivided into subgroups, which include flavanols, flavanones, flavones, flavonols, isoflavonoids, and anthocyanidins [142].
Flavanols are known for their antioxidant properties and occur abundantly in fruits and vegetables, tea, and red wine. The most well studied flavonols are kaempferol, quercetin, myricetin and fisetin. Quercetin (3,5,7,3′,4′-pentahydroxyflavone) is one of the most potent antioxidants among the flavanoids and has been extensively studied as a potential therapeutic for neurodegenerative diseases, cardiovascular disease, and cancer. Of importance for this review, quercetin has been shown to enhance gut barrier function by increasing the expression of tight junction proteins [143] and has demonstrated neuroprotectant properties [144,145,146], which can subsequently improve motor symptoms in animal models of PD [144,147].
The beneficial effects of quercetin in PD were first identified in the 6-hydroxydopamine rat model of PD, where intraperitoneal injection of quercetin two days after lesioning for 14 days was found to protect dopaminergic neurons in the CNS and increase dopamine levels in the striatum [148]. Quercetin inhibits neuronal cell death by downregulating the pro-inflammatory cytokines NF-κB and iNOS. In addition, it can reduce alpha-synuclein fibrillization in vitro [149], and mitochondrial dysfunction (Figure 2, panel B). Thus, coupled with its role in promoting gut barrier function/integrity, quercetin may have the potential to protect enteric neurons from damage, as observed in the CNS.
Although quercetin has many beneficial effects and shows promise in vitro and in in vivo models of disease, clinical trials/studies of quercetin in PD are yet to be carried out. A possible reason for this is the limited bioavailability of quercetin when orally administered, as it is extensively metabolized in the gut [150]. In addition, the long half-life of quercetin metabolites can lead to plasma accumulation [151]. However, studies have shown that when administered with piperine, an alkaloid found in black pepper, bioavailability is enhanced by inhibiting degradation enzymes [150,152].

6. Targeting the Intestinal Barrier

The GI tract is the largest mucosal surface in the body, and directly interfaces with external environmental factors. The epithelial cells that make up the epithelial lining of the intestine are held together by TJ proteins that allow the passage of ions, water, and solutes through the paracellular pathway and the movement of proteins and lipids between the apical and basolateral surfaces of the plasma membrane. This selectively permeable barrier plays an important role in protecting the body from potentially harmful substances in the lumen of the gut whilst continuing to absorb nutrients.
In addition to TJ proteins, the intestinal barrier also consists of a mucous layer, which is composed primarily of the gel-forming mucin MUC2 [153,154]. MUC2 is synthesized and secreted by epithelial goblet cells to protect the body from microbial antigens and lumen contents [155]. Reduced thickness/disruption of the mucus layer results in an increase in intestinal permeability that can lead to systemic inflammatory responses, enteric neuropathy, and potential GI dysfunction. Therefore, maintaining mucin production in the gut can improve gut health.
Increased GI barrier permeability, which results in toxins and bacteria leaking through the mucous layer and intestinal wall to the bloodstream, is associated with GI dysfunctions and the pathology of several GI inflammatory conditions, including IBS [156] and IBD [157]. Preliminary results indicate that intestinal barrier permeability is altered in PD patients [19,20,156,158]; however, the extent and precise location of this altered barrier integrity (small intestine vs. colon) remains controversial. Nevertheless, a correlation between gut permeability and the presence of alpha-synuclein aggregates was proposed in a mouse model of PD [159], and verified in human PD samples [19], suggesting that therapeutics that target the intestinal barrier could potentially stop the prion-like spread of disease.

6.1. Teduglutide

Glucagon-like peptide-2 (GLP-2) is a pleiotropic hormone that stimulates mucosal growth and repair, and regulates gastric motility, gastric acid secretion, and intestinal hexose transport. GLP-2 significantly enhances the surface area of the mucosal epithelium, reduces intestinal leakiness, and delays gastric transit, thereby increasing intestinal capacity for nutrient (and potentially drug) absorption [160]. Teduglutide (Revestive) is a GLP-2 analogue that binds to GLP-2 receptors located on intestinal enteroendocrine cells and enteric neurons. Teduglutide is currently approved for use in the USA, Europe, and Australia for the treatment of short bowel syndrome. Treatment with teduglutide improves monosaccharide absorption and intestinal fluid balance. Previous studies have shown that GLP-2 receptor activation by teduglutide restores intestinal structure and functional integrity by promoting growth of the mucosa, and protects neuronal nitric oxide synthase-positive neurons, a subset of neurons important for normal gut motility [161]. Moreover, GLP-2 administration enhances the survival of enteric neurons in culture and counteracts mast cell-induced neuronal cell death (Figure 2, panel C). Thus, manipulation of GLP-2 offers a unique dual efficacy, acting as both a mucosal regenerative agent to promote nutrient absorption and as a neuroprotective agent.
Several pre-clinical studies have highlighted the efficacy of GLP-2 in mouse and cell culture models of PD [162,163]. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) lesioned mice, GLP-2 analogue administration improved the bradykinesia and movement imbalance of mice, protected dopaminergic neurons and restored tyrosine hydroxylase expression in the substantia nigra and reduced markers of inflammation and mitochondrial dysfunction [162]. Similarly, in Neuro-2a cells treated with 1-Methyl-4-phenyl-pyridine ion, GLP-2 agonist administration protected cells against mitochondrial damage, autophagy impairments and apoptosis, whilst enhancing cell signalling for mitogenesis, and reducing oxidative stress levels [163]. Whilst the impact of GLP-2 administration on GI health and function is yet to be assessed in PD, these promising findings strongly suggest a beneficial role for GLP-2 in PD.

6.2. Metformin

Anti-diabetic drugs, such as glitazones and exenatide, are known to have pleiotropic actions and have been shown to reduce the risk of developing PD in diabetic patients and reduce neurodegeneration in animal models of disease [164,165,166]. Of note, the GLP-1 receptor agonist exenatide was also found to reduce cognitive decline associated with PD [167]. Further studies are currently being undertaken to explore the therapeutic potential of exenatide in PD and other neurodegenerative conditions.
Unlike exenatide, metformin can be orally administered, which makes it a more preferential treatment than other anti-diabetic interventions. Metformin is a biguanide hyperglyceamic agent that has recently been shown to cross the blood–brain barrier [168] and protect against stroke injury and cognitive decline in diabetic patients [169,170,171]. In addition, a large population-based study showed that people on long-term metformin therapy had a reduced incidence of developing PD [172]. Although the mechanism of metformin is not clear, the drug can reduce alpha-synuclein phosphorylation, aggregation, inflammation and improve autophagy [173].
Given the apparent neuroprotective effect of metformin in the CNS, it is tempting to assume that metformin may protect GI dysfunction-induced enteric neuropathy. However, metformin may also protect the gut by protecting barrier integrity, with a recent study indicating that metformin modulates the gut microbiome and promotes mucus production in ageing high-fat diet mice where it was found to increase MUC2 production and increase TJ protein expression [174]. An increase in the expression levels of the TJ proteins occludin and zonula occludins-1 expression was also observed in a mouse model of colitis following treatment with metformin [175], further confirming its protective action on the gut barrier (Figure 2, panel D).
At high doses, metformin can induce GI side-effects, such as diarrhoea and abdominal pain, which may limit its clinical application. Diarrhoea is the most common GI disturbance and is most likely caused by an increase in colonic bile salt concentrations [176], which results in abnormally high levels of water and salts entering the colon from the bloodstream. These GI side-effects are often manageable or only persist for the first few weeks after dosing; however, in 5% of cases, withdrawal is necessary [177,178]. Nevertheless, metformin’s dual action and extensive safety profile make it a promising therapeutic for improving gut health and function at the tissue and cellular level.

7. Targeting the Gut Microbiota

Emerging evidence suggests that dysbiosis in the gut and subsequent alterations in the gut–brain axis may play a role in the development of motor and non-motor symptoms [179,180], including GI dysfunction. Various studies have demonstrated alterations in both α and β diversity of faecal and mucosal microbial samples from PD patients relative to controls. Whilst results are relatively heterogeneous across studies, several trends have been identified at the family and genus level, with family Verrucomicrobiaceae and genera Akkermansia and Lactobacillus increased in PD patients [181,182,183,184], while the families Prevotellaceae, Lachnospiraceae, and Pasteurellaceae and genera Blautia, Roseburia, Prevotella, and Faecalibacterium appear to be consistently decreased [181,184,185]. Further to this, a recent meta-analysis that re-analysed ten currently available 16S microbiome datasets to investigate whether there were common themes in the alterations to the gut microbiota of PD patients uncovered significant alterations in the PD-associated microbiome [21]. Enrichment of the genera Lactobacillus, Akkermansia, and Bifidobacterium and depletion of bacteria belonging to the Lachnospiraceae family and the Faecalibacterium genus, both important short-chain fatty acids producers, emerged as the most consistent PD gut microbiome alterations [21]. However, it should be noted that the microbial dysbiosis observed in PD could be a consequence of PD-related gut dysfunction (e.g., slowed gastric emptying, delayed colonic transit time) and that alleviation of these GI symptoms may subsequently result in normalization of gut flora. Whilst it is universally acknowledged that data are influenced by PD duration and severity, and several confounders need to be taken into account, these findings have seen the gut microbiota emerge as a therapeutic target in recent years.

Prebiotics, Probiotics, and Fecal Transplants

Several key studies have investigated the impact of prebiotic and probiotic treatment on PD-related constipation [186,187,188]. A recent double-blind, randomized, placebo-controlled, single-centre trial investigating the effects of daily multi-strain probiotics treatment in PD patients with confirmed Rome IV criteria for functional constipation found a significant difference in primary outcomes of probiotic-treated patients compared to placebo [188]. The trial found that PD patients receiving one multi-strain probiotic capsule containing eight different commercially available bacterial strains (Lactobacillus acidophilus, Lactobacillus gasseri, Lactobacillus reuteri, Lactobacillus rhamnosus, Bifidobacterium bifidum, Bifidobacterium longum, Enterococcus faecalis, Enterococcus faecium) daily for 4 weeks had significantly increased spontaneous bowel movements (1.0 ± 1.2 per week) after treatment with probiotics, compared with decreased movements in the placebo group (0.3 ± 1.0 per week, mean difference 1.3, 95% confidence interval 0.8–1.8, p < 0.001) [188]. Significant improvements were also seen in the secondary outcomes of changes in stool consistency, constipation severity score, and quality of life related to constipation after treatment with probiotics [188].
Earlier randomized, double-blind, placebo-controlled trials in PD patients with Rome III-confirmed constipation investigated the effect of fermented milk containing multiple probiotic strains and prebiotic fibre—compared with a placebo, once daily for 4–5 weeks—and uncovered that probiotic treatment resulted in a higher number of complete bowel movements (mean difference 1.1, 95% confidence interval 0.4–1.8, p < 0.002) [186], a significant increase in the number of days per week in which stools were of normal consistency, and significant reductions in the number of days per week in which patients felt bloated, experienced abdominal pain, or the sensation of incomplete emptying [187] (Figure 2, panels E and F).
With regard to intestinal transit time (ITT), a meta-analysis of 11 randomized controlled clinical trials with 13 treatment effects representing 464 subjects determined that short-term probiotic supplementation decreases ITT with consistently greater treatment effects identified in constipated or older adults [189]; however, the effect of probiotics on ITT has not been investigated in PD.
Fecal microbiota transplants (FMT) are also emerging a therapeutic avenue to rectify microbial dysbiosis in PD patients [190,191]. An early case report, describing the effect of FMT on a 71-year-old male patient with 7 years of resting tremor, bradykinesia and intractable constipation concluded that microbial reconstitution may have therapeutic effects in PD after the patient successfully defecated within 5 min of FMT and maintained daily unobstructed defecation until the end of follow-up (3 months). A recent evaluation of FMT in 11 PD patients with constipation found similar results with a significant reduction in the Wexner constipation score following 6 and 12 weeks of FMT therapy [191].
Though some doubts about the use of probiotics, prebiotics, and fecal microbiota reconstitution have been raised [192], the latest Movement Disorder Society evidence-based medicine review update on treatments for the non-motor symptoms of PD determine probiotics and prebiotic fibre as “efficacious” and “clinically useful”. However, it was highlighted that they could pose an acceptable risk without specialized monitoring [105].
The normal functioning of the gut heavily relies on a healthy gut microbiome, and an intact mucosal and epithelial barrier that inhibit potentially pathogenic toxins/microbes and their metabolites from entering the bloodstream from the lumen of the gut. When microbial dysbiosis occurs and the mucosal barrier is disrupted, pathogenic microbes can enter the blood stream through changes in tight junction protein expression that make the barrier more permeable. These pathogens can cause damage to enteric neurons located in the submucosal and myenteric plexuses, and subsequent GI dysfunction and systemic inflammation. (A) Prucalopride, (B) quercetin, (C) teduglutide, and (D) metformin all have neuroprotective properties that may protect enteric neurons from damage. In addition, these therapeutics have pleiotropic properties that involve enhancing intestinal mucous production or increasing tight junction protein expression. (E and F) Prebiotics, probiotics, and fecal transplants all aim to improve gut health and function by promoting a healthy gut microbiome. Modified from [193] and adapted from “Jejunum Epithelium”, by BioRender.com (accessed on 17 January 2022) (2021). Retrieved from https://app.biorender.com/biorender-templates (accessed on 17 January 2022).

8. Conclusions

The incidence of PD is rising exponentially as life expectancy increases, and the number of individuals suffering from PD is conservatively estimated to increase to 12 million worldwide by 2050 [194]. Despite PD being recognized as a multi-system disorder, and growing awareness of the tremendous impact of non-motor symptoms on quality of life and disease management, the development of therapeutics to treat these debilitating symptoms has been largely overlooked.
Although not all PD patients have identical pathogenic mechanisms underlying their disease, GI dysfunctions are a common occurrence that require further, and better investigation. Many PD-related GI symptoms manifest early on in PD pathogenesis, and accumulating research shows that the GI tract may contribute to the spread of disease. GI function is highly dependent on gut health, which is tightly linked to intestinal gut barrier integrity, a healthy gut microbiome, and the proper functioning of enteric neurons responsible for the contraction and relaxation of the GI tract, all of which are compromised to some degree in PD patients. Therefore, using drugs that can target one, or all, parameters may prove to be beneficial and provide long-term relief for the GI symptoms that plague PD patients.
In this review, we have highlighted several potential therapeutics that either directly or indirectly target the GI tract at the tissue or cellular level. Although limited data is available on their effectiveness in PD-related GI dysfunctions, their efficacy in the CNS or other GI disorders warrants further investigation. In addition, the potential of repurposing drugs that have shown promise in the CNS or have pleiotropic properties may provide a faster identification of, and more cost-effective, therapeutics. Given the GI tract is integral for drug pharmacodynamics, improving gut health and potential drug metabolism is not only relevant for PD patients, but also other diseases/disorders where the GI system is affected and where oral administration of medications are used to treat disease. Thus, improving gut health has broader applications, and identifying drugs that can target one, or all parameters of gut heath and function will be highly beneficial to society overall.

Author Contributions

Conceptualization, M.N.H., R.M.M., S.D.; writing—original draft preparation, M.N.H., R.M.M., S.D.; writing—review and editing, R.M.M., S.D., D.I.F.; funding acquisition, R.M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Jack Brockhoff Early Career Research Grant (ID: 4506) and NHMRC Ideas Grant (APP1183420).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors acknowledge the use of software from BioRender.com for the preparation of all figures.

Conflicts of Interest

The authors declare that they have no competing interest.

Abbreviations

5-HT45-hydroxytryptamine/serotonin 4
CNSCentral nervous system
DMVDorsal motor nucleus of the vagus
ENSEnteric nervous system
FMTFecal microbiota transplant
FDAFood and Drug Administration
GIGastrointestinal
GLP-2Glucagon-like peptide-2
IBDInflammatory bowel disease
IBSInflammatory bowel syndrome
ITTIntestinal transit time
LBLewy bodies
MPTP1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
NMESNeuromuscular electrical stimulation
PDParkinson’s disease
PEGPolyethylene glycol
SNpcSubstantia nigra pars compacta
TJTight junction
VIPVasoactive intestinal peptide

References

  1. Nussbaum, R.L.; Ellis, C.E. Alzheimer’s disease and Parkinson’s disease. N. Engl. J. Med. 2003, 348, 1356–1364. [Google Scholar] [CrossRef] [Green Version]
  2. Fahn, S. Description of Parkinson’s disease as a clinical syndrome. Ann. N. Y. Acad. Sci. 2003, 991, 1–14. [Google Scholar] [CrossRef]
  3. Lees, A.J.; Hardy, J.; Revesz, T. Parkinson’s disease. Lancet 2009, 373, 2055–2066. [Google Scholar] [CrossRef]
  4. Shahed, J.; Jankovic, J. Motor symptoms in Parkinson’s disease. Handb. Clin. Neurol. 2007, 83, 329–342. [Google Scholar] [CrossRef]
  5. Wakabayashi, K.; Takahashi, H.; Takeda, S.; Ohama, E.; Ikuta, F. Parkinson’s disease: The presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol. 1988, 76, 217–221. [Google Scholar] [CrossRef] [PubMed]
  6. Del Tredici, K.; Duda, J.E. Peripheral Lewy body pathology in Parkinson’s disease and incidental Lewy body disease: Four cases. J. Neurol. Sci. 2011, 310, 100–106. [Google Scholar] [CrossRef] [PubMed]
  7. Chen, Z.; Li, G.; Liu, J. Autonomic dysfunction in Parkinson’s disease: Implications for pathophysiology, diagnosis, and treatment. Neurobiol. Dis. 2020, 134, 104700. [Google Scholar] [CrossRef]
  8. 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]
  9. Lorefalt, B.; Ganowiak, W.; Palhagen, S.; Toss, G.; Unosson, M.; Granerus, A.K. Factors of importance for weight loss in elderly patients with Parkinson’s disease. Acta Neurol. Scand. 2004, 110, 180–187. [Google Scholar] [CrossRef]
  10. Park, K.; Oeda, T.; Kohsaka, M.; Tomita, S.; Umemura, A.; Sawada, H. Low body mass index and life prognosis in Parkinson’s disease. Parkinsonism Relat. Disord. 2018, 55, 81–85. [Google Scholar] [CrossRef] [PubMed]
  11. Goldman, J.G.; Postuma, R. Premotor and nonmotor features of Parkinson’s disease. Curr. Opin. Neurol. 2014, 27, 434–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chaudhuri, K.R.; Healy, D.G.; Schapira, A.H.; National Institute for Clinical Excellence. Non-motor symptoms of Parkinson’s disease: Diagnosis and management. Lancet Neurol. 2006, 5, 235–245. [Google Scholar] [CrossRef]
  13. Furness, J.B.; Callaghan, B.P.; Rivera, L.R.; Cho, H.J. The enteric nervous system and gastrointestinal innervation: Integrated local and central control. Adv. Exp. Med. Biol. 2014, 817, 39–71. [Google Scholar] [CrossRef]
  14. Annerino, D.M.; Arshad, S.; Taylor, G.M.; Adler, C.H.; Beach, T.G.; Greene, J.G. Parkinson’s disease is not associated with gastrointestinal myenteric ganglion neuron loss. Acta Neuropathol. 2012, 124, 665–680. [Google Scholar] [CrossRef] [Green Version]
  15. Anderson, G.; Noorian, A.R.; Taylor, G.; Anitha, M.; Bernhard, D.; Srinivasan, S.; Greene, J.G. Loss of enteric dopaminergic neurons and associated changes in colon motility in an MPTP mouse model of Parkinson’s disease. Exp. Neurol. 2007, 207, 4–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Chaumette, T.; Lebouvier, T.; Aubert, P.; Lardeux, B.; Qin, C.; Li, Q.; Accary, D.; Bezard, E.; Bruley des Varannes, S.; Derkinderen, P.; et al. Neurochemical plasticity in the enteric nervous system of a primate animal model of experimental Parkinsonism. Neurogastroenterol. Motil. 2009, 21, 215–222. [Google Scholar] [CrossRef]
  17. Lebouvier, T.; Neunlist, M.; Bruley des Varannes, S.; Coron, E.; Drouard, A.; N’Guyen, J.M.; Chaumette, T.; Tasselli, M.; Paillusson, S.; Flamand, M.; et al. Colonic biopsies to assess the neuropathology of Parkinson’s disease and its relationship with symptoms. PLoS ONE 2010, 5, e12728. [Google Scholar] [CrossRef] [Green Version]
  18. Zhu, H.C.; Zhao, J.; Luo, C.Y.; Li, Q.Q. Gastrointestinal dysfunction in a Parkinson’s disease rat model and the changes of dopaminergic, nitric oxidergic, and cholinergic neurotransmitters in myenteric plexus. J. Mol. Neurosci. 2012, 47, 15–25. [Google Scholar] [CrossRef] [Green Version]
  19. 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] [Green Version]
  20. Clairembault, T.; Leclair-Visonneau, L.; Coron, E.; Bourreille, A.; Le Dily, S.; Vavasseur, F.; Heymann, M.F.; Neunlist, M.; Derkinderen, P. Structural alterations of the intestinal epithelial barrier in Parkinson’s disease. Acta Neuropathol. Commun. 2015, 3, 12. [Google Scholar] [CrossRef] [Green Version]
  21. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. NPJ Parkinsons Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
  22. Muller, T.; Erdmann, C.; Bremen, D.; Schmidt, W.E.; Muhlack, S.; Woitalla, D.; Goetze, O. Impact of gastric emptying on levodopa pharmacokinetics in Parkinson disease patients. Clin. Neuropharmacol. 2006, 29, 61–67. [Google Scholar] [CrossRef] [PubMed]
  23. Clifford, T.; Finnerty, J. The dental awareness and needs of a Parkinson’s disease population. Gerodontology 1995, 12, 99–103. [Google Scholar] [CrossRef] [PubMed]
  24. Coates, C.; Bakheit, A.M. Dysphagia in Parkinson’s disease. Eur. Neurol. 1997, 38, 49–52. [Google Scholar] [CrossRef]
  25. Kaye, J.; Gage, H.; Kimber, A.; Storey, L.; Trend, P. Excess burden of constipation in Parkinson’s disease: A pilot study. J. Mov. Disord. 2006, 21, 1270–1273. [Google Scholar] [CrossRef] [PubMed]
  26. Proulx, M.; de Courval, F.P.; Wiseman, M.A.; Panisset, M. Salivary production in Parkinson’s disease. J. Mov. Disord. 2005, 20, 204–207. [Google Scholar] [CrossRef]
  27. Edwards, L.L.; Quigley, E.M.; Harned, R.K.; Hofman, R.; Pfeiffer, R.F. Characterization of swallowing and defecation in Parkinson’s disease. Am. J. Gastroenterol. 1994, 89, 15–25. [Google Scholar]
  28. Yang, T.; Zhan, Z.; Zhang, L.; Zhu, J.; Liu, Y.; Zhang, L.; Ge, J.; Zhao, Y.; Zhang, L.; Dong, J. Prevalence and Risk Factors for Malnutrition in Patients with Parkinson’s Disease. Front. Neurol. 2020, 11, 533731. [Google Scholar] [CrossRef]
  29. Dauer, W.; Przedborski, S. Parkinson’s disease: Mechanisms and models. Neuron 2003, 39, 889–909. [Google Scholar] [CrossRef] [Green Version]
  30. Lang, A.E.; Lozano, A.M. Parkinson’s disease. First of two parts. N. Engl. J. Med. 1998, 339, 1044–1053. [Google Scholar] [CrossRef]
  31. Marsden, C.D. Parkinson’s disease. Lancet 1990, 335, 948–952. [Google Scholar] [CrossRef]
  32. Ross, G.W.; Petrovitch, H.; Abbott, R.D.; Nelson, J.; Markesbery, W.; Davis, D.; Hardman, J.; Launer, L.; Masaki, K.; Tanner, C.M.; et al. Parkinsonian signs and substantia nigra neuron density in decendents elders without PD. Ann. Neurol. 2004, 56, 532–539. [Google Scholar] [CrossRef] [PubMed]
  33. Barone, P.; Antonini, A.; Colosimo, C.; Marconi, R.; Morgante, L.; Avarello, T.P.; Bottacchi, E.; Cannas, A.; Ceravolo, G.; Ceravolo, R. The PRIAMO study: A multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2009, 24, 1641–1649. [Google Scholar] [CrossRef] [PubMed]
  34. Bernal-Pacheco, O.; Limotai, N.; Go, C.L.; Fernandez, H.H. Nonmotor manifestations in Parkinson disease. Neurologist 2012, 18, 1–16. [Google Scholar] [CrossRef] [PubMed]
  35. Dubow, J.S. Autonomic dysfunction in Parkinson’s disease. Dis. Mon. 2007, 53, 265–274. [Google Scholar] [CrossRef] [PubMed]
  36. Lyons, K.E.; Pahwa, R. The impact and management of nonmotor symptoms of Parkinson’s disease. Am. J. Manag. Care 2011, 17, S308. [Google Scholar] [PubMed]
  37. Knudsen, K.; Fedorova, T.D.; Bekker, A.C.; Iversen, P.; Østergaard, K.; Krogh, K.; Borghammer, P. Objective colonic dysfunction is far more prevalent than subjective constipation in Parkinson’s disease: A colon transit and volume study. J. Parkinson Dis. 2017, 7, 359–367. [Google Scholar] [CrossRef]
  38. Su, A.; Gandhy, R.; Barlow, C.; Triadafilopoulos, G. A practical review of gastrointestinal manifestations in Parkinson’s disease. Parkinsonism Relat. Disord. 2017, 39, 17–26. [Google Scholar] [CrossRef]
  39. Srivanitchapoom, P.; Pandey, S.; Hallett, M. Drooling in Parkinson’s disease: A review. Parkinsonism Relat. Disord. 2014, 20, 1109–1118. [Google Scholar] [CrossRef] [Green Version]
  40. Umemoto, G.; Tsuboi, Y.; Kitashima, A.; Furuya, H.; Kikuta, T. Impaired food transportation in Parkinson’s disease related to lingual bradykinesia. Dysphagia 2011, 26, 250–255. [Google Scholar] [CrossRef]
  41. Kikuchi, A.; Baba, T.; Hasegawa, T.; Kobayashi, M.; Sugeno, N.; Konno, M.; Miura, E.; Hosokai, Y.; Ishioka, T.; Nishio, Y.; et al. Hypometabolism in the supplementary and anterior cingulate cortices is related to dysphagia in Parkinson’s disease: A cross-sectional and 3-year longitudinal cohort study. BMJ Open 2013, 3, e002249. [Google Scholar] [CrossRef]
  42. Lee, K.D.; Koo, J.H.; Song, S.H.; Jo, K.D.; Lee, M.K.; Jang, W. Central cholinergic dysfunction could be associated with oropharyngeal dysphagia in early Parkinson’s disease. J. Neural. Transm. 2015, 122, 1553–1561. [Google Scholar] [CrossRef]
  43. Suntrup, S.; Teismann, I.; Bejer, J.; Suttrup, I.; Winkels, M.; Mehler, D.; Pantev, C.; Dziewas, R.; Warnecke, T. Evidence for adaptive cortical changes in swallowing in Parkinson’s disease. Brain 2013, 136, 726–738. [Google Scholar] [CrossRef]
  44. Picillo, M.; Amboni, M.; Erro, R.; Longo, K.; Vitale, C.; Moccia, M.; Pierro, A.; Santangelo, G.; De Rosa, A.; De Michele, G.; et al. Gender differences in non-motor symptoms in early, drug naive Parkinson’s disease. J. Neurol. 2013, 260, 2849–2855. [Google Scholar] [CrossRef] [PubMed]
  45. Johnston, B.T.; Li, Q.; Castell, J.A.; Castell, D.O. Swallowing and esophageal function in Parkinson’s disease. Am. J. Gastroenterol. 1995, 90, 1741–1746. [Google Scholar] [PubMed]
  46. Martinez-Ramirez, D.; Almeida, L.; Giugni, J.C.; Ahmed, B.; Higuchi, M.A.; Little, C.S.; Chapman, J.P.; Mignacca, C.; Wagle Shukla, A.; Hess, C.W.; et al. Rate of aspiration pneumonia in hospitalized Parkinson’s disease patients: A cross-sectional study. BMC Neurol. 2015, 15, 104. [Google Scholar] [CrossRef] [Green Version]
  47. Fuh, J.L.; Lee, R.C.; Wang, S.J.; Lin, C.H.; Wang, P.N.; Chiang, J.H.; Liu, H.C. Swallowing difficulty in Parkinson’s disease. Clin. Neurol. Neurosurg. 1997, 99, 106–112. [Google Scholar] [CrossRef]
  48. Potulska, A.; Friedman, A.; Krolicki, L.; Spychala, A. Swallowing disorders in Parkinson’s disease. Parkinsonism Relat. Disord. 2003, 9, 349–353. [Google Scholar] [CrossRef]
  49. Lin, C.W.; Chang, Y.C.; Chen, W.S.; Chang, K.; Chang, H.Y.; Wang, T.G. Prolonged swallowing time in dysphagic Parkinsonism patients with aspiration pneumonia. Arch. Phys. Med. Rehabil. 2012, 93, 2080–2084. [Google Scholar] [CrossRef]
  50. Heetun, Z.S.; Quigley, E.M. Gastroparesis and Parkinson’s disease: A systematic review. Parkinsonism Relat. Disord. 2012, 18, 433–440. [Google Scholar] [CrossRef]
  51. Tang, D.M.; Friedenberg, F.K. Gastroparesis: Approach, diagnostic evaluation, and management. Dis. Mon. 2011, 57, 74–101. [Google Scholar] [CrossRef]
  52. Goetze, O.; Nikodem, A.B.; Wiezcorek, J.; Banasch, M.; Przuntek, H.; Mueller, T.; Schmidt, W.E.; Woitalla, D. Predictors of gastric emptying in Parkinson’s disease. Neurogastroenterol. Motil. 2006, 18, 369–375. [Google Scholar] [CrossRef]
  53. Siddiqui, M.F.; Rast, S.; Lynn, M.J.; Auchus, A.P.; Pfeiffer, R.F. Autonomic dysfunction in Parkinson’s disease: A comprehensive symptom survey. Parkinsonism Relat. Disord. 2002, 8, 277–284. [Google Scholar] [CrossRef]
  54. Braak, H.; Rub, 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]
  55. Soliman, H.; Coffin, B.; Gourcerol, G. Gastroparesis in Parkinson Disease: Pathophysiology, and Clinical Management. Brain Sci. 2021, 11, 831. [Google Scholar] [CrossRef] [PubMed]
  56. Doi, H.; Sakakibara, R.; Sato, M.; Masaka, T.; Kishi, M.; Tateno, A.; Tateno, F.; Tsuyusaki, Y.; Takahashi, O. Plasma levodopa peak delay and impaired gastric emptying in Parkinson’s disease. J. Neurol. Sci. 2012, 319, 86–88. [Google Scholar] [CrossRef] [PubMed]
  57. Mathers, S.E.; Kempster, P.A.; Swash, M.; Lees, A.J. Constipation and paradoxical puborectalis contraction in anismus and Parkinson’s disease: A dystonic phenomenon? J. Neurol. Neurosurg. Psychiatry 1988, 51, 1503–1507. [Google Scholar] [CrossRef] [Green Version]
  58. Jost, W.; Schimrigk, K.J.K.W. Constipation in Parkinson’s disease. Klin. Wochenschr. 1991, 69, 906–909. [Google Scholar] [CrossRef] [Green Version]
  59. Singer, C.; Weiner, W.J.; Sanchez-Ramos, J.R. Autonomic dysfunction in men with Parkinson’s disease. Eur. Neurol. 1992, 32, 134–140. [Google Scholar] [CrossRef]
  60. Yu, B.; Xiao, Z.Y.; Li, J.Z.; Yuan, J.; Liu, Y.M. Study of an integrated non-motor symptoms questionnaire for Parkinson’s disease. Chin. Med. J. 2010, 123, 1436–1440. [Google Scholar]
  61. Postuma, R.; Romenets, S.R.; Rakheja, R. Physician Guide: Non-Motor Symptoms of Parkinson’s Disease; McGill University Health Centre: Montreal, QC, Canada, 2012. [Google Scholar]
  62. Knudsen, K.; Krogh, K.; Ostergaard, K.; Borghammer, P. Constipation in parkinson’s disease: Subjective symptoms, objective markers, and new perspectives. Mov. Disord. 2017, 32, 94–105. [Google Scholar] [CrossRef] [PubMed]
  63. Rodríguez-Ramallo, H.; Báez-Gutiérrez, N.; Prado-Mel, E.; Alfaro-Lara, E.R.; Santos-Ramos, B.; Sánchez-Fidalgo, S. Association between Anticholinergic Burden and Constipation: A Systematic Review. Healthcare 2021, 9, 581. [Google Scholar] [CrossRef] [PubMed]
  64. Andrews, C.N.; Storr, M. The pathophysiology of chronic constipation. Can. J. Gastroenterol. 2011, 25 (Suppl. B), 16B–21B. [Google Scholar] [CrossRef]
  65. Abbott, R.D.; Petrovitch, H.; White, L.R.; Masaki, K.H.; Tanner, C.M.; Curb, J.D.; Grandinetti, A.; Blanchette, P.L.; Popper, J.S.; Ross, G.W. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology 2001, 57, 456–462. [Google Scholar] [CrossRef]
  66. Ueki, A.; Otsuka, M. Life style risks of Parkinson’s disease: Association between decreased water intake and constipation. J. Neurol. 2004, 251 (Suppl. 7), vII18–vII23. [Google Scholar] [CrossRef] [PubMed]
  67. El Sharkawi, A.; Ramig, L.; Logemann, J.A.; Pauloski, B.R.; Rademaker, A.W.; Smith, C.H.; Pawlas, A.; Baum, S.; Werner, C. Swallowing and voice effects of Lee Silverman Voice Treatment (LSVT): A pilot study. J. Neurol. Neurosurg. Psychiatry 2002, 72, 31–36. [Google Scholar] [CrossRef]
  68. Troche, M.S.; Huebner, I.; Rosenbek, J.C.; Okun, M.S.; Sapienza, C.M. Respiratory-swallowing coordination and swallowing safety in patients with Parkinson’s disease. Dysphagia 2011, 26, 218–224. [Google Scholar] [CrossRef]
  69. Manor, Y.; Mootanah, R.; Freud, D.; Giladi, N.; Cohen, J.T. Video-assisted swallowing therapy for patients with Parkinson’s disease. Parkinsonism Relat. Disord. 2013, 19, 207–211. [Google Scholar] [CrossRef]
  70. Adamovich, S.V.; Berkinblit, M.B.; Hening, W.; Sage, J.; Poizner, H. The interaction of visual and proprioceptive inputs in pointing to actual and remembered targets in Parkinson’s disease. Neuroscience 2001, 104, 1027–1041. [Google Scholar] [CrossRef]
  71. Born, L.J.; Harned, R.H.; Rikkers, L.F.; Pfeiffer, R.F.; Quigley, E.M. Cricopharyngeal dysfunction in Parkinson’s disease: Role in dysphagia and response to myotomy. Mov. Disord. 1996, 11, 53–58. [Google Scholar] [CrossRef]
  72. Byrne, K.G.; Pfeiffer, R.; Quigley, E.M. Gastrointestinal dysfunction in Parkinson’s disease. A report of clinical experience at a single center. J. Clin. Gastroenterol. 1994, 19, 11–16. [Google Scholar] [CrossRef] [PubMed]
  73. Heijnen, B.J.; Speyer, R.; Baijens, L.W.; Bogaardt, H.C. Neuromuscular electrical stimulation versus traditional therapy in patients with Parkinson’s disease and oropharyngeal dysphagia: Effects on quality of life. Dysphagia 2012, 27, 336–345. [Google Scholar] [CrossRef] [Green Version]
  74. Warnecke, T.; Suttrup, I.; Schroder, J.B.; Osada, N.; Oelenberg, S.; Hamacher, C.; Suntrup, S.; Dziewas, R. Levodopa responsiveness of dysphagia in advanced Parkinson’s disease and reliability testing of the FEES-Levodopa-test. Parkinsonism Relat. Disord. 2016, 28, 100–106. [Google Scholar] [CrossRef]
  75. Tison, F.; Wiart, L.; Guatterie, M.; Fouillet, N.; Lozano, V.; Henry, P.; Barat, M. Effects of central dopaminergic stimulation by apomorphine on swallowing disorders in Parkinson’s disease. Mov. Disord. 1996, 11, 729–732. [Google Scholar] [CrossRef] [PubMed]
  76. Calne, D.; Shaw, D.; Spiers, A.; Stern, G. Swallowing in parkinsonism. Br. J. Radiol. 1970, 43, 456–457. [Google Scholar] [CrossRef] [PubMed]
  77. Lim, A.; Leow, L.; Huckabee, M.-L.; Frampton, C.; Anderson, T. A pilot study of respiration and swallowing integration in Parkinson’s disease: “On” and “Off” levodopa. Dysphagia 2008, 23, 76–81. [Google Scholar] [CrossRef]
  78. Bushmann, M.; Dobmeyer, S.M.; Leeker, L.; Perlmutter, J.S. Swallowing abnormalities and their response to treatment in Parkinson’s disease. Neurology 1989, 39, 1309–1314. [Google Scholar] [CrossRef]
  79. Muller, B.; Assmus, J.; Larsen, J.P.; Haugarvoll, K.; Skeie, G.O.; Tysnes, O.B.; ParkWest Study Group. Autonomic symptoms and dopaminergic treatment in de novo Parkinson’s disease. Acta Neurol. Scand. 2013, 127, 290–294. [Google Scholar] [CrossRef]
  80. Hunter, P.C.; Crameri, J.; Austin, S.; Woodward, M.C.; Hughes, A.J. Response of parkinsonian swallowing dysfunction to dopaminergic stimulation. J. Neurol. Neurosurg. Psychiatry 1997, 63, 579–583. [Google Scholar] [CrossRef]
  81. Miller, N.; Allcock, L.; Hildreth, A.J.; Jones, D.; Noble, E.; Burn, D.J. Swallowing problems in Parkinson disease: Frequency and clinical correlates. J. Neurol. Neurosurg. Psychiatry 2009, 80, 1047–1049. [Google Scholar] [CrossRef]
  82. Noble, E.; Jones, D.; Miller, N. Perceived changes to swallowing in people with Parkinson’s disease. Int. J. Ther. Rehabil. 2015, 22, 573–581. [Google Scholar] [CrossRef]
  83. Camilleri, M.; Parkman, H.P.; Shafi, M.A.; Abell, T.L.; Gerson, L. Clinical guideline: Management of gastroparesis. Am. J. Gastroenterol. 2013, 108, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Navas, C.M.; Patel, N.K.; Lacy, B.E. Symptomatic management of gastroparesis. Gastrointest. Endosc. Clin. 2019, 29, 55–70. [Google Scholar] [CrossRef] [PubMed]
  85. Parkman, H.P.; Yates, K.; Hasler, W.L.; Nguyen, L.; Pasricha, P.J.; Snape, W.J.; Farrugia, G.; Koch, K.L.; Abell, T.L.; McCallum, R.W.; et al. Clinical features of idiopathic gastroparesis vary with sex, body mass, symptom onset, delay in gastric emptying, and gastroparesis severity. Gastroenterology 2011, 140, 101–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Natale, G.; Pasquali, L.; Ruggieri, S.; Paparelli, A.; Fornai, F. Parkinson’s disease and the gut: A well known clinical association in need of an effective cure and explanation. Neurogastroenterol. Motil. 2008, 20, 741–749. [Google Scholar] [CrossRef] [PubMed]
  87. Brogden, R.N.; Carmine, A.A.; Heel, R.C.; Speight, T.M.; Avery, G.S. Domperidone. A review of its pharmacological activity, pharmacokinetics and therapeutic efficacy in the symptomatic treatment of chronic dyspepsia and as an antiemetic. Drugs 1982, 24, 360–400. [Google Scholar] [CrossRef] [PubMed]
  88. Van Nueten, J.M.; Schuurkes, J.A. Studies on the role of dopamine and dopamine blockers in gastroduodenal motility. Scand. J. Gastroenterol. Suppl. 1984, 96, 89–99. [Google Scholar]
  89. Nishikawa, N.; Nagai, M.; Tsujii, T.; Iwaki, H.; Yabe, H.; Nomoto, M. Coadministration of domperidone increases plasma levodopa concentration in patients with Parkinson disease. Clin. Neuropharmacol. 2012, 35, 182–184. [Google Scholar] [CrossRef]
  90. Soykan, I.; Sarosiek, I.; Shifflett, J.; Wooten, G.F.; McCallum, R.W. Effect of chronic oral domperidone therapy on gastrointestinal symptoms and gastric emptying in patients with Parkinson’s disease. Mov. Disord. 1997, 12, 952–957. [Google Scholar] [CrossRef]
  91. Johannes, C.B.; Varas-Lorenzo, C.; McQuay, L.J.; Midkiff, K.D.; Fife, D. Risk of serious ventricular arrhythmia and sudden cardiac death in a cohort of users of domperidone: A nested case-control study. Pharmacoepidemiol. Drug Saf. 2010, 19, 881–888. [Google Scholar] [CrossRef]
  92. van Noord, C.; Dieleman, J.P.; van Herpen, G.; Verhamme, K.; Sturkenboom, M.C. Domperidone and ventricular arrhythmia or sudden cardiac death: A population-based case-control study in the Netherlands. Drug Saf. 2010, 33, 1003–1014. [Google Scholar] [CrossRef]
  93. Ramprasad, C.; Douglas, J.Y.; Moshiree, B. Parkinson’s Disease and Current Treatments for Its Gastrointestinal Neurogastromotility Effects. Curr. Treat. Options Gastroenterol. 2018, 16, 489–510. [Google Scholar] [CrossRef]
  94. Avorn, J.; Gurwitz, J.H.; Bohn, R.L.; Mogun, H.; Monane, M.; Walker, A. Increased incidence of levodopa therapy following metoclopramide use. JAMA 1995, 274, 1780–1782. [Google Scholar] [CrossRef]
  95. Barone, J.A. Domperidone: A peripherally acting dopamine2-receptor antagonist. Ann. Pharmacother. 1999, 33, 429–440. [Google Scholar] [CrossRef]
  96. Hennessy, S.; Leonard, C.E.; Newcomb, C.; Kimmel, S.E.; Bilker, W.B. Cisapride and ventricular arrhythmia. Br. J. Clin. Pharmacol. 2008, 66, 375–385. [Google Scholar] [CrossRef] [Green Version]
  97. Loughlin, J.; Quinn, S.; Rivero, E.; Wong, J.; Huang, J.; Kralstein, J.; Earnest, D.L.; Seeger, J.D. Tegaserod and the risk of cardiovascular ischemic events: An observational cohort study. J. Cardiovasc. Pharmacol. Ther. 2010, 15, 151–157. [Google Scholar] [CrossRef]
  98. Ashraf, W.; Park, F.; Lof, J.; Quigley, E.M. Effects of psyllium therapy on stool characteristics, colon transit and anorectal function in chronic idiopathic constipation. Aliment. Pharmacol. Ther. 1995, 9, 639–647. [Google Scholar] [CrossRef] [PubMed]
  99. Johanson, J.F. Review of the treatment options for chronic constipation. MedGenMed 2007, 9, 25. [Google Scholar] [PubMed]
  100. Fernandez-Martinez, M.N.; Hernandez-Echevarria, L.; Sierra-Vega, M.; Diez-Liebana, M.J.; Calle-Pardo, A.; Carriedo-Ule, D.; Sahagun-Prieto, A.M.; Anguera-Vila, A.; Garcia-Vieitez, J.J. A randomised clinical trial to evaluate the effects of Plantago ovata husk in Parkinson patients: Changes in levodopa pharmacokinetics and biochemical parameters. BMC Complement. Altern. Med. 2014, 14, 296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Ernst, E. Abdominal massage therapy for chronic constipation: A systematic review of controlled clinical trials. Complement. Med. Res. 1999, 6, 149–151. [Google Scholar] [CrossRef]
  102. Sinclair, M. The use of abdominal massage to treat chronic constipation. J. Bodyw. Mov. Ther. 2011, 15, 436–445. [Google Scholar] [CrossRef] [PubMed]
  103. McClurg, D.; Hagen, S.; Dickinson, L. Abdominal massage for the treatment of constipation. Cochrane Database Syst. Rev. 2011. [Google Scholar] [CrossRef]
  104. Mokhtare, M.; Karimi, S.; Bahardoust, M.; Sotoudeheian, M.; Ghazi, A.; Babaei-Ghazani, A. How adding the abdominal massage to polyethylene glycol can improve symptom and quality of life in patients with functional constipation in comparison with each one of the treatment modalities alone: A randomized clinical trial. Complement. Ther. Med. 2020, 52, 102495. [Google Scholar] [CrossRef] [PubMed]
  105. Seppi, K.; Ray Chaudhuri, K.; Coelho, M.; Fox, S.H.; Katzenschlager, R.; Perez Lloret, S.; Weintraub, D.; Sampaio, C.; Collaborators of the Parkinson’s Disease Update on Non-Motor Symptoms Study Group on Behalf of the Movement Disorders Society Evidence-Based Medicine Committee; Chahine, L. Update on treatments for nonmotor symptoms of Parkinson’s disease—An evidence-based medicine review. Mov. Disord. 2019, 34, 180–198. [Google Scholar] [CrossRef] [Green Version]
  106. Rossi, M.; Merello, M.; Perez-Lloret, S. Management of constipation in Parkinson’s disease. Expert Opin. Pharmacother. 2015, 16, 547–557. [Google Scholar] [CrossRef] [PubMed]
  107. Zangaglia, R.; Martignoni, E.; Glorioso, M.; Ossola, M.; Riboldazzi, G.; Calandrella, D.; Brunetti, G.; Pacchetti, C. Macrogol for the treatment of constipation in Parkinson’s disease. A randomized placebo-controlled study. Mov. Disord. Off. J. Mov. Disord. Soc. 2007, 22, 1239–1244. [Google Scholar] [CrossRef]
  108. Eichhorn, T.E.; Oertel, W.H. Macrogol 3350/electrolyte improves constipation in Parkinson’s disease and multiple system atrophy. Mov. Disord. Off. J. Mov. Disord. Soc. 2001, 16, 1176–1177. [Google Scholar] [CrossRef]
  109. Coggrave, M.; Norton, C.; Cody, J.D. Management of faecal incontinence and constipation in adults with central neurological diseases. Cochrane Database Syst. Rev. 2014, 1, CD00211. [Google Scholar] [CrossRef] [Green Version]
  110. Di Palma, J.A.; Smith, J.R.; vB Cleveland, M. Overnight efficacy of polyethylene glycol laxative. Am. J. Gastroenterol. 2002, 97, 1776–1779. [Google Scholar] [CrossRef]
  111. DiPalma, J.A.; DeRidder, P.H.; Orlando, R.C.; Kolts, B.E.; vB Cleveland, M. A randomized, placebo-controlled, multicenter study of the safety and efficacy of a new polyethylene glycol laxative. Am. J. Gastroenterol. 2000, 95, 446–450. [Google Scholar] [CrossRef]
  112. Klaschik, E.; Nauck, F.; Ostgathe, C. Constipation—Modern laxative therapy. Support. Care Cancer 2003, 11, 679–685. [Google Scholar] [CrossRef]
  113. McKeage, K.; Plosker, G.L.; Siddiqui, M.A.A. Lubiprostone. Drugs 2006, 66, 873–879. [Google Scholar] [CrossRef] [PubMed]
  114. Ondo, W.; Kenney, C.; Sullivan, K.; Davidson, A.; Hunter, C.; Jahan, I.; McCombs, A.; Miller, A.; Zesiewicz, T. Placebo-controlled trial of lubiprostone for constipation associated with Parkinson disease. Neurology 2012, 78, 1650–1654. [Google Scholar] [CrossRef] [PubMed]
  115. Cuppoletti, J.; Blikslager, A.T.; Chakrabarti, J.; Nighot, P.K.; Malinowska, D.H. Contrasting effects of linaclotide and lubiprostone on restitution of epithelial cell barrier properties and cellular homeostasis after exposure to cell stressors. BMC Pharmacol. 2012, 12, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. De Lisle, R.C. Lubiprostone stimulates small intestinal mucin release. BMC Gastroenterol. 2012, 12, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. 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]
  118. Manfredsson, F.P.; Luk, K.C.; Benskey, M.J.; Gezer, A.; Garcia, J.; Kuhn, N.C.; Sandoval, I.M.; Patterson, J.R.; O’Mara, A.; Yonkers, R.; et al. Induction of alpha-synuclein pathology in the enteric nervous system of the rat and non-human primate results in gastrointestinal dysmotility and transient CNS pathology. Neurobiol. Dis. 2018, 112, 106–118. [Google Scholar] [CrossRef]
  119. Uemura, N.; Yagi, H.; Uemura, M.T.; Hatanaka, Y.; Yamakado, H.; Takahashi, R. Inoculation of alpha-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the vagus nerve. Mol. Neurodegener. 2018, 13, 21. [Google Scholar] [CrossRef] [Green Version]
  120. Holmqvist, S.; Chutna, O.; Bousset, L.; Aldrin-Kirk, P.; Li, W.; Bjorklund, T.; Wang, Z.Y.; Roybon, L.; Melki, R.; Li, J.Y. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 2014, 128, 805–820. [Google Scholar] [CrossRef] [Green Version]
  121. Giancola, F.; Torresan, F.; Repossi, R.; Bianco, F.; Latorre, R.; Ioannou, A.; Guarino, M.; Volta, U.; Clavenzani, P.; Mazzoni, M.; et al. Downregulation of neuronal vasoactive intestinal polypeptide in Parkinson’s disease and chronic constipation. Neurogastroenterol. Motil. 2017, 29, e12995. [Google Scholar] [CrossRef] [Green Version]
  122. Chalazonitis, A.; Rao, M. Enteric nervous system manifestations of neurodegenerative disease. Brain Res. 2018, 1693, 207–213. [Google Scholar] [CrossRef]
  123. Braak, H.; de Vos, R.A.I.; Bohl, J.; Del Tredici, K. Gastric a-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]
  124. Shannon, K.M.; Keshavarzian, A.; Mutlu, E.; Dodiya, H.B.; Daian, D.; Jaglin, J.A.; Kordower, J.H. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov. Disord. 2012, 27, 709–715. [Google Scholar] [CrossRef] [PubMed]
  125. Singaram, C.; Ashraf, W.; Gaumnitz, E.A.; Torbey, C.; Sengupta, A.; Pfeiffer, R.; Quigley, E.M. Dopaminergic defect of enteric nervous system in Parkinson’s disease patients with chronic constipation. Lancet 1995, 346, 861–864. [Google Scholar] [CrossRef]
  126. Bassotti, G.; Villanacci, V.; Nascimbeni, R.; Cadei, M.; Fisogni, S.; Antonelli, E.; Corazzi, N.; Salerni, B. Enteric neuroglial apoptosis in inflammatory bowel diseases. J. Crohn Colitis 2009, 3, 264–270. [Google Scholar] [CrossRef] [Green Version]
  127. McKeown, S.J.; Stamp, L.; Hao, M.M.; Young, H.M. Hirschsprung disease: A developmental disorder of the enteric nervous system. Wiley Interdiscip. Rev. Dev. Biol. 2013, 2, 113–129. [Google Scholar] [CrossRef] [PubMed]
  128. De Giorgio, R.; Cogliandro, R.F.; Barbara, G.; Corinaldesi, R.; Stanghellini, V. Chronic intestinal pseudo-obstruction: Clinical features, diagnosis, and therapy. Gastroenterol. Clin. N. Am. 2011, 40, 787–807. [Google Scholar] [CrossRef] [PubMed]
  129. Gershon, M.D.; Liu, M.T. Serotonin and neuroprotection in functional bowel disorders. Neurogastroenterol. Motil. 2007, 19 (Suppl. 2), 19–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Mawe, G.M.; Hoffman, J.M. Serotonin signalling in the gut--functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. 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]
  132. Poole, D.P.; Xu, B.; Koh, S.L.; Hunne, B.; Coupar, I.M.; Irving, H.R.; Shinjo, K.; Furness, J.B. Identification of neurons that express 5-hydroxytryptamine4 receptors in intestine. Cell Tissue Res. 2006, 325, 413–422. [Google Scholar] [CrossRef]
  133. Frampton, J.E. Prucalopride. Drugs 2009, 69, 2463–2476. [Google Scholar] [CrossRef]
  134. Müller-Lissner, S.; Rykx, A.; Kerstens, R.; Vandeplassche, L. A double-blind, placebo-controlled study of prucalopride in elderly patients with chronic constipation. Neurogastroenterol. Motil. 2010, 22, 991-e255. [Google Scholar] [CrossRef]
  135. Shin, A.; Camilleri, M.; Kolar, G.; Erwin, P.; West, C.; Murad, M. Systematic review with meta-analysis: Highly selective 5-HT4 agonists (prucalopride, velusetrag or naronapride) in chronic constipation. Aliment. Pharmacol. Ther. 2014, 39, 239–253. [Google Scholar] [CrossRef] [Green Version]
  136. Bouras, E.P.; Camilleri, M.; Burton, D.D.; McKinzie, S. Selective stimulation of colonic transit by the benzofuran 5HT4 agonist, prucalopride, in healthy humans. Gut 1999, 44, 682–686. [Google Scholar] [CrossRef] [Green Version]
  137. Bouras, E.P.; Camilleri, M.; Burton, D.D.; Thomforde, G.; McKinzie, S.; Zinsmeister, A.R. Prucalopride accelerates gastrointestinal and colonic transit in patients with constipation without a rectal evacuation disorder. Gastroenterology 2001, 120, 354–360. [Google Scholar] [CrossRef]
  138. Omer, A.; Quigley, E.M.M. An update on prucalopride in the treatment of chronic constipation. Ther. Adv. Gastroenterol. 2017, 10, 877–887. [Google Scholar] [CrossRef]
  139. Bianco, F.; Bonora, E.; Natarajan, D.; Vargiolu, M.; Thapar, N.; Torresan, F.; Giancola, F.; Boschetti, E.; Volta, U.; Bazzoli, F.; et al. Prucalopride exerts neuroprotection in human enteric neurons. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G768–G775. [Google Scholar] [CrossRef] [Green Version]
  140. Freitas, M.E.; Alqaraawi, A.; Lang, A.E.; Liu, L.W.C. Linaclotide and Prucalopride for Management of Constipation in Patients with Parkinsonism. Mov. Disord. Clin. Pract. 2018, 5, 218–220. [Google Scholar] [CrossRef] [Green Version]
  141. Pinyopornpanish, K.; Soontornpun, A.; Kijdamrongthum, P.; Teeyasoontranon, W.; Angkurawaranon, C.; Thongsawat, S. The effect of prucalopride on gastric emptying in Parkinson’s disease patients, a pilot randomized, open-label study. Dig. Syst. 2017, 1, 1–6. [Google Scholar] [CrossRef]
  142. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [Green Version]
  143. Suzuki, T.; Hara, H. Role of flavonoids in intestinal tight junction regulation. J. Nutr. Biochem. 2011, 22, 401–408. [Google Scholar] [CrossRef]
  144. El-Horany, H.E.; El-Latif, R.N.; ElBatsh, M.M.; Emam, M.N. Ameliorative Effect of Quercetin on Neurochemical and Behavioral Deficits in Rotenone Rat Model of Parkinson’s Disease: Modulating Autophagy (Quercetin on Experimental Parkinson’s Disease). J. Biochem. Mol. Toxicol. 2016, 30, 360–369. [Google Scholar] [CrossRef]
  145. Li, X.; Wang, H.; Gao, Y.; Li, L.; Tang, C.; Wen, G.; Zhou, Y.; Zhou, M.; Mao, L.; Fan, Y. Protective Effects of Quercetin on Mitochondrial Biogenesis in Experimental Traumatic Brain Injury via the Nrf2 Signaling Pathway. PLoS ONE 2016, 11, e0164237. [Google Scholar] [CrossRef] [Green Version]
  146. Sandhir, R.; Mehrotra, A. Quercetin supplementation is effective in improving mitochondrial dysfunctions induced by 3-nitropropionic acid: Implications in Huntington’s disease. Biochim. Biophys. Acta 2013, 1832, 421–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Lv, C.; Hong, T.; Yang, Z.; Zhang, Y.; Wang, L.; Dong, M.; Zhao, J.; Mu, J.; Meng, Y. Effect of Quercetin in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-Induced Mouse Model of Parkinson’s Disease. Evid. Based Complement. Altern. Med. 2012, 2012, 928643. [Google Scholar] [CrossRef] [PubMed]
  148. Haleagrahara, N.; Siew, C.J.; Mitra, N.K.; Kumari, M. Neuroprotective effect of bioflavonoid quercetin in 6-hydroxydopamine-induced oxidative stress biomarkers in the rat striatum. Neurosci. Lett. 2011, 500, 139–143. [Google Scholar] [CrossRef] [PubMed]
  149. Figueira, I.; Menezes, R.; Macedo, D.; Costa, I.; Dos Santos, C.N. Polyphenols Beyond Barriers: A Glimpse into the Brain. Curr. Neuropharmacol. 2017, 15, 562–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Sharma, S.; Raj, K.; Singh, S. Neuroprotective Effect of Quercetin in Combination with Piperine against Rotenone- and Iron Supplement-Induced Parkinson’s Disease in Experimental Rats. Neurotox. Res. 2020, 37, 198–209. [Google Scholar] [CrossRef]
  151. Khan, H.; Ullah, H.; Aschner, M.; Cheang, W.S.; Akkol, E.K. Neuroprotective Effects of Quercetin in Alzheimer’s Disease. Biomolecules 2019, 10, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Singh, S.; Jamwal, S.; Kumar, P. Neuroprotective potential of Quercetin in combination with piperine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity. Neural Regen. Res. 2017, 12, 1137–1144. [Google Scholar] [CrossRef] [PubMed]
  153. Johansson, M.E. Mucus layers in inflammatory bowel disease. Inflamm. Bowel Dis. 2014, 20, 2124–2131. [Google Scholar] [CrossRef] [PubMed]
  154. Johansson, M.E.; Sjovall, H.; Hansson, G.C. The gastrointestinal mucus system in health and disease. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 352–361. [Google Scholar] [CrossRef] [Green Version]
  155. Turner, J.R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef] [PubMed]
  156. Piche, T.; Barbara, G.; Aubert, P.; Bruley des Varannes, S.; Dainese, R.; Nano, J.L.; Cremon, C.; Stanghellini, V.; De Giorgio, R.; Galmiche, J.P.; et al. Impaired intestinal barrier integrity in the colon of patients with irritable bowel syndrome: Involvement of soluble mediators. Gut 2009, 58, 196–201. [Google Scholar] [CrossRef] [Green Version]
  157. Peeters, M.; Ghoos, Y.; Maes, B.; Hiele, M.; Geboes, K.; Vantrappen, G.; Rutgeerts, P. Increased permeability of macroscopically normal small bowel in Crohn’s disease. Dig. Dis. Sci. 1994, 39, 2170–2176. [Google Scholar] [CrossRef]
  158. Salat-Foix, D.; Tran, K.; Ranawaya, R.; Meddings, J.; Suchowersky, O. Increased intestinal permeability and Parkinson disease patients: Chicken or egg? Can. J. Neurol. Sci. 2012, 39, 185–188. [Google Scholar] [CrossRef] [Green Version]
  159. Kelly, L.P.; Carvey, P.M.; Keshavarzian, A.; Shannon, K.M.; Shaikh, M.; Bakay, R.A.; Kordower, J.H. Progression of intestinal permeability changes and alpha-synuclein expression in a mouse model of Parkinson’s disease. Mov. Disord. 2014, 29, 999–1009. [Google Scholar] [CrossRef]
  160. Thulesen, J. Glucagon-like peptide 2 (GLP-2), an intestinotrophic mediator. Curr. Protein Pept. Sci. 2004, 5, 51–65. [Google Scholar] [PubMed]
  161. Pini, A.; Garella, R.; Idrizaj, E.; Calosi, L.; Baccari, M.; Vannucchi, M. Glucagon-like peptide 2 counteracts the mucosal damage and the neuropathy induced by chronic treatment with cisplatin in the mouse gastric fundus. Neurogastroenterol. Motil. 2016, 28, 206–216. [Google Scholar] [CrossRef]
  162. Zhang, Z.; Hao, L.; Shi, M.; Yu, Z.; Shao, S.; Yuan, Y.; Zhang, Z.; Hölscher, C. Neuroprotective Effects of a GLP-2 Analogue in the MPTP Parkinson’s Disease Mouse Model. J. Parkinson Dis. 2021, 11, 529–543. [Google Scholar] [CrossRef] [PubMed]
  163. Su, Y.; Zhang, Z.; Li, H.; Ma, J.; Sun, L.; Shao, S.; Zhang, Z.; Hölscher, C. A GLP-2 Analogue Protects SH-SY5Y and Neuro-2a Cells Against Mitochondrial Damage, Autophagy Impairments and Apoptosis in a Parkinson Model. Drug Res. 2021, 71, 43–50. [Google Scholar] [CrossRef] [PubMed]
  164. Ren, Z.; Yang, N.; Ji, C.; Zheng, J.; Wang, T.; Liu, Y.; Zuo, P. Neuroprotective effects of 5-(4-hydroxy-3-dimethoxybenzylidene)-thiazolidinone in MPTP induced Parkinsonism model in mice. Neuropharmacology 2015, 93, 209–218. [Google Scholar] [CrossRef] [PubMed]
  165. Brakedal, B.; Flones, I.; Reiter, S.F.; Torkildsen, O.; Dolle, C.; Assmus, J.; Haugarvoll, K.; Tzoulis, C. Glitazone use associated with reduced risk of Parkinson’s disease. J. Mov. Disord. 2017, 32, 1594–1599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Cao, L.; Li, D.; Feng, P.; Li, L.; Xue, G.F.; Li, G.; Holscher, C. A novel dual GLP-1 and GIP incretin receptor agonist is neuroprotective in a mouse model of Parkinson’s disease by reducing chronic inflammation in the brain. NeuroReport 2016, 27, 384–391. [Google Scholar] [CrossRef]
  167. Aviles-Olmos, I.; Dickson, J.; Kefalopoulou, Z.; Djamshidian, A.; Kahan, J.; Ell, P.; Whitton, P.; Wyse, R.; Isaacs, T.; Lees, A.; et al. Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson’s disease. J. Parkinsons Dis. 2014, 4, 337–344. [Google Scholar] [CrossRef] [Green Version]
  168. Labuzek, K.; Suchy, D.; Gabryel, B.; Bielecka, A.; Liber, S.; Okopien, B. Quantification of metformin by the HPLC method in brain regions, cerebrospinal fluid and plasma of rats treated with lipopolysaccharide. Pharmacol. Rep. 2010, 62, 956–965. [Google Scholar] [CrossRef]
  169. Campbell, J.M.; Stephenson, M.D.; de Courten, B.; Chapman, I.; Bellman, S.M.; Aromataris, E. Metformin Use Associated with Reduced Risk of Dementia in Patients with Diabetes: A Systematic Review and Meta-Analysis. J. Alzheimers Dis. 2018, 65, 1225–1236. [Google Scholar] [CrossRef] [Green Version]
  170. Jiang, T.; Yu, J.T.; Zhu, X.C.; Wang, H.F.; Tan, M.S.; Cao, L.; Zhang, Q.Q.; Gao, L.; Shi, J.Q.; Zhang, Y.D.; et al. Acute metformin preconditioning confers neuroprotection against focal cerebral ischaemia by pre-activation of AMPK-dependent autophagy. Br. J. Pharmacol. 2014, 171, 3146–3157. [Google Scholar] [CrossRef]
  171. Ng, T.P.; Feng, L.; Yap, K.B.; Lee, T.S.; Tan, C.H.; Winblad, B. Long-term metformin usage and cognitive function among older adults with diabetes. J. Alzheimers Dis. 2014, 41, 61–68. [Google Scholar] [CrossRef]
  172. Wahlqvist, M.L.; Lee, M.-S.; Hsu, C.-C.; Chuang, S.-Y.; Lee, J.-T.; Tsai, H.-N. Metformin-inclusive sulfonylurea therapy reduces the risk of Parkinson’s disease occurring with Type 2 diabetes in a Taiwanese population cohort. Parkinsonism Relat. Disord. 2012, 18, 753–758. [Google Scholar] [CrossRef] [PubMed]
  173. Rotermund, C.; Machetanz, G.; Fitzgerald, J.C. The Therapeutic Potential of Metformin in Neurodegenerative Diseases. Front. Endocrinol. 2018, 9, 400. [Google Scholar] [CrossRef]
  174. Ahmadi, S.; Razazan, A.; Nagpal, R.; Jain, S.; Wang, B.; Mishra, S.P.; Wang, S.; Justice, J.; Ding, J.; McClain, D.A.; et al. Metformin Reduces Aging-Related Leaky Gut and Improves Cognitive Function by Beneficially Modulating Gut Microbiome/Goblet Cell/Mucin Axis. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, e9–e21. [Google Scholar] [CrossRef]
  175. Deng, J.; Zeng, L.; Lai, X.; Li, J.; Liu, L.; Lin, Q.; Chen, Y. Metformin protects against intestinal barrier dysfunction via AMPKalpha1-dependent inhibition of JNK signalling activation. J. Cell Mol. Med. 2018, 22, 546–557. [Google Scholar] [CrossRef] [Green Version]
  176. Scarpello, J.H.; Hodgson, E.; Howlett, H.C. Effect of metformin on bile salt circulation and intestinal motility in type 2 diabetes mellitus. Diabetes Med. 1998, 15, 651–656. [Google Scholar] [CrossRef]
  177. Dandona, P.; Fonseca, V.; Mier, A.; Beckett, A.G. Diarrhea and metformin in a diabetic clinic. Diabetes Care 1983, 6, 472–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Menzies, D.G.; Campbell, I.W.; McBain, A.; Brown, I.R. Metformin efficacy and tolerance in obese non-insulin dependent diabetics: A comparison of two dosage schedules. Curr. Med. Res. Opin. 1989, 11, 273–278. [Google Scholar] [CrossRef]
  179. Minato, T.; Maeda, T.; Fujisawa, Y.; Tsuji, H.; Nomoto, K.; Ohno, K.; Hirayama, M. Progression of Parkinson’s disease is associated with gut dysbiosis: Two-year follow-up study. PLoS ONE 2017, 12, e0187307. [Google Scholar] [CrossRef] [Green Version]
  180. Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609–10620. [Google Scholar] [CrossRef]
  181. Bedarf, J.R.; Hildebrand, F.; Coelho, L.P.; Sunagawa, S.; Bahram, M.; Goeser, F.; Bork, P.; Wullner, U. Functional implications of microbial and viral gut metagenome changes in early stage L-DOPA-naive Parkinson’s disease patients. Genome Med. 2017, 9, 39. [Google Scholar] [CrossRef]
  182. Heintz-Buschart, A.; Pandey, U.; Wicke, T.; Sixel-Doring, 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. J. Mov. Disord. 2018, 33, 88–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Hill-Burns, E.M.; Debelius, J.W.; Morton, J.T.; Wissemann, W.T.; Lewis, M.R.; Wallen, Z.D.; Peddada, S.D.; Factor, S.A.; Molho, E.; Zabetian, C.P.; et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. J. Mov. Disord. 2017, 32, 739–749. [Google Scholar] [CrossRef]
  184. 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. J. Mov. Disord. 2015, 30, 350–358. [Google Scholar] [CrossRef]
  185. Unger, M.M.; Spiegel, J.; Dillmann, K.U.; Grundmann, D.; Philippeit, H.; Burmann, J.; Fassbender, K.; Schwiertz, A.; Schafer, 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] [PubMed]
  186. Barichella, M.; Cereda, E.; Cassani, E.; Pinelli, G.; Iorio, L.; Ferri, V.; Privitera, G.; Pasqua, M.; Valentino, A.; Monajemi, F.; et al. Dietary habits and neurological features of Parkinson’s disease patients: Implications for practice. Clin. Nutr. 2017, 36, 1054–1061. [Google Scholar] [CrossRef]
  187. 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]
  188. Tan, A.H.; Lim, S.Y.; Chong, K.K.; MAA, A.M.; Hor, J.W.; Lim, J.L.; Low, S.C.; Chong, C.W.; Mahadeva, S.; Lang, A.E. Probiotics for Constipation in Parkinson Disease: A Randomized Placebo-Controlled Study. Neurology 2021, 96, e772–e782. [Google Scholar] [CrossRef]
  189. Miller, L.E.; Ouwehand, A.C. Probiotic supplementation decreases intestinal transit time: Meta-analysis of randomized controlled trials. World J. Gastroenterol. 2013, 19, 4718. [Google Scholar] [CrossRef]
  190. Huang, H.; Xu, H.; Luo, Q.; He, J.; Li, M.; Chen, H.; Tang, W.; Nie, Y.; Zhou, Y. Fecal microbiota transplantation to treat Parkinson’s disease with constipation: A case report. Medicine 2019, 98, e16163. [Google Scholar] [CrossRef]
  191. Kuai, X.Y.; Yao, X.H.; Xu, L.J.; Zhou, Y.Q.; Zhang, L.P.; Liu, Y.; Pei, S.F.; Zhou, C.L. Evaluation of fecal microbiota transplantation in Parkinson’s disease patients with constipation. Microb. Cell Fact. 2021, 20, 98. [Google Scholar] [CrossRef] [PubMed]
  192. Arnold, C. The pros and cons of probiotics. Lancet Infect. Dis. 2013, 13, 571–572. [Google Scholar] [CrossRef]
  193. Brescia, P.; Rescigno, M. The gut vascular barrier: A new player in the gut–liver–brain axis. Trends Mol. Med. 2021, 27, 844–855. [Google Scholar] [CrossRef] [PubMed]
  194. Rocca, W.A. The burden of Parkinson’s disease: A worldwide perspective. Lancet Neurol. 2018, 17, 928–929. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Common PD-related GI dysfunctions and current treatments.
Figure 1. Common PD-related GI dysfunctions and current treatments.
Jpm 12 00144 g001
Figure 2. Potential new therapeutics for PD-related GI dysfunctions and sites of action.
Figure 2. Potential new therapeutics for PD-related GI dysfunctions and sites of action.
Jpm 12 00144 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Han, M.N.; Finkelstein, D.I.; McQuade, R.M.; Diwakarla, S. Gastrointestinal Dysfunction in Parkinson’s Disease: Current and Potential Therapeutics. J. Pers. Med. 2022, 12, 144. https://doi.org/10.3390/jpm12020144

AMA Style

Han MN, Finkelstein DI, McQuade RM, Diwakarla S. Gastrointestinal Dysfunction in Parkinson’s Disease: Current and Potential Therapeutics. Journal of Personalized Medicine. 2022; 12(2):144. https://doi.org/10.3390/jpm12020144

Chicago/Turabian Style

Han, Myat Noe, David I. Finkelstein, Rachel M. McQuade, and Shanti Diwakarla. 2022. "Gastrointestinal Dysfunction in Parkinson’s Disease: Current and Potential Therapeutics" Journal of Personalized Medicine 12, no. 2: 144. https://doi.org/10.3390/jpm12020144

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