Preventing and Treating Parkinson’s Disease with a Plant-Based Diet

Image of brain scans combined with article header
Posted by: Plant-Based Diet Team Post Date: May 11, 2021

Preventing and Treating Parkinson’s Disease with a Plant-Based Diet

Header of published article

This article was published in the Open Access Journal of Neurology and Neurosurgery

Abstract

Parkinson’s disease (PD) is the second most common human neurodegenerative disorder, but no current therapy has been proven to be disease-modifying. Epidemiological as well as interventional studies indicate that the plant-based diet has the potential to prevent and treat PD. There are pathophysiological reasons that make this likely to be true.

The Western diet is among the greatest risk factors for developing neurodegenerative diseases such as PD. Consumption of high quantities of animal saturated fat has been widely reported to be associated with increased risk of developing Parkinson’s disease. Pesticide, herbicide, and heavy metal exposures through the consumption of meat are linked to an increased risk of Parkinson disease in some epidemiologic studies. Interventional studies with a plant-based diet have achieved positive results.

Accumulating evidence indicates that oxidative damage and mitochondrial dysfunction contribute to the cascade of events leading to degeneration of dopaminergic neurons. In addition, dysbiosis of the gut microbiota may be involved in the pathogenesis of PD, inducing immune cell activation and neuroinflammation of the central nervous system.

The benefits of a plant-based diet result from the increased levels of phytonutrients and the intake of fiber, which supports a beneficial gut microbiota and decreases the incidence of constipation, an independent risk factor. A plant-based diet can also facilitate the use of a protein-redistribution diet to improve the effectiveness of treatment with L-dopa.

Keywords: dopaminergic, gut dysbiosis, inflammation, microbiota, neurodegenerative, oxidative stress, Parkinson’s, pesticide, plant-based diet, protein-redistribution

Abbreviations: 6-OHDA – 6-hydroxydopamine, CNS – central nervous system, CSF – cerebrospinal fluid, FA – Ferulic acid, nAChR – nicotinic acetylcholine receptors, PD – Parkinson’s Disease, RBD – rapid eye movement sleep behavior disorder, ROS – reactive oxygen species, SCFA – short chain fatty acids, SNpc – substantia nigra pars compacta, UPDRS – unified Parkinson’s disease rating stage.

Cite this article: Rose S, Strombom A. (2021) Preventing and Treating of Parkinson’s Disease with a Plant-Based Diet. Open Access J Neurol Neurosurg 15(2): 555906

 

Introduction

With aging and increasing life span of the global population, age-related diseases like Parkinson’s Disease (PD) are receiving increased attention from the scientific community. Neurological disorders are now the leading source of disability in the world, and PD is the fastest growing of these disorders. (1) It is, after Alzheimer’s disease, the second most common human neurodegenerative disorder. The total annual cost of Parkinson’s Disease in the United States is almost $52 billion. (2)

The main signs of PD include bradykinesia, which is the cardinal symptom, plus muscular rigidity, rest tremor, and gait impairment. The characteristic pathological finding associated with the motor signs of PD is degeneration of the dopaminergic neurons of the pars compacta of the substantia nigra, resulting in loss of dopamine in the striatum. (3)

It is now thought that the involvement of non-dopaminergic pathways in the evolution of PD account for the increasingly recognized non-motor symptoms that adversely impact the quality of life of patients with PD. (4, 5, 6)

The prodromal phase (up to 15–20 years before onset of motor symptoms) occurs while clinical signs of disease are not evident, but underlying neurodegeneration has started and progressed. (7) Clinical studies have shown that rapid eye movement sleep behavior disorder (RBD), depression, olfactory dysfunction, constipation, and autonomic dysfunction may be present during this period. (8, 9) The 2019 Movement Disorders Society diagnostic criteria for prodromic PD have added other new markers (such as diabetes mellitus and physical inactivity), facilitating a web-based calculation of prodromic risk. (10)

Effective therapy alleviates the manifestations of the disease, moving the symptomatic progression curve to the right by several years, but does not affect the disease process as such. (11) No therapy has yet been proven to be disease-modifying. (12) Epidemiological as well as interventional studies indicate that the plant-based diet has the potential to prevent and treat PD. There are pathophysiological reasons that make this likely to be true.

 

Epidemiology

The etiology of PD involves both genetic and environmental factors. Although PD is generally an idiopathic disorder, there are a minority of cases (10–15%) that report a family history, and about 5% have Mendelian inheritance. (13) Furthermore, an individual’s risk of PD is partially the product of as-yet-poorly-defined polygenic risk factors. (13) The genes that have been found to potentially cause PD are assigned a “PARK” name in the order they were identified. To date, 23 PARK genes have been linked to PD. (14)

There is a growing body of epidemiological evidence to support the case that diet impacts (positively or negatively) the development of neurodegenerative diseases such as PD, so interest has been growing in the influence of food and nutrients on the development of PD. The Western diet is among the greatest risk factors for developing neurodegenerative diseases such as PD, (15) (16) whereas nicotine and caffeine use are associated with decreased risks. (17)

Age-adjusted prevalence rates of Parkinson’s disease tend to be relatively uniform throughout Europe and America. However, sub-Saharan black Africans, rural Chinese, and Japanese, groups whose diets tend to be quasi-vegetarian, appear to enjoy substantially lower rates.  Since current PD prevalence in African-Americans is little different from that in whites, environmental factors are likely to be responsible for the low PD risk in Black Africans. (18)

Consumption of high quantities of animal saturated fat has been widely reported to be associated with increased risk of developing Parkinson’s disease. (19)  Three recent case control studies conclude that diets high in animal fat or cholesterol are associated with a substantial increase in risk for Parkinson’s disease. However, fat of plant origin does not appear to increase risk, (18, 20) and may even lower it. (21)

Dairy product consumption and drinking milk may increase one’s risk of PD independently of calcium intake (22, 23, 24, 25) particularly in men. (26)  A positive association between milk consumption and PD risk was also observed in women in one study. (27) In contrast, studies have also shown that diets with high vegetable and fruit intake are associated with a decreased risk for PD, particularly in men. (18)

Insulin resistance and diabetes accelerates deterioration of motor function, while inhibiting the effectiveness of levodopa treatment in PD patients. (28)  Multiple epidemiological studies suggest that body mass index (BMI), insulin resistance, and diabetes increase the risk of PD. (29) For example, a study of over 45,000 people in Finland demonstrated a positive association between BMI and risk of PD, (30) and a study in Denmark showed that having diabetes increased the risk of PD by nearly 40%. (31) The progression of neuropathology in PD may be accelerated by insulin resistance, as suggested by a study showing that dementia is associated with insulin resistance in PD patients (32).

There are also environmental contributions to the risk of PD. Pesticide, herbicide, and heavy metal exposures are linked to an increased risk of Parkinson disease in some epidemiologic studies. (17) Based on several comprehensive epidemiological studies, pesticide exposure appears to be a particular risk factor for Parkinson’s disease. (33, 34, 35)  The data supporting a role for organochlorines in increasing the risk of PD continue to grow, including a recent family-based case-control study that demonstrated such an association. (36)

 

Pathophysiology

Among the various neuronal types that degenerate in this disease, there is little doubt that the degeneration or loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) is responsible for the characteristic motor symptoms and drives symptomatic therapies. (37, 38) The hallmark lesions called Lewy bodies, eosinophilic inclusion bodies, are produced by the progressive accumulation of protein inclusions containing α-synuclein and ubiquitin in the cytoplasm of selected neurons, which leads to their death by necrosis and/or apoptosis. Lewy bodies are present mainly in the surviving neurons and are considered as the biological marker of neuronal degeneration in PD. (3)

While the etiology of PD remains poorly understood, several underlying pathophysiological mechanisms such as oxidative stress, neuroinflammation, iron dysregulation, mitochondrial dysfunction, excitotoxicity, loss of neurotrophic factors, glial activation, and endoplasmic reticulum stress, as well as protein misfolding and dysfunction in their degradation, have been credited as significant pathways for the development of therapeutic approaches. (39, 40)

Accumulating evidence indicates that oxidative damage and mitochondrial dysfunction contribute to the cascade of events leading to degeneration of dopaminergic neurons. (41, 42, 43, 44, 45) Furthermore, evidence suggests that possible modification of the gut microbiota may be involved in the pathogenesis of PD, inducing immune cell activation and neuroinflammation of the central nervous system. (46)

Organochlorine compounds exhibit chemical properties, toxicokinetic features and temporal and geographic-use patterns that make them reasonable candidates to contribute to the incidence of PD. (36) The organochlorine pesticide dieldrin is an extremely persistent organic pollutant.  It does not easily break down in the environment, and tends to bioaccumulate as it is passed along the food chain. Long-term exposure has proven toxic to a very wide range of animals including humans, far greater than to the original insect targets. (47)

Dieldrin has been found in human PD postmortem brain tissues, suggesting that this pesticide has potential to promote nigral cell death. Although dieldrin has been banned, humans continue to be exposed to the pesticide through contaminated dairy products and meats, due to the persistent accumulation of the pesticide in the food chain including farm animals. (47) People exposed to dieldrin are at about a 250 percent higher risk of developing Parkinson’s disease than the rest of the population. (48, 49)  Since organochlorine compounds bioaccumulate in animal tissue, those following a vegan diet will have a much lower level of exposure.

Inflammation

In 1988, McGeer’s research team suggested that inflammation could be the first pathogenic mechanism of PD. (50)  At the same time, it has been observed that the use of non-steroidal anti-inflammatory drugs (NSAID) decreases the risk of PD, and this could be considered as a proof of inflammogenic characteristics of the disease. (51)

While neuronal death has been described as evidence of ongoing central nervous system (CNS) inflammation (52), several scientific reports documented microglial activation, cytokine production, and the presence of autoantibodies, univocally indicating inflammatory processes in PD. (53, 54, 55, 56) In vitro assays employing a dopaminergic neuron model showed some membrane proteins to be targeted by antibodies present in cerebrospinal fluid (CSF) of affected patients (57). Research performed on post-mortem excised brains revealed higher concentrations of cytokines and proapototic proteins in the striatum and CSF of PD patients compared to levels found in healthy controls, pointing at inflammation as a constant element of the disease (58).

Through a further immunohistological study, McGeer et al. discovered several alterations in striatal microglial cells of patients with PD, which appeared to be activated by an increased synthesis of proinflammatory cytokines. (59) Nonetheless, it remains to be explained whether inflammation represents the first cause determining neurodegeneration, or if it results from a selective damage process and cell degeneration.

A plant-based diet has been shown to reduce markers of inflammation such as CRP. Lower levels of hs-CRP were found in those following a vegetarian diet for more than 2 years. (60, 61) An interventional study found that after 8 weeks on a vegan diet, hs-CRP was reduced 32%, even more than the American Heart Association diet. (62)

Oxidative Stress

Oxidative stress is a well-accepted concept in the etiology and progression of Parkinson’s disease. (63) Oxidative stress plays an important role in the degeneration of dopaminergic neurons in PD. Disruptions in the physiologic maintenance of the redox potential in neurons interfere with several biological processes, ultimately leading to cell death. Evidence has been developed for oxidative and nitrative damage to key cellular components in the PD substantia nigra. (64)

In addition to PD, several other neurodegenerative disorders including Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis are associated with oxidative stress as well, despite having distinct pathological and clinical features, (65) suggesting that oxidative stress is a common mechanism contributing to neuronal degeneration. (66) (67)

The extensive production of reactive oxygen species (ROS) in the brain may provide an explanation for the magnitude of the role that these reactive molecules play in PD. The brain consumes about 20% of the oxygen supply of the body, and a significant portion of that oxygen is converted to ROS. (68) ROS can be generated in the brain from several sources, both in neurons and glia, with the electron transport chain being the major contributor at the mitochondrial level. (69) (70)

As one of the main sites of ROS production, mitochondria are particularly susceptible to oxidative stress-induced damage. Unlike nuclear DNA, mitochondrial DNA (mtDNA) are unprotected by histone proteins and therefore are easy targets of oxidation. (71)  ROS production and mtDNA damage have been shown to increase with age, up to 10–20 folds higher than in nuclear DNA. (72) (73)

A plant-based diet reduces oxidative stress through its rich supply of antioxidants. (74, 75)  Phytonutrients (also called phytochemicals), naturally occurring protective chemicals found in foods of plant origin and in plant based diets, are reported to have antioxidant properties. (75)

Parkin is an E3 ubiquitin ligase that promotes mitophagy of damaged depolarized mitochondria while also boosting mitochondrial biogenesis, thereby helping to maintain efficient mitochondrial function. Boosting Parkin expression in the substantia nigra (SN) with viral vectors is protective in multiple rodent models of PD. Conversely, homozygosity for inactivating mutations of Parkin results in early-onset PD.

Moderate-protein plant-based diets, relatively low in certain essential amino acids, have the potential to boost Parkin expression by activating the kinase GCN2, which in turn boosts the expression of ATF4, a factor that drives transcription of the Parkin gene. (76)

Microbiome Dysbiosis

A pathological characteristic for PD is the presence of cytoplasmatic eosinophilic alpha-synuclein inclusions in the form of Lewy bodies in cell somata and Lewy neurites in axons and dendrites. (77) (78) (79) The alpha-synuclein protein is generally expressed in the CNS, mainly in presynaptic terminals. It is thought to be involved in the regulation of neurotransmission and synaptic homeostasis (80) (81).  Studies suggest that it plays a role in modulating the supply and release of dopamine.

There is some evidence that proinflammatory dysbiosis is present in PD patients, and could trigger inflammation-induced misfolding of alpha-synuclein and development of PD pathology. (82) It has been suggested that alpha-synuclein could act like a prion protein during PD pathogenesis. In this theory, pathologic misfolded alpha-synuclein is an ‘infectious’ protein, spreading pathology by forming a template that seeds misfolding for nearby alpha-synuclein protein, turning the previously healthy protein into a pathogenic protein. (83) (84)

Emerging evidence has indicated that gut microbiota dysbiosis plays a role in several neurological diseases, including PD. (79)  Evidence suggests that the enteric nerves are involved in the PD pathological progression towards the central nervous system.  In the course of PD, the enteric nerves and parasympathetic nerves are amongst the structures earliest and most frequently affected by alpha-synuclein pathology. (85) One of the most common non-motor symptoms of PD is gastrointestinal dysfunction, usually associated with alpha-synuclein accumulations and low-grade mucosal inflammation in the enteric nerves.

The gut-brain axis is believed to be a bidirectional signaling pathway between the gastrointestinal tract and central nervous system. (86, 87, 88, 89)  The role of the vagus nerve and its branches in the pathogenesis of PD has recently been brought into focus. A retrospective study demonstrated that individuals undergoing bilateral truncal vagotomy and super selective vagotomy were at a reduced risk of developing PD as compared to the general population. This observation supports a strong association of vagal nerve fibers with the pathogenesis of PD. (90)

PD pathogenesis may be caused or exacerbated by dysbiotic microbiota-induced inflammatory responses that could promote alpha-synuclein pathology in the intestine and the brain or by rostral to caudal cell-to-cell transfer of alpha-synuclein pathology caused by increased oxidative stress (due to an increase in pro-inflammatory bacteria). (79)

Dietary components might influence the gut-brain axis by altering microbiota composition or by affecting neuronal functioning in both the enteric nerves and the central nervous system (CNS). (91) Recent research has shown that intestinal microbiota interact with the autonomic and central nervous system via diverse pathways including the enteric nerves and vagal nerve. (85)

There has been detection of abnormalities in the GI microbiome (gut dysbiosis) in patients with PD (92, 93, 94) and the discovery of inflammatory changes in the intestinal mucosa, enteric nervous system, vagus nerve, and the brain of patients with PD. (95) One study has provided evidence that bacterial flora causes enteric inflammation in PD, and further reinforces the role of peripheral inflammation in the initiation and/or the progression of the disease. (96)  Additionally, intestinal permeability was increased and beneficial metabolites of microbiota function, such as short chain fatty acids (SCFA), were lower in those with PD compared to healthy controls (97)

SCFA butyrate has anti-inflammatory properties thought to be owing to an epigenetic mechanism or to the activation of SCFA receptors, leading to anti-inflammatory effects, anti-microbial effects, and to a decreased intestinal barrier leakiness. (98, 99, 100)

PD patients show an increased intestinal permeability that correlates with intestinal alpha-synuclein accumulation. (97) The increased intestinal permeability and the translocation of bacteria and inflammatory bacterial products such as lipopolysaccharides (LPS) might lead to inflammation and oxidative stress in the GI tract, thereby initiating alpha-synuclein accumulation in the enteric nerves. (97) (101) (102)  In addition, gut-derived LPS can promote the disruption of the blood brain barrier, (103) and thus facilitate neuroinflammation and injury in the SN that is triggered by dysbiosis.

It is not possible to determine for sure if changes in the gut microbiota are a cause or a consequence of PD pathogenesis. However, it might still play a role in neuronal loss by perpetuating inflammatory cascades and oxidative injury in the brain through a lipopolysaccharide-mediated mechanism. (91) The LPS from pro-inflammatory intestinal flora bacteria can induce a chronic subclinical inflammatory process.

The importance of fiber

The term prebiotics was first introduced in 1995 by Gibson and Roberfroid as “a non-digestible food ingredient that beneficially affects the host by selectively stimulating the growth and/or activity of one or a limited number of bacteria in the colon, and thus improves host health” (104). Since then, the original definition has been revised several times and recently broadened to ‘a substrate that is selectively utilized by host microorganisms conferring a health benefit’ (105). This should not be confused with probiotics, defined as “live microorganisms that confer a health benefit on the host when administered in adequate amounts” (106)

The difference in gut microbiota composition between individuals following vegan or vegetarian diets and those following omnivorous diets is well documented. One way that a plant-based diet appears to be beneficial for human health is by promoting the development of more diverse and stable microbial systems. (91)

Such diets are high in dietary fiber and fermentable substrate (i.e. non digestible or undigested carbohydrates), which are sources of metabolic fuel and encourage the growth of species that ferment fiber into metabolites as short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate, that may be used by the host. These end products may have direct or indirect effects on modulating the health of their host. (107) Apart from being the preferred energy source for colonic epithelial cells, butyrate is involved in anti-inflammatory, enteroendocrine and epigenetic mechanisms that influence colonic and systemic health, including improved immunity against pathogens, blood–brain barrier integrity, brain function, provision of energy substrates, and regulation of critical functions of the intestine. (108) (109) Fecal SCFA concentrations have been found to be significantly reduced in PD patients. (110)

Phytochemicals

A plant-based diet includes phytochemicals that have a therapeutic effect, including flavonoids, nicotine and caffeine.

Flavonoids are the most common groups of polyphenols in human diet (111). Many plant-based foods and beverages are rich in flavonoids, such as berry fruits and citrus fruits (112). Flavonoids have high antioxidant capacity (113). They have been shown to modulate oxidative-related enzymes and regulate mitochondrial function in neurons. (111, 114) These findings point to a potential protective role of flavonoids in PD. In experimental studies, administration of flavonoids or flavonoid-rich foods (eg. berry fruits) protected dopamine neurons from oxidative damage and apoptosis and inhibited formation of α-synuclein fibrils. (115)

Nicotine

Nicotine is the addictive phytochemical in tobacco, which is derived from plants in the Nicotiana species of the Solanaceae family.  Nicotine accounts for approximately 95% of the total alkaloid content of tobacco, while the structurally-related nornicotine and anatabine are the most abundant minor pyridine alkaloids, accounting for 4 to 5% of total alkaloids (116). Other pyridine alkaloids in tobacco, such as anabasine, anabaseine, and cotinine, are present in smaller amounts (117).

Nicotine and all minor tobacco alkaloids have been shown to be pharmacologically active upon binding to several nicotinic acetylcholine receptors (nAChRs) (118). Tobacco nAChR agonists such as nicotine, anatabine, anabasine, anabaseine, and cotinine, display protective effects in animal models of several inflammatory conditions, including sepsis, (119) Parkinson’s disease, (120) Alzheimer’s disease, (121) and Inflammatory Bowel Disease. (122)

In a neuro-image study, a substantial portion of nicotine receptors became occupied when exposed to relatively small amount of nicotine. (123) This notion has further been supported by the observation that long-term smoking is more important than smoking intensity in the smoking-PD relationship. (124, 125) However, this does not necessarily indicate that cigarette smoking is advisable for the prevention or treatment of PD.

Besides cigarettes, nicotine is found in some common vegetables that belong to the biological family of nightshades. Other species in this family include Capsicum and Solanum, whose edible fruits and tubers include peppers, tomatoes, potatoes and eggplants. All these nightshades contain nicotine. (126, 127, 128, 129, 130) The nicotine levels in fresh potatoes, tomatoes and sweet peppers were only up to 10 μg/kg. Processed products contained equivalent or slightly higher levels of nicotine than fresh products (up to 34 μg/kg). Somewhat higher levels were found in fresh eggplant fruits (up to 100 μg/kg). (131)

The amount of nicotine absorbed from these foods is negligible relative to the amount obtained from active smoking. (131) However, even nicotine blood levels reached from environmental tobacco smoke exposure, much lower than that from active smoking, are sufficient to saturate a substantial portion of α4β2 nicotine receptors in the human brain. (123) Stimulation of nicotine receptors protects dopaminergic neurons in animal models of PD using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (132) or rotenone. (133) Therefore, given the strength of the association observed between PD and environmental tobacco smoke, (134) the small amounts found in solanaceae vegetables may confer a reduced risk of PD.

In a recent case-control study including 241 PD cases, each additional serving of edible solanaceae was associated with 31% lower risk of PD among never-smokers (135). It remains unclear as to whether the observed protective effect was due to the nicotine content or other components of this group of vegetables, but consumption of all other vegetables combined showed no association.  Another study showed a 30% decreased risk of PD for those in the highest quintile of nicotine in their diet. (136)

Constituents of peppers other than nicotine may also be neuroprotective. Another alkyloid, anatabine, is an intriguing possibility because it has anti-inflammatory properties (137, 121, 138) and might be more feasibly employed as a neuroprotective chemical than nicotine, due to its longer half-life and perhaps lower toxicity and addictive potential.

Capsinoids in peppers and capsaicinoids in spicy peppers may also be neuroprotective. They activate transient receptor potential cation channel subfamily vanilloid member 1 (TRPV1) receptors. (139) These receptors are in the substantia nigra, and they and capsaicin may affect survival of midbrain dopaminergic neurons. (140, 141)

Cruciferous vegetables such as cauliflower, cabbage, and broccoli, are another group of vegetables rich in antioxidants with neuroprotective capacity. For example, sulforaphane and erucin are potent, naturally occurring, isothiocyanates found in cruciferous vegetables with antioxidant properties. Treatment with sulforaphane ameliorated motor deficits, and protected dopaminergic neurons, in a 6-OHDA mouse model of PD (142). Similarly, erucin provided neuroprotective effects by preventing oxidative damage induced by 6-OHDA in an in vitro model (143). Both sulforaphane and erucin appear to be promising neuroprotective agents in chronic neurodegenerative diseases. (144) Taken together, these findings highlight the effects of some vegetables, fruits, and constituents they contain, as having neuroprotective potential.

Caffeine

The consumption of coffee or caffeinated food is associated with the reduction of the risk of PD. Patients with PD are less frequent habitual consumers of caffeinated food (145) (22). The consumption of either tea or coffee exhibited similar effects on the reduction of the risk of PD in a dose dependent manner (146), thus establishing caffeine as a neuroprotective phytochemical.

Caffeine is an adenosine A2A receptor antagonist (147). Different types of adenosine receptors (A1, A2A, A2B, and A3) are widely distributed in the brain. Adenosine A2A receptors are coupled with G-proteins and exclusively expressed in dopaminergic neurons. The activation of adenosine A2A receptors causes an increase in intracellular cAMP levels and the extracellular release of glutamate, resulting in neural excitotoxicity (148). The neuroprotective effects of caffeine involved the antagonism of the adenosine A2A receptor, down-regulating the down streaming phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, and avoiding excessive calcium releasing-related neurotoxicity and neuroinflammation (149), which has been experimentally demonstrated in several in vivo models of PD. (150, 151, 152, 153)

 

Interventional studies

In spite of extensive research, the pharmacotherapeutic options for Parkinson’s disease are limited to those which offer only symptomatic relief and cannot prevent the disease progression, so there is still need for effective disease-modifying agents for PD. (154, 155) The pharmacotherapeutic approach with antioxidants is gradually being preferred as disease-modifying strategies in neurodegenerative diseases, including PD. (155, 156, 157, 158, 159, 160)  Chronic consumption of anti PD drugs by PD patients, and their reduced efficacy, are leading researchers to look for probable synthetic or natural compounds and dietary treatment for the PD therapeutics.

A number of pharmacological, genetic, and clinical studies, including postmortem PD brain studies, show that mitochondrial defects, increased reactive oxygen species (ROS), and induction of inflammatory mediators play a very critical role in the development of PD. (161, 162)  Accordingly, oxidative stress and inflammatory processes are the major therapeutic targets of intervention to delay the development and progression of PD. (154)

The concept of neuroprotection referring to the prevention of dopamine cell death, and hence to retarding or halting disease progression, appears to be a promising strategy. It is convincing that naturally occurring molecules, possessing antioxidant and anti-inflammatory activities along with other pharmacological properties, could be effective in preventing or halting the neurodegenerative processes. (158, 159, 160) Various substances exhibiting anti-inflammatory, antioxidant, and metal chelating activity in the central nervous system (CNS) have been tested for use to facilitate the management of PD.

In this context, natural polyphenols have recently raised much attention. (163) Numerous studies have indicated the neuroprotective effects of natural polyphenols including epigallocatechin, quercetin, baicalein, resveratrol, luteolin, curcumin, puerarin, genistein, and hyperoside naringin, against dopaminergic neuronal death with relatively safe with uncommon, mild or transient side effects. (164)

Another potential treatment is the use of ferulic acid (FA).  FA is an important component of widely used medicinal herbs and belongs to the family of hydroxycinnamic acid. Pure form of FA appears as yellowish powder, and it has structural resemblance to curcumin, one of the well-studied natural molecules with potent neuroprotective effects. FA is highly abundant in the leaves and seeds of many plants, especially in cereals such as brown rice, whole wheat, and oats. It has been credited with many pharmacological properties including neuronal progenitor cell proliferation, anti-inflammatory, antioxidant, and neuroprotective activities, (165, 166, 167, 168, 169, 170, 171, 172, 173, 174)

The neuroprotective effect of FA has been reported in several experimental studies including brain injury, spinal ischemia, and Alzheimer-like pathology, (168, 169, 171) so it is reasonable to assume that it could also have a therapeutic effect for PD patients.

Food also appears to affect the pharmacokinetic and pharmacodynamic of levodopa, a prodrug of dopamine which remains the most effective agent to alleviate motor dysfunction in Parkinson’s disease. L-Dopa can enter the brain and be decarboxylated to dopamine only after crossing the blood–brain barrier by means of a specific saturable carrier system (the large neutral amino acid transporter, LAT). At this level, it competes with some dietary essential amino acids (the large neutral amino acid, LNAA, including tyrosine) which block L-dopa entry into the brain, even if blood concentrations of the drug are adequate. (175)

To avoid the risk of nutritional deficiencies linked to an extreme restriction in total protein intake, some doctors use the “protein redistribution diet.” This intervention consists of a normoproteic diet (protein calories about 10–15% of total calories, about  0.8–1.0 g/kg/day) with the main protein intake concentrated in the evening meal, in order to limit the negative interaction of LNAA on L-dopa response during daytime, and let the negative effect act at night-time during sleep. (176) An effect of this intervention was noticed within one week in patients who had previously benefited from L-dopa therapy (177, 178).  The effect of the protein distribution lasted for years. (179)

Some researchers have described an increased bioavailability of L-dopa by increasing insoluble fiber consumption. (179, 180) A study was designed to ascertain whether a plant-food normoproteic protein-redistributed diet can be as effective as a protein-redistributed omnivorous one in improving motor performances in PD patients in the short term. (180)

This study compared the effect of a plant-based diet with an omnivorous diet on motor performance. After 4 weeks, patients following a plant-based diet showed a significant reduction (Mann–Whitney test) in the Unified Parkinson’s Disease Rating Scale, total score (47.67 vs. 74.46) and sub-score III motor performances (25.42 vs. 46.46), and the modified Hoehn and Yahr Staging Scale (1.96 vs. 3.15).  The patients in this study had L-dopa daily dosage of over 350 mg, but under 850 mg, their age was over 50 years, and their BMI was over 18.5 but under 30. Two thirds of the plant protein was consumed at night. (180)

So a plant food (vegan) diet can be a convenient way to conjugate the positive effect of limited protein intake and high fiber intake without limiting total food amount. Due to its high fiber content, a plant-food diet can also potentially raise levodopa bioavailability by reducing the phenomenon of constipation. (181)

Another intervention that was tried was the use of omega-3 fatty acids and Vitamin E. In a randomized double-blind placebo-controlled clinical trial, patients received either 1000 mg omega-3 fatty acids from flaxseed oil plus 400 IU vitamin E supplements for 12 weeks, or a placebo. Unified Parkinson’s disease rating stage (UPDRS) were recorded at baseline and after 3 months of intervention.  After 12 weeks of intervention, omega-3 fatty acids and vitamin E co-supplementation led to a significant improvement in UPDRS. Furthermore, co-supplementation decreased high-sensitivity C-reactive protein (CRP) and increased total antioxidant capacity (TAC) and glutathione (GSH). Insulin resistance improved along with beta cell function. (182) The omega-3 fatty acids were from flaxseed oil, showing that animal sources of omega-3 such as fish are not required.

Riboflavin-sensitive mechanisms involved in PD may include glutathione depletion, cumulative mitochondrial DNA mutations, disturbed mitochondrial protein complexes, and abnormal iron metabolism. (183) Another study combined the elimination of red meat with riboflavin supplementation. All PD patients received 30 mg riboflavin orally at about 8-hr intervals (90 mg/day) and their usual symptomatic medications. This dosage was used to avoid decreased absorption associated with higher doses or shorter intervals between administrations. (183) Due to the renal excretion of riboflavin, the treatment was only initiated after confirmation of normal blood levels of creatinine (0.5-1.4 mg/dl). (154)

Because the PD patients had a higher consumption of red meat (beef and pork) than sex-matched controls (19 healthy non-consanguineous relatives or neighbors of similar age recruited for controlling the dietary habits), all PD patients were required to eliminate all red meat from their diets. The symptomatic drugs for PD in use included L-Dopa with carbidopa (200/50 mg tablets), L-Dopa with benserazide hydrochloride (200/50 mg tablets), biperiden (2 or 4 mg tablets), amantadine hydrochloride (100 mg tablets), selegiline (5 mg tablets), and pramipexole (0.25 or 1.0 mg tablets) taken alone or in diverse combinations. The treatment paradigm when the study began, with symptomatic drugs for PD for each patient, was maintained.

All patients who completed 6 months of treatment showed improved motor capacity during the first three months, and most reached a plateau while 5/19 continued to improve in the 3- to 6-month interval. Their average motor capacity increased from 44 to 71% after 6 months, increasing significantly every month compared with their own pretreatment status (P < 0.001, Wilcoxon signed rank test).  Discontinuation of riboflavin for several days did not impair motor capacity and yellowish urine was the only side effect observed. (183)

People with PD tend to be at greater odds of having a CoQ10 deficiency, an antioxidant that is important for the detoxification system, which may be partially responsible for their toxic burden (184). Several high-quality studies have shown that supplementation with 100–1,200 mg of CoQ10 daily, particularly larger dosages, reduces inflammatory markers and improves motor symptoms. (185, 186, 187, 188, 189)

 

Case study

Increasing evidence suggests that Parkinson disease consists of heterogeneous subtypes. Subtypes have implications for diagnosis, prognosis, and expected treatment response. Initial sub-typing focused on motor features. (190, 191)  Definitive diagnoses for both Parkinsonisms and Parkinson’s disease can only be done via an autopsy. (192)  A case study was done on a patient with Parkinsonism that includes Parkinson’s disease in the differential diagnosis. (193)

A 64-year-old man had depression since his 30’s that was treated with the MAO inhibitor tranylcypromine.  At age 51, he had onset of urinary frequency/urgency and erectile dysfunction. About 4 years later, he developed bradykinesia, bilateral rigidity, start hesitation, and sudden transient freezing. He did not have tremor and there were no vascular risk factors. An MRI of the brain was unremarkable.

This patient was diagnosed with levodopa-responsive Parkinsonism, characterized mainly by start hesitation, gait freezing, and prominent autonomic dysfunction. The differential diagnosis included PD with dysautonomia or multiple system atrophy. Vascular parkinsonism was unlikely given the absence of both vascular risk factors and vascular abnormalities on brain imaging. He was tried on trihexyphenidyl and amantadine with no response, and experienced only modest benefit from dopamine agonists.

The patient changed his diet to avoid protein during breakfast and lunch, and found that he had a better, more predictable response to levodopa. After 2 months, he adopted a vegan diet and since then has experienced steady and dramatic improvement in his motor symptoms. His gait has returned to almost normal, with near complete resolution of freezing and start hesitation. He now runs and ice skates, activities nearly impossible previously, with no difficulty. He was able to reduce levodopa from 2175 mg/day to 1305 mg/day, although symptoms recur when levodopa is reduced further. (193)

A protein redistribution diet (PRD) in this patient was useful, probably by reducing competition from diet-derived amino acids for transport across the intestinal and the blood-brain barrier, as has been described previously (194). More remarkable, however, was his response to a vegan diet that included only organic, plant-based foods and eliminated all animal-derived products such as eggs, cheese, and other milk products.

The benefits of a vegan diet may be derived from its protein-sparing qualities, which may be stricter and more consistent than the protein redistribution diet he used. A plant-based diet is also generally rich in fiber, which may improve bowel motility, thereby promoting the bioavailability of levodopa. (179)

Although its influence on levodopa pharmacokinetics is one mechanism for the fairly quick clinical benefit produced, a vegan diet may have other benefits for PD that contributed to the patient’s improvement. This includes an antioxidative effect, with its high levels of antioxidants slowing the loss of surviving dopaminergic neurons, thus retarding progression of the syndrome. Other benefits are the anti-inflammatory properties, caloric reduction, promotion of vascular health and aiding blood-brain barrier transport of L-dopa. (195, 18) All of these actions are relevant to the current understanding of factors that influence neurodegeneration in PD.

 

Clinical Considerations

The incidence of Parkinson’s disease rapidly increases over the age of 60 years, with only 4% of the cases being under the age of 50. (196)  The prevalence of chronic diseases such as type 2 diabetes, (197) coronary artery disease, (198) prostate (199) and colon cancer (200) increases during this time as well. Hence comorbidities will be common.

A plant-based diet protects against chronic oxidative-stress-related diseases. Dietary plants contain variable chemical families and amounts of antioxidants. Plant antioxidants may contribute to the beneficial health effects of dietary plants. (201) On average plant foods provide 11.57 mmol/ 100gm antioxidant content, while animal foods provide only on average 0.18 mmol/100gm. (201)

Recently, clinical and scientific attention has shifted to treating additional nonmotor symptoms that in the past have often passed unheeded. (202) Constipation is one of the most frequent non-motor symptoms in the autonomic system (203, 204) and gastrointestinal disturbance of PD (205). Between 50% and 80% of PD patients suffer from constipation. (206, 207, 208) (205) (209)   It has been reported that constipation can precede motor symptoms by as much as 20 years (210) and people with constipation may have a relatively high risk of developing PD (211). Accordingly, constipation may predict the occurrence of PD. However, PD patients may not talk about their symptom of constipation actively, leading to this problem not being reported in time. (212)

Underlying causes for constipation in PD are multifaceted. Besides physical weakness, lifestyle risks such as lack of fiber and reduced fluid intake may substantially promote its emergence. (213) Moreover, side effects of medication and disease-related pathomechanisms have been identified. (214, 215, 216) Regarding the latter, two usually concomitant alterations require distinction: slow intestinal transit and outlet obstruction. Increasing evidence indicates that delayed colonic transit in PD stems from disordered central, as well as peripheral, parasympathetic system dysregulation. (217)

On top of functional impairment, psychosocial distress increases with constipation in PD, strongly suggesting a negative impact on the quality of life. (212, 218, 219, 220) These manifold characteristics of PD-associated constipation highlight an urgent demand for efficacious treatment. Comprehensive and valuable reviews have emerged on the topic of PD-related constipation in recent years. (221, 222, 223)

In one study, the effects of a diet rich in insoluble fiber (DRIF) on motor disability, and the peripheral pharmacokinetics of orally administered L-dopa, in Parkinsonian patients with marked constipation were analyzed. (179) A useful effect of a DRIF on plasma L-dopa concentration and motor function was found. The greatest effect on the plasma L-dopa levels was found early (at 30 and 60 min) after oral administration. There was a relationship between the improvement of constipation and the higher bioavailability of L-dopa. DRIF can be a coadjuvant treatment in patients with Parkinson’s disease.

In PD subjects with confirmed constipation, adding psyllium to the diet increased stool frequency and weight, but did not alter colonic transit or anorectal function. Psyllium produced both subjective and objective improvements in constipation related to PD. (209) Prospectively obtained stool diaries should be employed to confirm constipation in PD.

Discussion

There are no disease modifying drugs to treat Parkinson’s disease. This makes prevention all the more important. A plant-based diet can help reduce the risk of Parkinson’s disease. In particular, several phytochemicals present in plant foods help reduce the risk of Parkinson’s disease. The increased fiber in a plant-based diet promotes butyrate-producing flora thus reducing inflammation, and it treats constipation, a risk factor for Parkinson’s disease.  Eliminating meat from the diet plays its part in reducing the risk. Therefore, both the presence of plant foods and the absence of meat and dairy combine to reduce risk.

Treatment with a plant-based diet can enable the dosage of medications used to treat the symptoms to be reduced. This may be especially advantageous in treating constipation caused by anticholinergics. The long-term benefit of a plant-based diet in slowing the progression of Parkinson’s disease should be researched. Its efficacy in treating nonmotor symptoms should also be researched further.

Since Parkinson’s disease often occurs later in life, the fact that a plant-based diet can reduce the risk, as well as treat common comorbidities such as type 2 diabetes, cardiovascular disease, prostate and colon cancer, makes prophylaxis with it all the more valuable.

Treating the Parkinson’s disease patients with a plant-based diet has no contraindications or adverse reactions. It offers a safe treatment as an adjunct to treatment with standard pharmacotherapy. A plant-based diet is affordable and can also relieve the financial stress on the Parkinson’s disease patient which is considerable.

 

References

1.

Dorsey ER, Bloem BR. (2018) The Parkinson Pandemic-A Call to Action. JAMA Neurol. 75(1):9-10. Pubmed

2.

Hamilton J, The Michael J. Fox Foundation for Parkinson’s Research, Yang W, The Lewin Group, et al. (2019) The Economic Burden of Parkinson’s Disease. Vol Silver Book. Washington DC: Alliance for Aging Research. MichaelJFox.org

3.

Kalia LV, Lang AE. (2015) Parkinson’s disease. Lancet. 2015;386: 896–912. The Lancet

4.

Chaudhuri KR, Sauerbier A. (2016) Parkinson disease:Unravelling the nonmotor mysteries of Parkinson disease. Nat Rev Neurol. 12:10–11. Pubmed

5.

Aarsland D, Creese B, Politis M, Chaudhuri KR, Ffytche D, et al. (2017) Cognitive decline in Parkinson disease. Nat Rev Neurol. 13:217–231. Pubmed

6.

Hughes KC, Gao X, Baker JM, Stephen C, Kim I, et al. (2018) Non-motor features of Parkinson’s disease in a nested case–control study of US men. J Neurol Neurosurg Psychiatry. 89:1288–1295. Pubmed

7.

Mantri S, Morley J, Siderowf A. (2019) The importance of preclinical diagnostics in Parkinson disease. Parkinsonism Relat Disord. 64:20–28. Pubmed

8.

Postuma RB, Aarsland D, Barone P, Burn D, Hawkes C, et al. (2012) Identifying prodromal Parkinson’s disease: pre-motor disorders in Parkinson’s disease. Mov Disord. 27:617–626. Pubmed

9.

Poewe W, Seppi K, Tanner CM, Halliday G, Brundin P, et al. (2019) Parkinson disease. Nat Rev Dis Primers. 2017;3:1701. Pubmed

10.

Heinzel S, Berg D, Gasser T, Chen H, Yao C, et al. (2019) Update of the MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 34(10):1464–1470. Pubmed

11.

Mattle H, Mumenthaler M. (2006) Fundamentals of neurology: Thieme. Amazon

12.

Hauser S, Josephson S. (2013) Harrison’s Neurology in Clinical Medicine. 3 ed: McGraw-Hill Education. Amazon

13.

Deng H, Wang P, Jankovic J. (2018) The genetics of Parkinson disease. Ageing Res Rev. 42:72–85. Pubmed

14.

Schulte C, Gasser T. (2011) Genetic basis of Parkinson’s disease: Inheritance, penetrance, and expression. Appl Clin Genet. 4:67–80. Pubmed

15.

Mattson M. (2014) Interventions that improve body and brain bioenergetics for Parkinson’s disease risk reduction and therapy. J Parkinsons Dis. 4:1–13. EuropePMC

16.

Mischley L, Lau R, Bennett R. (2017) Role of diet and nutritional supplements in Parkinson’s disease progression. Oxid Med Cell Longev. 2017:6405278. Pubmed

17.

Kouli A, Torsney K, Kuan W. (2018) Parkinson’s disease: etiology, neuropathology, and pathogenesis. In: Stoker T, Greenland J, eds. Parkinson’s disease: pathogenesis and clinical aspects [internet]. Brisbane, Australia: Codon Publications. Pubmed

18.

McCarty MF. (2001) Does a vegan diet reduce risk for Parkinson’s disease? Medical Hypotheses. 57(3):318-323. Pubmed

19.

Anderson C, Checkoway H, Franklin G, Beresford S, Smith-Weller T, et al. (1999) Dietary factors in Parkinson’s disease: the role of food groups and specific foods. Mov Disord. 14:21–27. Pubmed

20.

Logroscino G, Marder K, Cote L, Tang M, Shea S, et al. (1996) Dietary lipids and antioxidants in Parkinson’s disease: a population-based, case-control study. Ann Neurol. 39(1):89-94. Pubmed

21.

de Lau L, Bornebroek M, Witteman J, Hofman A, Koudstaal P, et al. (2005) Dietary fatty acids and the risk of Parkinson disease: the Rotterdam study. Neurology. 64(12):2040-2045. Pubmed

22.

Hellenbrand W, Seidler A, Boeing H, Robra B, Vieregge P, et al. (1996) Diet and Parkinson’s disease. I: A possible role for the past intake of specific foods and food groups. Results from a self-administered food-frequency questionnaire in a case-control study. Neurology. 47(3):636-643. Pubmed

23.

Chen H, Zhang S, Hernán M, Willett W, Ascherio A. (2002) Diet and Parkinson’s disease: a potential role of dairy products in men. Ann Neurol. 52(6):793-801. Pubmed

24.

Park M, Ross GW, Petrovitch H, White L, Masaki K, et al. (2005) Consumption of milk and calcium in midlife and the future risk of Parkinson disease. Neurology. 64:1047–1051. Pubmed

25.

Kyrozis A, Ghika A, Stathopoulos P, Vassilopoulos D, Trichopoulos D, et al. (2013) Dietary and lifestyle variables in relation to incidence of Parkinson’s disease in Greece. Eur J Epidemiol. 28(1):67-77. Pubmed

26.

Chen H, O’Reilly E, McCullough M, Rodriguez C, Schwarzschild M et al. (2007) Consumption of dairy products and risk of Parkinson’s disease. Am J Epidemiol. 165(9):998-1006. Pubmed

27.

Sääksjärvi K, Knekt P, Lundqvist A, Männistö S, Heliövaara M, et al. (2013) A cohort study on diet and the risk of Parkinson’s disease: the role of food groups and diet quality. Br J Nutr. 109(2):329-337. Pubmed

28.

Sandyk R. (1993) The relationship between diabetes mellitus and Parkinson’s disease. Int J Neurosci. 69(1-4):125-130. Pubmed

29.

Aviles-Olmos I, Limousin P, Lees A, Foltynie T. (2013) Parkinson’s disease, insulin resistance and novel agents of neuroprotection. Brain. 136(2):374–384. Pubmed

30.

Hu G, Jousilahti P, Nissinen A, Antikainen R, Kivipelto M, et al. (2006) Body mass index and the risk of Parkinson disease. Neurology. 67:1955-1959. Pubmed

31.

Schernhammer E, Hansen J, Rugbjerg K, Wermuth L, Ritz B. (2011) Diabetes and the risk of developing Parkinson’s disease in Denmark. Diabetes Care;34:1102-1108. Pubmed

32.

Boscoa D, Plastino M, Cristiano D, Colica C, Ermio C, et al. (2012) Dementia is associated with insulin resistance in patients with Parkinson’s disease. J Neurol Sci. 315(1-2):39-43. Pubmed

33.

Ascherio A, Chen H, Weisskopf MG, O’Reilly E, McCullough M, et al. (2006) Pesticide exposure and risk for Parkinson’s disease. Ann Neurol. 60:197–203. Pubmed

34.

Frigerio R, Elbaz A, Sanft KR,  Peterson B, Bower J, et al. (2005) Education and occupations preceding Parkinson disease: a population-based case-control study. Neurology. 65:1575–1583. Pubmed

35.

Priyadarshi A, Khuder S, Schaub E, Shrivastava S. (2000) A meta-analysis of Parkinson’s disease and exposure to pesticides. Neurotoxicology. 21:435–440. Pubmed

36.

Hancock DB, Martin ER, Mayhew GM, Stajich J, Jewett R, et al. (2008) Pesticide exposure and risk of Parkinson’s disease: a family-based case-control study. BMC Neurology. 8:6. Pubmed

37.

Fahn S, Sulzer D. (2004) Neurodegeneration and neuroprotection in Parkinson disease. Neuro Rx. 1(1):139–154. Pubmed

38.

Sulzer D, Surmeier DJ. (2013) Neuronal vulnerability, pathogenesis, and Parkinson’s disease. Mov Disord. 28(1):41–50. Pubmed

39.

Reglodi D, Renaud J, Tamas A, Tizabi Y, Socias S, et al. (2017) Novel tactics for neuroprotection in Parkinson’s disease: Role of antibiotics, polyphenols and neuropeptides. Prog Neurobiol. 155:120–148. Pubmed

40.

Costa SL, Silva VDA, Souza CdS, Santos C, Paris I, et al. (2016) Impact of plant-derived flavonoids on neurodegenerative diseases. Neurotox Res. 2016;30:41–52. Pubmed

41.

Schapira AH, Jenner P. (2011) Etiology and pathogenesis of Parkinson’s disease. Mov Disord. 26(6):1049–1055. Pubmed

42.

Zhu J, Chu C. (2010) Mitochondrial dysfunction in Parkinson’s disease. J Alzheimers Dis. 20(Suppl 2):S325–S334. Pubmed

43.

Parker WD, Parks JK, Swerdlow RH. (2008) Complex I deficiency in Parkinson’s disease frontal cortex. Brain Res. 16:215–218. Pubmed

44.

Jenner P, Olanow CW. (2006) The pathogenesis of cell death in Parkinson’s disease. Neurol. 66(10 suppl 4):S24–S36. Pubmed

45.

Beal MF. (2005) Mitochondria take center stage in aging and neurodegeneration. Ann Neurol. 58(4):495–505. Pubmed

46.

Roy Sarkar S, Banerjee S. (2019) Gut microbiota in neurodegenerative disorders. J Neuroimmunol. 328:98–104. Pubmed

47.

Kanthasamy AG, Kitazawa M, Kanthasamy A, Anantharam V. (2005) Dieldrin-induced neurotoxicity: relevance to Parkinson’s disease pathogenesis. Neurotoxicology. 26(4):701-719. Pubmed

48.

Tanner CM, Kamel F, Ross GW, Hoppin J, Goldman S, et al. (2011) Rotenone, paraquat and Parkinson’s disease. Environ Health Perspect. 119(6):866-72. Pubmed

49.

Stykel MG, Humphries K, Kirby MP, Czaniecki C, Wang T, et al. (2018) Nitration of microtubules blocks axonal mitochondrial transport in a human pluripotent stem cell model of Parkinson’s disease. The FASEB Journal. 32(10):5354. Pubmed

50.

McGeer PL, Itagaki S, Boyes BE, McGeer EG. (1988) Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 38(8):1285–1291. Pubmed

51.

Chen H, Zhang SM, Hernán MA, Schwarzschild M, Willett W, et al. (2003) Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Arch Neurol. 60(8):1059–1064. Pubmed

52.

Orr CF, Rowe DB, Halliday GM. (2002) An inflammatory review of Parkinson’s disease. Prog Neurobiol. 68(5):325–340. Pubmed

53.

Mogi M, Harada M, Kondo T, Riederer P, Inagaki H, et al. (1994) Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from Parkinsonian patients. Neurosci Lett. 180(2):147–150. Pubmed

54.

Müller T, Blum‐Degen D, Przuntek H, Kuhn W. (1998) Interleukin‐6 levels in cerebrospinal fluid inversely correlate to severity of Parkinson’s disease. Acta Neurol Scand. 98:142–144. Pubmed

55.

McGeer PL, Itagaki S, McGeer EG. (1988) Expression of the histocompatibility glycoprotein HLA-DR in neurological disease. Acta Neuropathol. 76:550–557. Pubmed

56.

Caggiu E, Paulus K, Arru G, Piredda R, Sechi GP, et al. (2016) Humoral cross reactivity between a α-synuclein and Herpes simplex-1 epitope in Parkinson’s disease, a triggering role in the disease? J Neuroimmunol. 291:110–114. Pubmed

57.

Carvey PM, McRae A, Lint TF, Ptak L, Lo E, et al. (1991) The potential use of a dopamine neuron antibody and a striatal-derived neurotrophic factor as diagnostic markers in Parkinson’s disease. Neurology. 41(5 Suppl 2):53–58. Pubmed

58.

Nagatsu T, Mogi M, Ichinose H, Togari A. (2000) Cytokines in Parkinson’s disease. J Neural Transm. 58:143–151. Pubmed

59.

McGeer PL, McGeer EG. (2004) Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 10:S3–S7. Pubmed

60.

Szeto Y, Kwok T, Benzie I. (2004) Effects of long-term vegetarian diet on biomarkers of antioxidant status and cardiovascular disease risk. Nutrition. 20(10):863-6. Pubmed

61.

Haghighatdoost F, Bellissimo N, Totosy de Zepetnek J, Rouhani M. (2017) Association of vegetarian diet with inflammatory biomarkers: a systematic review and meta-analysis of observational studies. Public Health Nutrition. 1-9. Pubmed

62.

Shah B, Newman J, Woolf K, et al. (2018) Anti-Inflammatory Effects of a vegan diet versus the American Heart Association-recommended diet in coronary artery disease trial. J Am Heart Assoc. 7(23):e011367. Pubmed

63.

Magalingam KB, Radhakrishnan AK, Haleagrahara N. (2015) Protective Mechanisms of Flavonoids in Parkinson’s Disease. Oxid Med Cell Longev. 2015:314560. Pubmed

64.

Dias V, Junn E, Mouradian MM. (2013) The role of oxidative stress in Parkinson’s disease. J Parkinsons Dis. 3(4):461-491. Pubmed

65.

Lin MT, Beal MF. (2006) Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 443:787–795. Pubmed

66.

Beal MF. (2003) Bioenergetic approaches for neuroprotection in Parkinson’s disease. Ann Neurol. 53(Suppl 3):S39–S47. Pubmed

67.

Andersen JK. (2004) Oxidative stress in neurodegeneration: Cause or consequence? Nat Med. 10:S18–S25. Pubmed

68.

Johnson WM, Wilson-Delfosse AL, Mieyal JJ. (2012) Dysregulation of glutathione homeostasis in neurodegenerative diseases. Nutrients. 4(10):1399–1440. Pubmed

69.

Dumont M, Beal MF. (2011) Neuroprotective strategies involving ROS in Alzheimer’s disease. Free Radic Biol Med. 51(5):1014–1026. Pubmed

70.

Yan MH, Wang X, Zhu X. (2011) Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic Biol Med. 62:90–101. Pubmed

71.

Richter C, Park JW, Ames BN. (1988) Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc Natl Acad Sci. 85(17):6465–6467. Pubmed

72.

Cadenas E, Davies KJA. (2000) Mitochondrial free radical generation, oxidative stress, and aging. Free Radic Biol Med. 29(3-4):222–230. Pubmed

73.

Lee HC, Chang CM, Chi CW. (2010) Somatic mutations of mitochondrial DNA in aging and cancer progression. Ageing Res Rev. 9(Suppl 1):S47–S58. Pubmed

74.

Gordon M. (2012) Significance of dietary antioxidants for health. Int J Mol Sci. 13(1):173-179. Pubmed

75.

Poe K. (2017) Plant-based diets and phytonutrients:potential health benefits and disease prevention. iMedPub Journals. 9(6):7. Archives of Medicine

76.

McCarty MF, Lerner A. (2020) Low risk of Parkinson’s disease in quasi-vegan cultures may reflect GCN2-mediated upregulation of Parkin. Advan Nutr. nmaa112. Pubmed

77.

Braak H, Sandmann-Keil D, Gai W, Braak E. (1999) Extensive axonal Lewy neurites in Parkinson’s disease: a novel pathological feature revealed by alpha-synuclein immunocytochemistry. Neurosci Lett. 265(1):67-69. Pubmed

78.

Gibb WR, Lees AJ. (1989) The significance of the Lewy body in the diagnosis of idiopathic Parkinson’s disease. Neuropathol Appl Neurobiol. 15(1):27-44. Pubmed

79.

Yang D, Zhao D, Ali Shah S, Wu W, Lai M, et al. (2019) The role of the gut microbiota in the pathogenesis of Parkinson’s disease. Front Neurol. 10:1155. Pubmed

80.

Burré J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, et al. (2010) Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science. 329 (5999):1663-1667. Pubmed

81.

Dikiy I, Eliezer D. (2012) Folding and misfolding of alpha-synuclein on membranes. Biochim Biophys Acta. 1818(4):1013-1018. Pubmed

82.

Keshavarzian A, Green SJ, Engen PA, Voigt R, Naqib A, et al. (2015) Colonic bacterial composition in Parkinson’s disease. Mov Disord. 30(10):1351–1360. Pubmed

83.

Jucker M, Walker LC. (2013) Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature. 501:45-51. Pubmed

84.

Visanji NP, Brooks PL, Hazrati LN, Lang AE. (2013) The prion hypothesis in Parkinson’s disease: braak to the future. Acta Neuropathol Commun. 1:2. Pubmed

85.

Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, et al. (2015) Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 30(3):350-386. Pubmed

86.

Mayer EA. (2011) Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci. 12:453-466. Pubmed

87.

Cryan JF, O’Mahony SM. (2011) The microbiome-gut-brain axis: from bowel to behavior. Neurogastroenterol Motil. 23(3):187-192. Pubmed

88.

De Palma G, Collins SM, Bercik P, Verdu EF. (2014) The microbiota-gut-brain axis in gastrointestinal disorders: stressed bugs, stressed brain or both? J Physiol. 592:2989-2997. Pubmed

89.

Yarandi SS, Peterson DA, Treisman GJ, Moran TH, Pasricha PJ. (2016) Modulatory effects of gut microbiota on the central nervous system: how gut could play a role in neuropsychiatric health and diseases. J Neurogastroenterol Motil. 22(2):201-212. Pubmed

90.

Svensson E, Horváth‐Puhó E, Thomsen RW, Djurhuus JC, Pedersen L, et al. (2015) Vagotomy and subsequent risk of Parkinson’s disease. Ann Neurol. 78(4):522-529. Pubmed

91.

Perez-Pardo P, Kliest T, Dodiya HB, Broersen L, Garssen J, et al. (2017) The gut-brain axis in Parkinson’s disease: Possibilities for food-based therapies. Eur J Pharmacol. 817:86-95. Pubmed

92.

Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, et al. (2015) Intestinal dysbiosis and lowered serum lipopolysaccharide-binding protein in Parkinson’s disease. PLoS One. 10(11):e0142164. Pubmed

93.

Borody T, Torres M, Campbell J, Hills L, Ketheeswaran S. (2009) Treatment of severe constipation improves Parkinson’s disease (PD) symptoms. Am J Gastroenterol. 104:S367. AJG

94.

Hill‐Burns E, Debelius J, Morton J, Wissemann W, Lewis M, et al. (2017) Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 32(5):739-749. Pubmed

95.

Dutta SK, Verma S, Jain V, Surapaneni B, Vinayek R, et al. (2019) Parkinson’s Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J Neurogastroenterol Motil. 25(3):363-376. Pubmed

96.

Devos D, Lebouvier T, Lardeux B, Biraud M, Rouaud T, et al. (2013) Colonic inflammation in Parkinson’s disease. Neurobiol Dis. 50:42-48. Pubmed

97.

Forsyth CB, Shannon KM, Kordower JH, Voigt R, Shaikh M, et al. (2011) Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLOS ONE. 6(12):e28032. Pubmed

98.

Forsythe P, Kunze WA. (2013) Voices from within: gut microbes and the CNS. Cell Mol Life Sci. 70:55-69. Pubmed

99.

Ganapathy V, Thangaraju M, Prasad PD, Martin PM, Singh N. (2013) Transporters and receptors for short-chain fatty acids as the molecular link between colonic bacteria and the host. Curr Opin Pharmacol. 13(6):869-874. Pubmed

100.

Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, et al. (2014) Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 40 (1):128-139. Pubmed

101.

Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. (2010) Mechanisms underlying inflammation in neurodegeneration. Cell. 140 (6):918-934. Pubmed

102.

Quigley E, Quera R. (2006) Small intestinal bacterial overgrowth: roles of antibiotics, prebiotics, and probiotics. Gastroenterology. 130 (2):S78-S90.  Pubmed

103.

Banks WA, Dohgu S, Lynch JL, Fleegal-DeMotta M, Erickson M, et al. (2008) Nitric oxide isoenzymes regulate lipopolysaccharide-enhanced insulin transport across the blood-brain barrier. Endocrinology. 149 (4):1514-1523. Pubmed

104.

Gibson GR, Roberfroid MB. (1995) Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr. 125(6):1401–1412. Pubmed

105.

Gibson G, Hutkins R, Sanders M, Prescott S, Reimer R, et al. (2017) Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. 14:491. Pubmed

106.

Ciorba MA. (2012) A gastroenterologist’s guide to probiotics. Clin Gastroenterol Hepatol. 10(9):960–968. Pubmed

107.

Wong J. (2014) Gut microbiota and cardiometabolic outcomes: influence of dietary patterns and their associated components. Am J Clin Nutr. 100(Suppl 1):369S-77S. Pubmed

108.

Cantu-Jungles TM, Rasmussen HE, Hamaker BR. (2019) Potential of prebiotic butyrogenic fibers in Parkinson’s disease. Front Neurol. 10:663. Pubmed

109.

Tomova A, Bukovsky I, Rembert E, Yonas W, Alwarith J, et al. (2019) The effects of vegetarian and vegan diets on gut microbiota. Front Nutr. 6:47. Pubmed

110.

Unger M, Spiegel J, Dillmann K, Grundmann D, Philippeit H, et al. (2016) Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat Disord. 32:66–72. Pubmed

111.

Spencer JPE. (2008) Flavonoids: modulators of brain function? Br J Nutr. 99(E-S1):ES60–ES77. Pubmed

112.

Manach C, Scalbert A, Morand C, Remesy C, Jimenez L. (2004) Polyphenols: food sources and bioavailability. Am J Clin Nutr. 79(5):727–747. Pubmed

113.

Waterhouse AL, Shirley JR, Donovan JL. (1996) Antioxidants in chocolate. Lancet. 348(9030):834. Pubmed

114.

Ramassamy C. (2006) Emerging role of polyphenolic compounds in the treatment of neurodegenerative diseases: a review of their intracellular targets. Eur J Pharmacol. 545(1):51–64. Pubmed

115.

Meng X, Munishkina L, Fink A, Uversky V. (2009) Molecular mechanisms underlying the flavonoid-induced inhibition of alpha-synuclein fibrillation. Biochemistry. 48(34):8206-8224. Pubmed

116.

Saitoh F, Noma M, Kawashima N. (1985) The alkaloid contents of sixty Nicotiana species. Phytochemistry. 24(3):477–480. Elsevier

117.

Schmeltz I, Hoffmann D. (1977) Nitrogen-containing compounds in tobacco and tobacco smoke. Chem Rev. 77:295–311. ACS

118.

Clark MS, Rand MJ, Vanov S. (1965) Comparison of pharmacological activity of nicotine and related alkaloids occurring in cigarette smoke. Arch Int Pharmacodyn Ther. 156(2):363–379. Pubmed

119.

Zabrodskii P, Gromov M, Maslyakov V. (2015) Effect of alpha7n-acetylcholine receptor activation and antibodies to TNF-alpha on mortality of mice and concentration of Proinflammatory cytokines during early stage of Sepsis. Bull Exp Biol Med. 159(6):740–742. Springer

120.

Barreto G, Iarkov A, Moran V. (2014) Beneficial effects of nicotine, cotinine and its metabolites as potential agents for Parkinson’s disease. Front Aging Neurosci. 6:340. Pubmed

121.

Paris D, Beaulieu-Abdelahad D, Bachmeier C, Reed J, Ait-Ghezala G, et al. (2011) Anatabine lowers Alzheimer’s Aβ production in vitro and in vivo. Eur J Pharmacol. 670(2–3):384–391. Pubmed

122.

Bai A, Guo Y, Lu N. (2007) The effect of the cholinergic anti-inflammatory pathway on experimental colitis. Scand J Immunol. 66(5):538–545. Pubmed

123.

Brody AL, Mandelkern MA, London ED, Olmstead R, Farahi J, et al. (2006) Cigarette smoking saturates brain alpha 4 beta 2 nicotinic acetylcholine receptors. Arch Gen Psychiatry. 63:907–915. Pubmed

124.

Chen H, Huang X, Guo X, Mailman R, Park Y, et al. (2010) Smoking duration, intensity, and risk of Parkinson disease. Neurology. 74:878–884. Pubmed

125.

Kenborg L, Lassen CF, Ritz B, Andersen K, Christensen J, et al. (2015) Lifestyle, family history, and risk of idiopathic Parkinson disease: a large Danish case-control study. Am J Epidemiol. 181:808–816. Pubmed

126.

Siegmund B, Leitner E, Pfannhauser W. (1999) Determination of the nicotine content of various edible nightshades (Solanaceae) and their products and estimation of the associated dietary nicotine intake. J Agric Food Chem. 47:3113–3120. Pubmed

127.

Sheen S. (1988) Detection of nicotine in foods and plant material. J Food Sci. 53:1572–1573. Wiley

128.

Castro A, Monji D. (1986) Dietary nicotine and its significance in studies on tobacco smoking. Biochem Arch. 2:91–97.

129.

Davis RA, Stiles MF, deBethizy JD, Reynolds JH. (1991) Dietary nicotine: a source of urinary cotinine. Food Chem Toxicol. 29:821–827. Pubmed

130.

Domino E, Hornbach E, Demana T. (1993) Relevance of nicotine content of common vegetables to the identification of passive tobacco smokers. Med Sci Res. 21:571–572.

131.

Andersson C, Wennström P, Gry J. (2003) Nicotine alkaloids in Solanaceous food plants. Copenhagen: Nordic Council of Ministers. 2003:531. TemaNord

132.

Liu Y, Hu J, Wu J, Zhu C, Hui Y, et al. (2012) α7 nicotinic acetylcholine receptor-mediated neuroprotection against dopaminergic neuron loss in an MPTP mouse model via inhibition of astrocyte activation. J Neuroinflammation. 9:98. Pubmed

133.

Takeuchi H, Yanagida T, Inden M, Takata K, Kitamura Y, et al. (2009) Nicotinic receptor stimulation protects nigral dopaminergic neurons in rotenone-induced Parkinson’s disease models. J Neurosci Res. 87:576–585. Pubmed

134.

Searles Nielsen S, Gallagher LG, Lundin JI, Longstreth W, Smith-Weller T, et al. (2012) Environmental tobacco smoke and Parkinson’s disease. Mov Disord. 27:293–296. Pubmed

135.

Nielsen SS, Franklin GM, Longstreth Jr WT, Swanson PD, Checkoway H. (2013) Nicotine from edible Solanaceae and risk of Parkinson disease. Ann Neurol. 74(3):472–477. Pubmed

136.

Ma C, Molsberry S, Li Y, Schwarzschild M, Ascherio A, et al. (2020) Dietary nicotine intake and risk of Parkinson disease: a prospective study. Am J Clin Nutr. 112(4):1080-1087. Pubmed

137.

Caturegli P, DeRemigis A, Ferlito M, Landek-Salgado M, Iwama S, et al. (2012) Anatabine ameliorates experimental autoimmune thyroiditis. Endocrinology. 153:4580–4587. Pubmed

138.

Paris D, Beaulieu-Abdelahad D, Abdullah L, Bachmeier C, Ait-Ghezala G, et al. (2013) Anti-inflammatory activity of anatabine via inhibition of STAT3 phosphorylation. Eur J Pharmacol. 698:145–153. Pubmed

139.

Sasahara I, Furuhata Y, Iwasaki Y, Inoue N, Sato H, et al. (2010) Assessment of the biological similarity of three capsaicin analogs (capsinoids) found in non-pungent chili pepper (CH-19 Sweet) fruits. Biosci Biotechnol Biochem. 74:274–278. Pubmed

140.

Kim SR, Lee DY, Chung ES, Oh UT, Kim SU, et al. (2005) Transient receptor potential vanilloid subtype 1 mediates cell death of mesencephalic dopaminergic neurons in vivo and in vitro. J Neurosci. 25:662–671. Pubmed

141.

Park ES, Kim SR, Jin BK. (2012) Transient receptor potential vanilloid subtype 1 contributes to mesencephalic dopaminergic neuronal survival by inhibiting microglia-originated oxidative stress. Brain Res Bull. 89:92–99. Elsevier

142.

Morroni F, Tarozzi A, Sita G, Bolondi C, Moraga J, et al. (2013) Neuroprotective effect of sulforaphane in 6-hydroxydopamine-lesioned mouse model of Parkinson’s disease. Neurotoxicology. 36:63–71. Pubmed

143.

Tarozzi A, Morroni F, Bolondi C, Sita G, Hrelia P, et al. (2012) Neuroprotective effects of erucin against 6-hydroxydopamine-induced oxidative damage in a dopaminergic-like neuroblastoma cell line. Int J Mol Sci. 13(9):10899–10910. Pubmed

144.

Tarozzi A, Angeloni C, Malaguti M, Morroni F, Hrelia S, et al. (2013) Sulforaphane as a potential protective phytochemical against neurodegenerative diseases. Oxid Med Cell Longev. 2013:415078. Pubmed

145.

Jiméanez‐Jiméanez FJ, Mateo D, Giméanez‐Roldan S. (1992) Premorbid smoking, alcohol consumption, and coffee drinking habits in Parkinson’s disease: A case-control study. Mov Disord. 7(4):339–344. Mov Disord

146.

Tan EK, Tan C, Fook-Chong S, Lum S, Chai A, et al. (2003) Dose-dependent protective effect of coffee, tea, and smoking in Parkinson’s disease: A study in ethnic Chinese. J Neurol Sci. 216(1):163–167. Pubmed

147.

Fredholm BB, Bättig K, Holmén J, Nehlig A, Zvartau EE. (1999) Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol Rev. 51(1):83–133. Pubmed

148.

Carpenter B, Lebon G. (2017) Human Adenosine A(2A) Receptor: Molecular Mechanism of Ligand Binding and Activation. Front Pharmacol. 8:898. Pubmed

149.

Kolahdouzan M, Hamadeh MJ. (2017) The neuroprotective effects of caffeine in neurodegenerative diseases. CNS Neurosci Ther. 23:272–290. Pubmed

150.

Chen JF, Xu K, Petzer JP,Staal R, Xu Y, et al. (2001) Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson’s disease. J Neurosci. 21:RC143. Pubmed

151.

Bové J, Serrats J, Mengod G, Cortés R, Tolosa E, et al. (2005) Neuroprotection induced by the adenosine A2A antagonist CSC in the 6-OHDA rat model of parkinsonism: Effect on the activity of striatal output pathways. Exp Brain Res. 165(3):362–374. Pubmed

152.

Kelsey JE, Langelier NA, Oriel BS, Reedy C. (2009) The effects of systemic, intrastriatal, and intrapallidal injections of caffeine and systemic injections of A2A and A1 antagonists on forepaw stepping in the unilateral 6-OHDA-lesioned rat. Psychopharmacology. 201:529–539. Europe PMC

153.

Reyhani-Rad S, Mahmoudi J. (2016) Effect of adenosine A2A receptor antagonists on motor disorders induced by 6-hydroxydopamine in rat. Acta Cir Bras. 31(2):133–137. Pubmed

154.

Schapira A, Olanow C, Greenamyre J, Bezard E. (2014) Slowing of neurodegeneration in Parkinson’s disease and Huntington’s disease: future therapeutic perspectives. Lancet. 384(9942):545–555. Pubmed

155.

Al Dakheel A, Kalia L, Lang A. (2014) Pathogenesis-targeted disease-modifying therapies in Parkinson disease. Neurotherapeutics. 11(1):6–23. Pubmed

156.

Sutachan JJ, Casas Z, Albarracin SL, Stab BR, Samudio I, et al. (2012) Cellular and molecular mechanisms of antioxidants in Parkinson’s disease. Nutr Neurosci. 15(3):120–126. Pubmed

157.

Albarracin SL, Stab B, Casas Z, Sutachan JJ, Samudio I, et al. (2012) Effects of natural antioxidants in neurodegenerative disease. Nutr Neurosci. 15(1):1–9. Pubmed

158.

Song JX, Sze SCW, Ng TB, Lee CK, Leung G, et al. (2012) Anti-Parkinsonian drug discovery from herbal medicines: what have we got from neurotoxic models? J Ethnopharmacol. 139(3):698–711. Pubmed

159.

Takeda A, Nyssen O, Syed A, Jansen E, Bueno-de-Mesquita B, et al. (2014) Vitamin A and carotenoids and the risk of Parkinson’s disease: a systematic review and meta-analysis. Neuroepidemiology. 42(1):25–38. Pubmed

160.

Koppula S, Kumar H, More S, Lim H, Hong S, et al. (2012) Recent updates in redox regulation and free radical scavenging effects by herbal products in experimental models of Parkinson’s disease. Molecules. 17(10):11391–11420. Pubmed

161.

Olanow C, Schapira A, Agid Y. (2003) Causes of cell death and prospects for neuroprotection in Parkinson’s disease. Ann Neurol. 53(suppl 3):1–170. Wiley

162.

Jenner P, Olanow C. (1996) Oxidative stress and the pathogenesis of Parkinson’s disease. Neurology. 47(6 suppl 3):S161–S170. Pubmed

163.

Kujawska M, Jodynis-Liebert J. (2018) Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies. Nutrients. 10(5):642. Pubmed

164.

Singh A, Tripathi P, Yadawa A, Singh S. (2020) Promising polyphenols in Parkinson’s disease therapeutics. Neurochem Res. 45(8):1731-1745. Pubmed

165.

Picone P, Bondi ML, Montana G, Bruno A, Pitarresi G, et al. (2009) Ferulic acid inhibits oxidative stress and cell death induced by Ab oligomers: improved delivery by solid lipid nanoparticles. Free Radic Res. 43(11):1133–1145. Pubmed

166.

Yabe T, Hirahara H, Harada N, Ito N, Nagai T, et al. (2010) Ferulic acid induces neural progenitor cell proliferation in vitro and in vivo. Neuroscience. 165(2):515–524. Pubmed

167.

Jin Y, Yan E, Li X, Fan Y, Zhao Y, et al. (2008) Neuroprotective effect of sodium ferulate and signal transduction mechanisms in the aged rat hippocampus. Acta Pharmacol Sin. 29(12):1399–1408. Pubmed

168.

Mori T, Koyama N, Guillot-Sestier M, Tan J, Town T. (2013) Ferulic acid is a nutraceutical β-secretase modulator that improves behavioral impairment and alzheimer-like pathology in transgenic mice. PLoS One. 8(2):e55774. Pubmed

169.

Sultana R, Ravagna A, Mohmmad-Abdul H, Calabrese V, Butterfield D. (2005) Ferulic acid ethyl ester protects neurons against amyloid beta-peptide (1–42)-induced oxidative stress and neurotoxicity: relationship to antioxidant activity. J Neurochem. 92(4):749–758. Pubmed

170.

Zhang Z, Wei T, Hou J, Li G, Yu S, et al. (2003) Iron-induced oxidative damage and apoptosis in cerebellar granule cells: attenuation by tetramethylpyrazine and ferulic acid. Eur J Pharmacol. 467(1–3):41–47. Pubmed

171.

Koh P. (2013) Ferulic acid prevents cerebral ischemic injury-induced reduction of hippocalcin expression. Synapse. 67(7):390–398. Pubmed

172.

Kanski J, Aksenova M, Stoyanova A, Butterfield D. (2002) Ferulic acid antioxidant protection against hydroxyl and peroxyl radical oxidation in synaptosomal and neuronal cell culture systems in vitro: structure-activity studies. J Nutr Biochem. 13(5):273–281. Pubmed

173.

Scapagnini G, Butterfield D, Colombrita C, Sultana R, Pascale A, et al. (2004) Ethyl ferulate, a lipophilic polyphenol, induces HO-1 and protects rat neurons against oxidative stress. Antioxid Redox Signal. 6(5):811–818. Pubmed

174.

Kim BW, Koppula S, Park SY, Park PJ, Lim JH, et al. (2015) Attenuation of neuroinflammatory responses and behavioral deficits by Ligusticum officinale (Makino) Kitag in stimulated microglia and MPTP-induced mouse model of Parkinson’s disease. J Ethnopharmacol. 164:388–397. Pubmed

175.

Pincus JH, Barry KM. (1987) Plasma levels of amino acids correlate with motor fluctuations in Parkinsonism. Arch Neurol. 44:1006–1009. Pubmed

176.

Bracco F, Malesani R, Saladini M, Battistin L. (1991) Protein redistribution diet and antiparkinsonian response to levodopa. Eur Neurol. 31(2):68–71. Pubmed

177.

Riley D, Lang AE. (1988) Practical application of a low-protein diet for Parkinson’s disease. Neurology. 38(7):1026–1031. Pubmed

178.

Karstaedt PJ, Pincus JH. (1992) Protein redistribution diet remains effective in patients with fluctuating Parkinsonism. Arch Neurol. 49(2):149–151. Pubmed

179.

Astarloa R, Mena M, Sánchez V, de la Vega L, de Yébenes J. (1992) Clinical and pharmacokinetic effects of a diet rich in insoluble fiber on Parkinson disease. Clin Neuropharmacol. 15(5):375–380. Pubmed

180.

Baroni L, Bonetto C, Tessan F, Goldin D, Cenci L, et al. (2011) Pilot dietary study with normoproteic protein-redistributed plant-food diet and motor performance in patients with Parkinson’s disease. Nutr Neurosci. 14(1):1-9. Pubmed

181.

Key TJ, Davey GK, Appleby PN. (1999) Health benefits of a vegetarian diet. Proc Nutr Soc. 58(2):271–275. Pubmed

182.

Taghizadeh M, Tamtaji OR, Dadgostar E, Kakhaki R, Bahmani F, et al. (2017) The effects of omega-3 fatty acids and vitamin E co-supplementation on clinical and metabolic status in patients with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Neurochem Int. 108:183-189. Pubmed

183.

Coimbra CG, Junqueira VBC. (2003) High doses of riboflavin and the elimination of dietary red meat promote the recovery of some motor functions in Parkinson’s disease patients. Braz J Med Biol Res. 36(10):1409-1417. Pubmed

184.

Mischley LK, Allen J, Bradley R. (2012) Coenzyme Q10 deficiency in patients with Parkinson’s disease. J Neurol Sci. 318(1-2):72–75. Pubmed

185.

Seet RCS, Lim ECH, Tan JJH, Quek A, Chow A, et al. (2014) Does high-dose coenzyme Q10 improve oxidative damage and clinical outcomes in Parkinson’s disease? Antioxid Redox Signal. 21(2):211–217. Pubmed

186.

Shults CW, Oakes D, Kieburtz K, Beal MF, Haas R, et al. (2002) Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol. 59(10):1541–1550. Pubmed

187.

Müller T, Büttner T, Gholipour AF, Kuhn W. (2003) Coenzyme Q10 supplementation provides mild symptomatic benefit in patients with Parkinson’s disease. Neurosci Lett. 341(3):201–204. Pubmed

188.

Storch A, Jost WH, Vieregge P, Spiegel J, Greulich W, et al. (2007) Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme Q(10) in Parkinson disease. Arch Neurol. 64(7):938–944. Pubmed

189.

Yoritaka A, Kawajiri S, Yamamoto Y, Nakahara T, Ando M, et al. (2015) Randomized, double-blind, placebo-controlled pilot trial of reduced coenzyme Q10 for Parkinson’s disease. Parkinsonism Relat Disord. 21(8):911–916. Pubmed

190.

Fereshtehnejad S, Postuma R. (2017) Sub types of Parkinson’s disease: what do they tell us about disease progression? Curr Neurol Neurosci Rep. 17(4):34. Pubmed

191.

Thenganatt M, Jankovic J. (2014) Parkinson disease subtypes. JAMA Neurol. 71(4):499-504. Pubmed

192.

Parkinson’s Foundation. Parkinson’s Disease vs. Parkinsonism. Parkinson’s Foundation. Available at: https://www.parkinson.org/pd-library/fact-sheets/parkinsonism-vs-parkinsons-disease.

193.

Kurlan R, Kumari R, Ganihong I. (2016) Dramatic Response of Parkinsonism to a Vegan Diet: Case Report. J Parkinsons Dis Alzheimer Dis. 3(1):2. Avens online

194.

Cereda E, Barichella M, Pedrolli C, Pezzoli G. (2010) Low-protein and protein-redistribution diets for Parkinson’s disease patients with motor fluctuations: a systematic review. Mov Disord. 25(13):2021-2034. Pubmed

195.

Pistollato F, Battino M. (2014) Role of plant-based diets in the prevention and regression of metabolic syndrome and neurodegenerative diseases. Trends in Food Science & Technology. 40(1):62-81. Europe PMC

196.

Van Den Eeden SK, Tanner CM, Bernstein AL, Fross R, Leimpeter A, et al. (2003) Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. Am J Epidemiol. 157(11):1015–1022. Pubmed

197.

Strombom A, Rose S. (2017) The prevention and treatment of Type II Diabetes Mellitus with a plant-based diet. Endocrin Metab Int J. 5(5):00138. Medcrave

198.

Rose S, Strombom A. (2018) A comprehensive review of the prevention and treatment of heart disease with a plant-based diet. J Cardiol & Cardiovas Ther. 12(5):555847. Juniper

199.

Rose S, Strombom A. (2018) A plant-based diet prevents and treats prostate cancer. Canc Therapy & Oncol Int J. 11(3):555813. Juniper

200.

Rose S, Strombom A. (2019) Colorectal Cancer Prevention with a Plant-Based Diet. Canc Therapy & Oncol Int J. 15(2):555906. Juniper

201.

Carlsen MH, Halvorsen BL, Holte K, Bøhn S, Dragland S, et al. (2010) The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 9:3. Pubmed

202.

Siddiqui M, Rast S, Lynn M, Auchus A, Pfeiffer R. (2002) Autonomic dysfunction in Parkinson’s disease: A comprehensive symptom survey. Park Relat Disord. 8:277–284. Pubmed

203.

Savica R, Carlin J, Grossardt B, Bower J, Ahlskog J, et al. (2009) Medical records documentation of constipation preceding Parkinson disease: A case-control study. Neurology. 73(21):1752–1758. Pubmed

204.

Gao X, Chen H, Schwarzschild MA, Ascherio A. (2011) A prospective study of bowel movement frequency and risk of Parkinson’s disease. Am J Epidemiol. 174(5):546–551. Pubmed

205.

Sakakibara R, Odaka T, Uchiyama T, Asahina M, Yamaguchi K, et al. (2003) Colonic transit time and rectoanal videomanometry in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 74(2):268–272. Pubmed

206.

Cheon S, Ha M, Park M, Kim J. (2008) Nonmotor symptoms of Parkinson’s disease: Prevalence and awareness of patients and families. Park Relat Disord. 14:286–290. Pubmed

207.

Martinez‐Martin P, Schapira AHV, Stocchi F, Sethi K, Odin P, et al. (2007) Prevalence of nonmotor symptoms in Parkinson’s disease in an international setting; study using nonmotor symptoms questionnaire in 545 patients. Mov Disord. 22(11):1623–1629. Pubmed

208.

Saleem TZ, Higginson IJ, Chaudhuri KR, Martin A, Burman R, et al. (2013) Symptom prevalence, severity and palliative care needs assessment using the Palliative Outcome Scale: a cross-sectional study of patients with Parkinson’s disease and related neurological conditions. Palliat Med. 27:722–731. Pubmed

209.

Ashraf W, Pfeiffer RF, Park F, Lof J, Quigley EM. (1997) Constipation in Parkinson’s disease: objective assessment and response to psyllium. Mov Disord. 12(6):946–951. Pubmed

210.

Verbaan D, Marinus J, Visser M, van Rooden SM, Stiggelbout AM, et al. (2007) Patient-reported autonomic symptoms in Parkinson disease. Neurology. 69(4):333–341. Pubmed

211.

Lin C, Lin J, Liu Y, Chang C, Wu R. (2014) Risk of Parkinson’s disease following severe constipation: a nationwide population-based cohort study. Parkinsonism Relat Disord. 20(12):1371–1375. Pubmed

212.

Kaye J, Gage H, Kimber A, Storey L, Trend P. (2006) Excess burden of constipation in Parkinson’s disease: a pilot study. Mov Disord. 21(8):1270–1273. Pubmed

213.

Ueki A, Otsuka M. (2004) Lifestyle risks of Parkinson’s disease: Association between decreased water intake and constipation. J Neurol. 251:VII/18–VII/23. Pubmed

214.

Meek PD, Evang SD, Tadrous M, Roux-Lirange D, Triller DM, et al. (2011) Overactive bladder drugs and constipation: a meta-analysis of randomized, placebo-controlled trials. Dig Dis Sci. 56:7–18. Pubmed

215.

Pfeiffer R. (2003) Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 2:107–116. Pubmed

216.

Winge K, Rasmussen D, Werdelin L. (2003) Constipation in neurological diseases. J Neurol Neurosurg Psychiatry. 74:13–19. Pubmed

217.

Cersosimo M, Benarroch E. (2012) Pathological correlates of gastrointestinal dysfunction in Parkinson’s disease. Neurobiol Dis. 46(3):559–564. Pubmed

218.

Berrios G, Campbell C, Politynska B. (1995) Autonomic failure, depression and anxiety in Parkinson’s disease. Br J Psychiatry. 166:789–792. Pubmed

219.

Rahman S, Griffin H, Quinn N, Jahanshahi M. (2008) Quality of life in Parkinson’s disease: the relative importance of the symptoms. Mov Disord. 23:1428–1434. Pubmed

220.

McClurg D, Walker K, Aitchison P, Jamieson K, Dickinson L, et al. (2016) Abdominal massage for the relief of constipation in people with Parkinson’s: A qualitative study. Parkinson’s Disease. 2016:4842090. Hindawi

221.

Knudsen K, Krogh K, Østergaard K, Borghammer P. (2017) Constipation in Parkinson’s disease: Subjective symptoms, objective markers, and new perspectives. Mov Disord. 32:94–105. Pubmed

222.

Fasano A, Visanji N, Liu L, Lang A, Pfeiffer R. (2015) Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 14:1474–4422. Pubmed

223.

Mukherjee A, Biswas A, Das S. (2016) Gut dysfunction in Parkinson’s disease. World J Gastroenterol. 22:5742. Pubmed

 

Share this post