Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-24T18:15:31.477Z Has data issue: false hasContentIssue false

Parkinson's disease: the nutrition perspective

Published online by Cambridge University Press:  04 October 2021

Mícheál Ó Breasail*
Affiliation:
Population Health Sciences, Bristol Medical School, University of Bristol, 1-5 Whiteladies Road, Bristol BS8 1NU, UK
Matthew D. Smith
Affiliation:
Population Health Sciences, Bristol Medical School, University of Bristol, 1-5 Whiteladies Road, Bristol BS8 1NU, UK
Emma Tenison
Affiliation:
Population Health Sciences, Bristol Medical School, University of Bristol, 1-5 Whiteladies Road, Bristol BS8 1NU, UK
Emily J. Henderson
Affiliation:
Population Health Sciences, Bristol Medical School, University of Bristol, 1-5 Whiteladies Road, Bristol BS8 1NU, UK Royal United Hospital Bath NHS Foundation Trust, Combe Park, Bath BA1 3NG, UK
Fiona E. Lithander
Affiliation:
Population Health Sciences, Bristol Medical School, University of Bristol, 1-5 Whiteladies Road, Bristol BS8 1NU, UK
*
*Corresponding author: Mícheál Ó Breasail, email micheal.obreasail@bristol.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease and affects about 1% of the population over the age of 60 years in industrialised countries. The aim of this review is to examine nutrition in PD across three domains: dietary intake and the development of PD; whole body metabolism in PD and the effects of PD symptoms and treatment on nutritional status. In most cases, PD is believed to be caused by a combination of genetic and environmental factors and although there has been much research in the area, evidence suggests that poor dietary intake is not a risk factor for the development of PD. The evidence about body weight changes in both the prodromal and symptomatic phases of PD is inconclusive and is confounded by many factors. Malnutrition in PD has been documented as has sarcopaenia, although the prevalence of the latter remains uncertain due to a lack of consensus in the definition of sarcopaenia. PD symptoms, including those which are gastrointestinal and non-gastrointestinal, are known to adversely affect nutritional status. Similarly, PD treatments can cause nausea, vomiting and constipation, all of which can adversely affect nutritional status. Given that the prevalence of PD will increase as the population ages, it is important to understand the interplay between PD, comorbidities and nutritional status. Further research may contribute to the development of interventional strategies to improve symptoms, augment care and importantly, enhance the quality of life for patients living with this complex neurodegenerative disease.

Type
Conference on Nutrition, health and ageing – translating science into practice
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press on behalf of The Nutrition Society

Parkinson's disease (PD) is the second most common neurodegenerative condition with an estimated prevalence of 1 % in those over 60 years(Reference Parkinson1,Reference de Lau and Breteler2 ). First described in 1817 in ‘An Essay on the Shaking Palsy’, James Parkinson described both the motor and non-motor features of the condition. Parkinsonism is a symptom complex consisting of akinesia, rigidity, tremor and postural instability, the latter of which tends to emerge as the disease progresses. The onset of motor signs is often pre-dated by non-motor symptoms which encompass neuropsychiatric, sleep and autonomic dysfunction(Reference Chaudhuri, Healy and Schapira3). Nutrition is an often overlooked but important factor in the management of the disease; dysfunction is invariably multifactorial in aetiology, different features emerge over the disease course and vary in the degree to which they impact quality of life (Fig. 1).

Fig. 1. Nutrition-related issues faced by patients with Parkinson's disease.

Idiopathic PD is the commonest cause of parkinsonism and is diagnosed clinically, usually with at least two of the four clinical signs in accordance with the UK Queens Square Brain Bank criteria(Reference Hughes, Daniel and Kilford4). Imaging has a supportive role particularly where features such as early falls, lack of response to levodopa, lack of tremor, rapid progression or dysautonomia raise the possibility of an alternative diagnosis(Reference Suchowersky, Reich and Perlmutter5). The mainstay of treatment is focused on compensating for the primary loss of dopamine that results from presynaptic degeneration of dopaminergic cells in the substantia nigra and striatum. Dopaminergic drugs offer a gratifying response, particularly early in the disease. The heterogeneity of the disease is such that the choice of treatment regimen is highly individualised.

Hoehn and Yahr described five levels of clinical disability in PD from stage 1, in which there is unilateral involvement and minimal or no functional impairment, through to stage 5, in which the individual is confined to bed/wheelchair(Reference Hoehn and Yahr6). Although these stages may not correlate with pathophysiological stages, this scale aims to have practical utility and be reproducible when scored by different clinicians. The Movement Disorder Society-Unified Parkinson's Disease Rating Scale, which evaluates motor signs, the impact of motor and non-motor symptoms on daily living and motor complications, is the most widely used clinical rating scale for PD(Reference Goetz, Tilley and Shaftman7). Macmahon and Thomas proposed a scale to describe the clinical course of PD from the ‘diagnostic’ phase; through a relatively stable ‘maintenance’ phase; progressing to a ‘complex’ phase, characterised by motor fluctuations, development of cognitive impairment and appearance of axial symptoms, including falls, freezing of gait and dysphagia; finally reaching a palliative phase(Reference MacMahon and Thomas8). It is now recognised that several symptoms, in particular rapid-eye-movement sleep behaviour disorder, constipation, anosmia and depression, known as premotor symptoms, may precede the onset of motor symptoms by many years, during what is referred to as the ‘prodromal’ phase(Reference Schrag, Horsfall and Walters9,Reference Kaiserova, Grambalova and Kurcova10) .

Pathophysiology and aetiology of Parkinson's disease

PD is characterised pathologically by the loss of dopaminergic neurons in the substantia nigra, thought to be due to the accumulation within neurons of aggregated forms of the protein α-synuclein, known as Lewy bodies. By studying post-mortem brains from individuals with and without prior symptoms of parkinsonism, Braak and colleagues described a sequential progression of neuronal damage throughout the nervous system, categorised into six neuropathological stages: stage 1 consisting of lesions in the dorsal motor nucleus of the glossopharyngeal and vagal nerves, which later progress to reach the brainstem, finally involving the cerebral cortex in stage 6(Reference Braak, Del Tredici and Rüb11). Crucially, the hallmark lesions of PD were noted to develop before the clinical appearance of motor and non-motor dysfunction(Reference Braak, Del Tredici and Rüb11). In a subsequent neuropathological study, Braak and colleagues identified Lewy bodies within the myenteric and submucosal plexuses, the collections of neurons which comprise the enteric nervous system which controls the function of the gastrointestinal (GI) tract(Reference Braak, de Vos and Bohl12). This has led to the suggestion that a neurotropic pathogen, which triggers PD pathology, may enter the central nervous system via a nasal and gastric route; the so-called ‘dual-hit hypothesis’(Reference Hawkes, Del Tredici and Braak13).

A minority of patients with PD have a Mendelian form of the disease, such that the disease is caused by the inheritance of a single causative gene (e.g. SNCA, LRRK2, PARK2 and PINK1) in either an autosomal dominant or recessive pattern(Reference Lill, Roehr and McQueen14). However, such cases of familial parkinsonism are rare and, in most cases, PD is believed to be caused by a combination of genetic and environmental factors. Genome-wide association studies look for variability across the genome to determine whether any identified SNPs occur at a differing frequency between PD cases and controls(Reference Vázquez-Vélez and Zoghbi15). Multiple loci have been potentially implicated in PD(Reference Lill, Roehr and McQueen14); these low penetrance variants, which each have only a minimal impact on risk, may collectively increase an individual's risk(Reference Vázquez-Vélez and Zoghbi15). Similarly, numerous observational studies have examined the impact of environmental factors on the risk of PD, including dietary factors such as alcohol consumption and caffeine intake; exposure to environmental toxins, such as pesticides; biomarkers, such as BMI; drugs; comorbid illness and lifestyle factors including smoking(Reference Bellou, Belbasis and Tzoulaki16). However, apparent associations may not reflect a causal relationship due to potential residual confounding and reverse causation. There may also be interactions between genetic and environmental risk factors such that the impact of any environmental factors is modified by the background genetic risk(Reference Jacobs, Belete and Bestwick17).

In this review, we will examine nutrition in PD across three domains: dietary intake and the development of PD; whole body metabolism in PD including an overview of energy balance and musculoskeletal health and the effects of PD symptoms and treatment on nutritional status.

Dietary intake and the development of Parkinson's disease

Single nutrients

Oxidative stress plays a role in the development of PD(Reference Trist, Hare and Double18Reference Puspita, Chung and Shim20). Antioxidant vitamins such as vitamins A, C, E and β-carotene have established roles in reducing cell damage from free radicals, and there has been much interest in whether higher intake of these nutrients reduces PD risk. Data from the Health Professionals Follow-Up Study and the Nurses' Health Study cohorts suggest lower PD risk for the highest v. the lowest quintile of dietary vitamin E(Reference Zhang, Hernán and Chen21), although at later follow-up, no association was found with vitamins C, E or carotenoids(Reference Hughes, Gao and Kim22). In two Swedish cohorts, higher dietary β-carotene intake was associated with lower PD risk, and there was also an inverse association with vitamin C and E intake but only in women(Reference Yang, Wolk and Håkansson23). Two meta-analyses suggest a neuroprotective effect of dietary vitamin E but not vitamins C and A or β-carotene(Reference Etminan, Gill and Samii24,Reference Takeda, Nyssen and Syed25) , although these predate more recent findings from the Health Professionals Follow-Up Study, the Nurses' Health Study and the Swedish cohort studies. Elevated plasma homocysteine has been described in neurodegenerative disorders(Reference Selhub26,Reference Coppedè27) and evidence suggests that polymorphisms in C1 metabolism may increase PD risk(Reference Murray and Jadavji28). Due to the importance of B vitamins in C1 metabolism(Reference Selhub26), B6, folate and B12 have been investigated prospectively in the context of PD risk(Reference de Lau, Koudstaal and Witteman29,Reference Chen, Zhang and Schwarzschild30) although the findings of these studies have been inconclusive.

The potential association between vitamin D intake and status has been explored with respect to PD risk (Reference Knekt, Kilkkinen and Rissanen31,Reference Larsson, Singleton and Nalls32) and progression(Reference Evatt, Delong and Khazai33,Reference Ding, Dhima and Lockhart34) outwith of its established roles in musculoskeletal health(Reference Hiller, Murchison and Lobb35). The literature in relation to vitamin D and the risk of developing PD has been distorted by several studies(Reference Sato, Honda and Iwamoto36Reference Sato, Honda and Kaji40), which, prior to retraction, influenced widely cited reviews and meta-analyses(Reference Hiller41). A Finnish cohort study reported an inverse relationship between serum 25-hydroxyvitamin D (25(OH)D) and PD risk in those in the highest and lowest quartiles(Reference Knekt, Kilkkinen and Rissanen31). In contrast, a Mendelian randomisation study reported a lack of support for a causal association between low 25(OH)D and risk of PD(Reference Larsson, Singleton and Nalls32). Although there is limited evidence that vitamin D is protective against the development of PD(Reference Fullard and Duda42), low serum 25(OH)D in those with established PD may partially explain poorer musculoskeletal health(Reference Zhang, Zhang and Mao43).

Macronutrients such as dietary fat and its individual fatty acids may influence PD risk, although there is significant heterogeneity in the literature(Reference Chen, Zhang and Hernán44Reference Tan, Methawasin and Tan49). Three meta-analyses, each with a different focus, investigated the relationship between dietary fat and the risk of developing PD(Reference Kamel, Goldman and Umbach50Reference Zhang, Chen and Qiu52). The first, by Kamel and colleagues, reported a negative association between total fat intake and PD risk(Reference Kamel, Goldman and Umbach50). Although they demonstrated the same for dietary MUFA, SFA, PUFA, α-linolenic acid and linoleic acid, associations were greatest for α-linolenic acid and weakest for SFA(Reference Kamel, Goldman and Umbach50). In contrast, Wang and colleagues found no association between total fat intake and PD risk, however, they did report an inverse relationship between PD risk and intake of total PUFA, n-3 PUFA, n-6 PUFA and linoleic acid(Reference Wang, Lin and Wu51). The third study by Zhang and colleagues had a wider scope whereby they examined the relationship between fish and PUFA intake and PD risk in patients with mild-to-severe cognitive impairment(Reference Zhang, Chen and Qiu52). They reported that greater PUFA intake decreased PD risk, although not through the intake of the PUFAs DHA, EPA and α-linolenic acid(Reference Zhang, Chen and Qiu52). In summary, evidence about the intake of total fat and individual fatty acids is inconclusive and further research is needed in this area.

Food and food groups

Epidemiological evidence for the neuroprotective effects of caffeine-containing food and beverages, particularly from coffee and tea, emerged in the early 2000s(Reference Ross, Abbott and Petrovitch53Reference Hu, Bidel and Jousilahti61). Meta-analyses which included several of these cohorts in addition to smaller case–control studies(Reference Tan, Chua and Fook-Chong62Reference Tanaka, Miyake and Fukushima69) have consistently found an inverse association between caffeine intake and PD risk(Reference Liu, Guo and Park70Reference Hong, Chan and Bai72). Hong and colleagues also included PD cohorts and suggested that consuming caffeine may slow the rate of disease progression(Reference Hong, Chan and Bai72Reference Scott, Macleod and Counsell75). Recent case–control studies not included in these meta-analyses further support this inverse relationship between caffeine intake and PD risk(Reference Bakshi, Macklin and Hung76). The novel treatment istradefylline, recently approved for the treatment in PD, acts at the same adenosine A2A receptor as caffeine, which further supports this position(Reference Torti, Vacca and Stocchi77).

Cohort studies report positive associations between dairy intake and the risk of developing PD(Reference Kyrozis, Ghika and Stathopoulos46,Reference Sääksjärvi, Knekt and Lundqvist58,Reference Hughes, Gao and Kim78Reference Park, Ross and Petrovitch81) . In the Health Professionals Follow-Up Study and Nurses' Health Study cohorts, dairy intake was reported to be positively associated with PD; men consuming ≥2⋅9 servings/d had an 80 % increased risk compared to those consuming <1 serving/d(Reference Chen, Zhang and Hernán80). More recent data from these cohorts found total dairy intake was not significantly associated with PD, while low-fat dairy intake was(Reference Hughes, Gao and Kim78), perhaps suggesting that the fat component of dairy foods may not increase PD risk(Reference Hughes, Gao and Kim78). The American Cancer Society's Cancer Prevention Study II found a positive association between dairy consumption in both sexes and PD risk; those in the top quintile had a risk ratio 1⋅6 greater compared to the lowest(Reference Chen, O'Reilly and McCullough79). The Honolulu Heart Program which followed up men over 30 years, found those consuming the most milk (>450 ml/d) had a 2⋅3-fold excess of PD v. non-milk drinkers, and no association was found for calcium from dairy or non-dairy sources(Reference Park, Ross and Petrovitch81). In prospective cohorts in Greece and Finland, associations were also observed between milk consumption and the increased risk of PD(Reference Kyrozis, Ghika and Stathopoulos46,Reference Sääksjärvi, Knekt and Lundqvist58) . In the Finnish cohort, a positive association was reported between reduced-fat dairy products and PD risk(Reference Sääksjärvi, Knekt and Lundqvist58). A meta-analysis including several of these studies(Reference Kyrozis, Ghika and Stathopoulos46,Reference Sääksjärvi, Knekt and Lundqvist58,Reference Chen, O'Reilly and McCullough79Reference Park, Ross and Petrovitch81) , estimated the absolute risk differences were two–four PD cases per 100 000 person-years for every 200 g/d increment in milk intake, and one–three PD cases per 100 000 person-years for every 10 g/d increment in cheese intake(Reference Jiang, Ju and Jiang82). In turn, this was included in two umbrella reviews which considered it as Class III evidence(Reference Bellou, Belbasis and Tzoulaki16) and low by AMSTAR and GRADE scores(Reference Zhang, Chen and Xu83). Despite the positive associations between dairy intake and PD risk, there is currently not sufficient evidence to advise against dairy consumption at a population level.

Dietary patterns

Studies which focus on single nutrients and foods risk missing the synergistic effects at a whole diet level. Dietary patterns with well-established relationships to cardiovascular and metabolic health(Reference Chrysohoou, Panagiotakos and Pitsavos84Reference Sofi, Abbate and Gensini86) have been investigated for their potential neuroprotective properties(Reference Sääksjärvi, Knekt and Lundqvist58,Reference Gu, Brickman and Stern87Reference Anastasiou, Yannakoulia and Kosmidis89) . Although many studies used the healthy eating index and alternate healthy eating index to explore relationships between diet and neurological outcomes, in recent years indices based on the ‘Mediterranean diet’ (MeDi) have generated greatest research interest(Reference Gardener and Caunca90). Data from two US cohorts found that those in the highest alternate healthy eating index and alternate Mediterranean diet score quintiles were 30 and 25 %, respectively, less likely to develop PD v. the lowest(Reference Gao, Chen and Fung88); similar relationships were observed at later follow-up(Reference Molsberry, Bjornevik and Hughes91). However, a Finnish cohort using a modified alternate healthy eating index found no such relationship(Reference Sääksjärvi, Knekt and Lundqvist58). In a Greek population, MeDi adherence was associated with better cognitive performance and lower dementia rates(Reference Anastasiou, Yannakoulia and Kosmidis89), and subsequently it was reported that adherence was associated with fewer prodromal PD symptoms(Reference Maraki, Yannakoulia and Stamelou92). In a cohort of over 47 000 Swedish women, greater adherence to a MeDi at middle age was associated with a lower risk for PD(Reference Yin, Löf and Pedersen93). Studies have examined a combined Mediterranean-Dietary Approaches to Stop Hypertension Diet Intervention for Neurodegenerative Delay(Reference Metcalfe-Roach, Yu and Golz94,Reference Agarwal, Wang and Buchman95) : Agarwal and colleagues found that it appeared to reduce PD risk and slow disease progression whereas neither the MeDi nor Dietary Approaches to Stop Hypertension diets alone achieved this(Reference Agarwal, Wang and Buchman95). Moreover, in a smaller study, Metcalfe-Roach and colleagues found similar findings but in women only, whereas a Greek MeDi was protective in both sexes. There has been speculation that other diets such as the ketogenic, vegetarian and vegan can reduce PD risk although the evidence is limited(Reference Włodarek96). Interest in plant-based diets and PD stems from the protective attributes of the MeDi or other diets rich in fruit and vegetables(Reference Okubo, Miyake and Sasaki97), and the apparent lower prevalence of PD outside of North America and Europe, where animal-derived foods form a smaller proportion of dietary intake.

Whole body metabolism in Parkinson's disease

Impaired nutritional status is a common occurrence in patients once they have established features of PD and weight loss may occur with PD progression, holding implications for treatment, quality of life and mortality. Numerous factors occur that may contribute to this process across the course of the condition, visually conceptualised in Fig. 2.

Fig. 2. Putative representation of the dynamic and interacting factors that impact nutritional status in Parkinson's disease. GI, gastrointestinal; M & T, MacMahon and Thomas(Reference MacMahon and Thomas8); H & Y, Hoehn and Yahr(Reference Hoehn and Yahr6).

Body weight

One way in which nutritional dysfunction can be observed in PD is the variation in body weight that is observed throughout the lifespan of the disease. Conflicting results have been reported in epidemiological studies likely due to the considerable heterogeneity between different populations and varying stages of the condition when studied. The investigation of weight in populations with PD poses a multitude of difficulties with confounding occurring at a population level with various levels of obesity in the country in which the study was undertaken, coupled with confounders from comorbidities such as diabetes, as well as the drug treatment diet regimens. Furthermore, the possible existence of specific disease phenotypes in which weight loss may feature differentially may further complicate the picture(Reference Sharma and Turton98). Weight loss in PD may predispose patients not only to an increased risk of malnutrition, but has been postulated to lead to the worsening of symptoms(Reference Sharma and Turton98).

Changes in body weight may occur as a prodromal feature, before the typical symptoms of PD can be observed in an individual, however there are conflicting results. In a prospective study, Chen and colleagues demonstrated significant weight loss prior to diagnosis in over four hundred patients with PD(Reference Chen, Zhang and Hernán99). They suggested that the average weight of patients with PD was stable until shortly before diagnosis and then declined. However, in a case–control study, Ragonese and colleagues reported that no change in BMI occurred during the period preceding disease onset(Reference Ragonese, D'Amelio and Callari100). Both studies used different methods and assessed two geographically disparate populations some decades apart. When symptomatic individuals with PD have been studied, the literature is also inconclusive. A low BMI in symptomatic individuals with PD, relative to healthy controls, has been demonstrated in a meta-analysis of twelve studies and in over eight hundred patients(Reference van der Marck, Dicke and Uc101). Weight variation over the disease course is of increasing interest and is relevant clinically as the therapeutic window of the drug treatments diminishes over time and therefore dosing needs to be reviewed in light of weight changes. Yong and colleagues found progressive weight loss in PD in a 3-year longitudinal study(Reference Yong, Tan and Ng102), whereas a further cross-sectional study of n = 125 adults with a median PD duration of 6 years reported unintentional weight loss in 38 and 50 % of men and women, respectively(Reference Sheard, Ash and Mellick103). A study of a smaller cohort also suggested that BMI is low early in PD; however, a retrospective chart review was used to collect height and weight data in that instance(Reference Cheshire and Wszolek104). Other studies have shown no evidence of low weight or BMI in PD. Barichella and colleagues assessed BMI in n = 364 Italian men and women with a mean duration of PD of 10⋅6 years and found 65 % of the cohort were overweight(Reference Barichella, Marczewska and Vairo105). Similarly, when compared with controls in a cross-sectional study, overweight and obesity were more common among n = 177 Mexican patients with PD(Reference Morales-Briceño, Cervantes-Arriaga and Rodríguez-Violante106). Where it has been demonstrated, a low BMI in PD has been shown to increase the risk of cognitive decline(Reference Kim, Oh and Lee107), and is a risk factor for mortality, particularly in males(Reference Park, Oeda and Kohsaka108), and has also been correlated with decreased olfaction(Reference Sharma and Turton98). Early weight loss, occurring within the first year, has been associated with excess mortality(Reference Cumming, Macleod and Myint109).

Energy expenditure

Changes in energy expenditure have been investigated in PD. Increased energy expenditure has been demonstrated in a number of studies(Reference Ma, Xiong and Shen110) associated, in particular, with rigidity and the medication ‘off’ state which are periods of time in which symptoms significantly worsen, usually due to wearing off of dopaminergic medication(Reference Ross, Abbott and Petrovitch111,Reference Abbott, Ross and Petrovitch112) . Further studies have shown either no evidence of increased resting energy expenditure or even a lower energy expenditure associated with the decreased level of physical activity that occurs due to disability in PD(Reference Barichella, Cereda and Faierman113,Reference Toth, Fishman and Poehlman114) . Investigating this association is likely confounded by the dynamic nature of ‘off’ and ‘on’ medication states and the influence of varying medication states that can change throughout the course of a day.

The symptom of tremor, which predominantly occurs at rest, is a potential cause of an increase in energy expenditure. This association has seldom been investigated aside from extrapolation based on the observation of weight gain following deep brain stimulation (DBS) surgery(Reference Tuite, Maxwell and Ikramuddin115). In contrast, in studies where DBS has been carried out for the unlinked condition of essential tremor, weight gain has not been shown to be significant, disputing the link to increased energy expenditure due to excess movement from tremor(Reference Strowd, Cartwright and Passmore116). Excess energy can also be spent with the excessive movement associated with dyskinaesia, a fidgeting-like motion occurring due to a paradoxical surfeit of dopamine. This is common in association with levodopa therapy, with the risk worsening throughout the disease course, and is associated with a decreased body weight(Reference Yong, Tan and Ng102). The link of DBS with weight gain may also be a function of this negative effect of dyskinaesia on weight, as one of the primary indications for undertaking DBS is to improve the nature of dyskinaesia, particularly by reducing required levodopa dosing. Additionally, a central metabolic effect may be observed given the close proximity of one DBS target, the subthalamic nucleus, to the hypothalamus(Reference Steinhardt, Münte and Schmid117).

Energy intake

Alterations in energy intake have also been found in PD. Changes in the quality of the diet may be driven by specific cognitive changes pertaining to food selection or as part of an impulse control disorder, the latter of which may be related to dopaminergic medication(Reference Kelly, Baig and Hu118). Compulsive eating has been documented in PD, with one case–control study suggesting that 25 % of patients with PD consumed excessive energy compared with 12 % of healthy controls(Reference de Chazeron, Durif and Lambert119). This can manifest as binge-eating and correlates with the presence of anxiety more strongly than medication type or dose(Reference de Chazeron, Durif and Chereau-Boudet120). Dietary intake in PD may be subtly affected by more than simply over-eating, with several studies suggesting a preference for sweet foods including chocolate, cakes and ice cream, even relative to household controls(Reference Lorefält, Granérus and Unosson121Reference Wolz, Kaminsky and Löhle124). Higher carbohydrate intake, associated with lower protein, folate, magnesium and phosphorus intake has also been observed(Reference Ådén, Carlsson and Poortvliet125). Other studies have examined the intake of food groups and low intake of fruit, vegetables and meat has been observed(Reference Lorefält, Granérus and Unosson121). The dopaminergic system is highly implicated in reward and motivation, and dysfunction likely contributes to the pattern of abnormal eating observed in PD, as suggested empirically by the association of dopamine agonist therapy with overeating. It has been suggested that overeating of rewarding foods that triggers a small dopamine ‘hit’ may be in response to the more generalised hypodopaminergic state that defines PD(Reference Ådén, Carlsson and Poortvliet125).

Metabolic homoeostasis

Evidence for dysfunction of metabolic homoeostasis has been found in PD, in particular pathology affecting the hypothalamus as demonstrated in post-mortem and functional imaging studies(Reference Ma, Xiong and Shen110). Decreased orexin is observed in the cerebrospinal fluid of individuals with PD(Reference Drouot, Moutereau and Lefaucheur126,Reference Chieffi, Carotenuto and Monda127) , although correlation with weight has not been undertaken. Decreased ghrelin is observed in the plasma of patients with PD who experience weight loss. This suggests a degree of neurohormonal dysregulation, with an opposite relationship (an increase) usually observed with weight loss in healthy subjects(Reference Chieffi, Carotenuto and Monda127,Reference Fiszer, Michałowska and Baranowska128) . Decreased dynamic control of ghrelin has been demonstrated at prodromal stages, prompting the question of whether this is an early symptomatic change or causative mechanism(Reference Unger, Möller and Mankel129). Although conflicting results for leptin levels have been published in recent years, a recent meta-analysis suggested that this remains normal(Reference Rahnemayan, Mirghafourvand and Fathalizadeh130). Evidence exists for a role of mitochondrial dysfunction in the aetiology of PD(Reference Park, Davis and Sue131), suggesting that an intrinsic dysfunction of energy handling may exist in affected individuals, with neuronal cells particularly sensitive to abnormalities of cellular respiration.

Malnutrition, muscle and bone

Malnutrition is common in older adults and is prevalent in PD. In a systematic review, Sheard and colleagues assessed the prevalence in over 1100 patients with PD in eleven studies. The mean age of the PD samples ranged from 54 to 75 years and the duration of the disease ranged from 5 to 13 years. Irrespective of whether the patient was hospitalised, measured in a PD clinic or living in the community, the prevalence of malnutrition in PD ranged from 0 to –24 % and the risk of malnutrition from 3 to 60 %(Reference Sheard, Ash and Silburn132). Other studies of either hospitalised patients(Reference Yang, Zhan and Zhang133) or those attending an outpatient clinic(Reference Paul, Singh and Paul134) support this finding, although different methods were used to measure malnutrition across the studies. In addition to being at an increased risk of malnutrition because of the symptoms of PD, the treatment used to manage PD may also increase malnutrition risk(Reference Sheard, Ash and Silburn132).

Malnutrition is a known risk factor for sarcopaenia(Reference Beaudart, Sanchez-Rodriguez and Locquet135Reference Gómez-Gómez and Zapico137), the age-related loss of muscle and strength (Reference Morley, Baumgartner and Roubenoff138,Reference Cruz-Jentoft, Baeyens and Bauer139) , which combined with low serum 25(OH)D in PD may negatively impact muscle strength and balance, increasing falls risk(Reference Hiller, Murchison and Lobb35). Determining the prevalence of sarcopaenia in PD is complicated by a lack of consensus and a multitude of clinical definitions in widespread use. These include The European Working Group on Sarcopenia in Older Persons (EWGSOP-1 and EWGSOP-2)(Reference Cruz-Jentoft, Baeyens and Bauer139,Reference Cruz-Jentoft, Bahat and Bauer140) , Foundation of the National Institutes of Health(Reference Studenski, Peters and Alley141) and International Working Group on Sarcopenia(Reference Fielding, Vellas and Evans142) definitions. Although the European Society for Parenteral and Enteral Nutrition guideline for clinical nutrition in neurology suggests that sarcopaenia risk in PD is low(Reference Burgos, Bretón and Cereda143) they cite only a single study that used a clinical definition of sarcopaenia. This study of Italians with parkinsonian syndromes (n = 364) reported a prevalence of 7 % using the EWGSOP-1 definition(Reference Barichella, Pinelli and Iorio144). Several more recent studies have reported higher rates. A Turkish case–control study (n = 155) used EWGSOP-1 and reported a prevalence of 50 % in PD and 31 % in controls(Reference Ozer, Akın and Gultekin145), whereas a Brazilian study reported a prevalence of 22 % in PD using EWGSOP-2 compared to 55 % with EWGSOP-1(Reference da Luz, Bezerra and Asano146). A recent Malaysian study reported a prevalence of 26 % in PD using EWGSOP-2 and 4 % in controls(Reference Tan, Lim and Yong147), greater than previous Malaysian rates based on The Asian Working Group on Sarcopenia criteria(Reference Chen, Liu and Woo148). Vetrano and colleagues compared the criteria and found the prevalence of sarcopaenia in PD varied between 29 and 41 % in men and 18 and 33 % in women, with poor agreement between EWGSOP-1, Foundation of the National Institutes of Health and IGWS(Reference Vetrano, Pisciotta and Laudisio149).

Although in a young healthy individual, a fall from standing height would not likely result in a fracture, in PD, reduced bone mineral density(Reference Torsney, Noyce and Doherty150Reference Tan, Wang and Zhou152) and a higher frequency of falls increase fracture risk(Reference Zhang, Zhang and Mao43,Reference Hely, Reid and Adena153Reference Lee, Choi and Shin159) . Studies suggest that in PD, osteoporosis and fracture are twice as likely(Reference Torsney, Noyce and Doherty150,Reference Kalilani, Asgharnejad and Palokangas160,Reference Sleeman, Che and Counsell161) , and in women, PD is the strongest single contributor to fracture risk(Reference Dennison, Compston and Flahive162). Patients with PD also appear to have a greater prevalence of hip fracture(Reference Coomber, Alshameeri and Masia163Reference Mühlenfeld, Söhling and Marzi165), which is associated with increased morbidity and mortality(Reference Walker, Chaplin and Hancock166,Reference Crego-Vita, Sanchez-Perez and Gomez-Rico167) . Of particular relevance to bone health, a number of case–control studies have suggested serum 25(OH)D is reduced in PD(Reference Evatt, Delong and Khazai33,Reference Ding, Dhima and Lockhart34,Reference Zhang, Zhang and Mao43,Reference Abou-Raya, Helmii and Abou-Raya168Reference Peterson, Mancini and Horak170) , and those with lower 25(OH)D have a greater frequency of falls(Reference Zhang, Zhang and Mao43). Given the vital role that nutrition plays in musculoskeletal function, further research would be valuable to specifically determine the benefit of dietary interventions on bone health and fracture risk.

The effects of Parkinson's disease symptoms and treatment on nutritional status

Gastrointestinal symptoms

Anosmia and dysgeusia

Anosmia is a frequently reported symptom in PD, often occurring many years before the onset of motor symptoms and affecting up to 90 % of patients(Reference Tarakad and Jankovic171). Ageusia is less commonly reported, however taste is heavily dependent on the sense of smell and the two interlinked may affect up to 27 % of patients with PD(Reference Shah, Deeb and Fernando172). Other symptoms such as xerostomia may contribute to taste abnormalities. A decreased sense of smell and taste is known to negatively influence nutritional status(Reference Andersson and Sidenvall173). On examination of the relationship between anosmia, dysgeusia and body weight, it was found that olfactory and gustatory deficits may negatively influence weight(Reference Masala, Loy and Piras174). In a prospective study of patients with PD, Sharma and Turton found that 39 % of patients were characterised as ‘weight losers’ and 60 % as ‘non-weight losers’, with ‘weight losers’ more likely to be older, female and have more severe olfactory impairment(Reference Sharma and Turton98). Since smell is an important aspect of food appeal, it is possible that altered olfaction can negatively impact dietary intake that may be important in the pathophysiology of PD(Reference Ådén, Carlsson and Poortvliet125). Ådén and colleagues found associations between a lower intake of nutrients such as protein and olfactory function in patients newly diagnosed with PD compared to a control group(Reference Ådén, Carlsson and Poortvliet125). Roos and colleagues suggested that hyposmia, and not hypogeusia, may contribute to weight loss in PD and hence increase the risk of malnutrition. This cross-sectional study found a significant correlation between olfactory function and BMI, but not between gustatory function and BMI(Reference Roos, Oranje and Freriksen175).

Dysphagia and the oral cavity

Dysphagia is common in PD, having an estimated symptomatic prevalence of 35 % based on a meta-analysis of ten studies. The prevalence is further increased to 82 % when considering objective measures of swallowing function(Reference Kalf, de Swart and Bloem176), suggesting that asymptomatic dysphagia is widespread and occurs silently. Symptoms arise due to the disruption of swallowing at all stages of the process including the oral stage (reduced oral bolus control), pharyngeal phase (impaired coordination of pharyngeal structures) and the oeosophageal stage (impaired sphincter relaxation)(Reference Simons177). Hypersalivation or conversely xerostomia that can be linked to levodopa therapy are other factors that may impair the swallowing of food(Reference Umemoto, Fujioka and Iwasa178). Patients with PD also suffer from worse dentition than similarly aged individuals, further impairing their ability to eat, with tooth decay in turn contributed to by xerostomia(Reference Hanaoka and Kashihara179). Orofacial pain phenomena such as burning mouth syndrome occur at an increased prevalence in PD, causing potential distress with eating(Reference Coon and Laughlin180). Facial and lip tremors may also occur, affecting confidence with eating(Reference Ou, Wei and Hou181). Each of these factors can individually or collectively impair dietary intake and have a negative impact on nutritional status. Moreover, difficulties ingesting food may impair the intake of specific micronutrients such as vitamin D and calcium, subsequently increasing the risk of osteoporosis and fracture risk. Swallowing issues can also affect quality of life(Reference Carneiro, das Graças Wanderley de Sales Coriolano and Belo182) which may be directly through embarrassment in social situations, or due to fear induced by choking episodes, as well as the impact that dysphagia can have on dietary intake. The presence of dysphagia is a risk factor for adverse outcomes such as pneumonia through silent aspiration, a primary cause of death associated with PD(Reference Pennington, Snell and Lee183).

Changes to the gut microbiome

Small bowel gastrointestinal overgrowth has been commonly observed in PD, affecting an estimated 46 % of patients and is characterised by an overgrowth of bacterial colonies beyond normally observed limits or the growth of abnormal types(Reference Li, Feng and Jiang184). Diagnosis relies on the use of a hydrogen breath test to infer overgrowth, as used in studies defining prevalence in PD, although the gold standard is the physical culture of jejunal aspirate. The gut microbiome in PD is well documented to be abnormal relative to healthy controls and the overabundance of particular species are associated with the risk of GI symptoms in PD such as constipation(Reference Boulos, Yaghi and El Hayeck185). Vitamin B12 deficiency can result from the consumption of this vitamin by anaerobic micro-organisms(Reference Bures, Cyrany and Kohoutova186) and bacteria can disrupt bile acid which adversely affects the absorption of fat and fat-soluble vitamins(Reference Bures, Cyrany and Kohoutova186). Hasuike and colleagues hypothesised that bacterial overgrowth has various effects via bile acid metabolism in PD(Reference Hasuike, Endo and Koroyasu187). It has been suggested that serum bilirubin increases as bilirubin metabolism declines with decreases in the intestinal bacteria(Reference Hasuike, Endo and Koroyasu187). Simultaneously, bile acid is degraded due to increased intestinal bacteria, and lipid absorption decreases leading to low serum TAG levels and loss of body mass(Reference Hasuike, Endo and Koroyasu187). Similarly, there is decreased absorption of vitamin D aligned with a decrease in bile acid, a risk factor for osteoporosis and fractures. It is clear that consideration needs to be given to the hypothesis that some of the non-motor manifestations accompanying PD are caused by intestinal dysbiosis(Reference Hasuike, Endo and Koroyasu187). Helicobacter pylori (H. pylori) infection has been linked to PD, with higher rates of infection observed PD(Reference Dardiotis, Tsouris and Mentis188). The benefit of eradication therapy has been addressed and is linked to improved outcomes such as UPDRS score and ‘on’ time(Reference Bai and Li189), suggesting that untreated infection may impair levodopa absorption.

Constipation

The most commonly recognised GI symptom in PD is constipation, referring to both difficulty defecating and a decreased frequency of stool, and affects between 20 and 81 % of patients(Reference Sakakibara190). Constipation can be observed as a prodromal symptom predating the onset of defining motor symptoms(Reference Picillo, Palladino and Erro191), and is linked to worse outcomes that include earlier onset of dementia(Reference Camacho, Macleod and Maple-Grødem192). Symptoms arise from the degeneration of brain structures involved in controlling defecatory storage and expulsion, as well as the nerves of the myenteric plexus within the bowel that control motility(Reference Ohlsson and Englund193). A similar effect on motility can also be observed in the upper GI tract, causing delayed gastric emptying(Reference Yu, Ramsey and Norton194) and both can adversely affect quality of life. Bowel motility issues also affect dopaminergic medication absorption(Reference Doi, Sakakibara and Sato195), worsening motor control and cause further impact on quality of life. Constipation is a risk factor for malnutrition(Reference Fávaro-Moreira, Krausch-Hofmann and Matthys196). In a sample of community-dwelling patients with PD, nutritional status was measured using two validated tools: the Subjective Global Assessment and the Patient-Generated Subjective Global Assessment(197). Forty three percent of the cohort reported previous unintentional weight loss following diagnosis, and 15 % were moderately malnourished wherein constipation was one of the symptoms reported to adversely affect dietary intake(Reference Sheard, Ash and Mellick103). Sheard and colleagues assessed nutritional status using the Patient-Generated Subjective Global Assessment in patients with PD awaiting DBS surgery, and of the nutrition impact symptoms listed, 58 % recorded that constipation adversely affected their dietary intake over the previous 2 weeks(Reference Sheard, Ash and Silburn198). In a cross-sectional study of PD in China, constipation was considered to be one of the two most important predictors of nutritional impairment(Reference Wang, Wan and Cheng199). Delayed gastric emptying and the resultant nausea, bloating and early satiety(Reference Yu, Ramsey and Norton194) can adversely affect appetite and in turn dietary intake, and increase the risk of malnutrition in this patient group.

The effect of non-gastrointestinal symptoms on nutritional status

Difficulty with eating

Patients with PD experience a motor disorder that affects manual dexterity and control, therefore leading to problems with the physical act of eating. Difficulty manipulating food on the plate and transporting it to the mouth accurately is observed with the risk of spillage due to tremor(Reference Athlin, Norberg and Axelsson200). Increased upper limb tremor has been associated with a lower energy intake, as has the observation of fewer spoonfuls being taken during a meal by individuals with advanced stage disease(Reference Fagerberg, Klingelhoefer and Bottai201) and represents the direct effect of the cardinal motor features of PD on the risk of malnutrition.

Cognition

In PD, cognitive impairment is a common symptom later in the disease course. Specific decline in frontal lobe function has been shown to correlate with the BMI, fitting with the potential sweet food preference that can be observed in frontal executive dysfunction(Reference De Lucia, Peluso and Esposito202). Cognitive impairment as it progresses becomes a risk factor for becoming dependent on others for food intake(Reference Jung, De Gagne and Lee203). Individuals are less likely to ask for food and less likely to be able to physically prepare or access food at will due to motor disability in PD, especially in an environment that is unfamiliar such as physical care settings. Overall, the prevalence of malnutrition in care homes is high, estimated at approximately 15 % worldwide(Reference Kaiser, Bauer and Rämsch204). This may be impacted by staffing constraints commonly encountered in such environments, and a lack of social interaction may also remove some aspects of reward associated with eating(Reference Liu, Jao and Williams205). Depression is also a common occurrence in PD and may also negatively influence volition to eat with a demonstrated association of weight loss(Reference Reijnders, Ehrt and Weber206,Reference Kim, Chung and Yoo207) .

The effect of treatment on nutritional status

Dopaminergic-based treatments for PD primarily target motor symptoms, but are associated with recognised side-effects (Table 1). Dopaminergic medication, including levodopa, dopamine agonists and catechol-O-methyltransferase inhibitors commonly cause nausea and vomiting which is usually transient during an initial period of acclimatisation, but may adversely affect dietary intake in the short term. Dopamine agonist medications are associated with weight gain in the long term(Reference Wills, Li and Pérez208,Reference Artaud, Lee and Mangone209) , potentially due to an increased incidence of compulsive eating associated with impulse control disorder. Levodopa use has conversely been associated with a decrease in body weight, although historically dopamine agonists have been used in earlier disease and may therefore simply be a function of disease staging. Weight loss is also observed with levodopa intestinal gel-infusions; however, this is also a treatment reserved for late-stage disease(Reference Fabbri, Zibetti and Beccaria210). Dyskinaesia is another common phenomena related to levodopa therapy and is associated with increased weight loss(Reference Yong, Tan and Ng102).

Table 1. Common medications and their effects on nutritional status

MAO, monoamine oxidase type B; NMDA, N-methyl-d-aspartate.

Adapted from BDA Best Practice guidance for dietitians on the nutritional management of Parkinson's, 2021(219).

Medications can also influence the handling of nutrients important for metabolism. As such, raised homocysteine levels are observed in those patients with oral levodopa dosing and decreased vitamins B12, B6 and folate levels associated with intestinal gel use(Reference Muhlack, Kinkel and Herrman211,Reference Taher, Naranian and Poon212) . As described previously, DBS has been found to be consistently associated with weight gain. This may result from an improvement in dyskinaesia, a primary indication for this therapy, thus reducing overall energy expenditure(Reference Balestrino, Baroncini and Fichera213).

Nutritional status is known to affect the response to PD treatments, likely by affecting pharmacokinetic handling. Individuals with lower body weight are at an increased risk of peak-dose dyskinaesia on levodopa, with a greater area under the curve exposure and longer elimination observed, necessitating the consideration of weight and anticipation of ongoing nutritional care when dosing(Reference Arabia, Zappia and Bosco214Reference Zappia, Crescibene and Arabia216). When normalised for body weight and adjusted for age, sex and disease severity, levodopa has been associated with the impaired nutritional status in a dose-dependent manner(Reference Adams, Boschmann and Lobsien217). In addition, a cumulative dose of levodopa has been reported to be associated with micronutrient deficiency through changes in homocysteine, vitamin B6 and B12 levels(Reference Ceravolo, Cossu and Bandettini di Poggio218). Other common medications used in the treatment of PD and the effects that they may have on nutritional status are described in Table 1(219).

Conclusions and future research

We have presented an overview of nutrition in PD in this review and have covered three domains: dietary intake and the development of PD; whole body metabolism in PD, including energy balance and musculoskeletal health and the effects of PD symptoms and treatment on nutritional status. We have highlighted areas where future research on the effect of PD on nutritional status is particularly relevant and important. Determining modifiable risk factors for PD with further research specifically on Mediterranean-style diets in particular, the Mediterranean-Dietary Approaches to Stop Hypertension Diet Intervention for Neurodegenerative Delay diet, could be vital in enhancing our knowledge of the pathophysiology of PD and therefore the development of novel treatments for the condition. Additionally, there is evidence that patients with PD are at risk of malnutrition and a better understanding of the mechanisms behind this is important in developing holistic care to optimise outcomes and quality of life.

Little is known about appetite in PD aside from limited studies that explore a tendency towards certain foods. Additionally, the burden of disability that can affect dexterity, cognitive impairment and the motor impairments that negatively impact the physical act of eating have not yet been fully determined. Tackling this need would enable techniques to be developed to assist with eating and thus improve nutritional status in this patient group. We have highlighted the studies that have sought to characterise patients' weight in the years before the onset of symptoms. Further research will allow us to understand the role of weight change in the pathophysiology of PD, addressing the question of whether weight change is integral to the underlying pathology or merely an additional early symptom.

Much has been discussed about the link between PD and the GI tract, archetyped by the Braak hypothesis(Reference Braak, Del Tredici and Rüb11) although currently only circumstantial evidence exists. The role played by empirically observed phenomena such as small bowel gastrointestinal overgrowth is not yet fully understood, in particular the extent to which this is important in the prodromal phase, nor is the impact of H. pylori infection, where the impact on dyskinaesia is not yet clear. Adequately powered and well-designed randomised controlled trials are required to assess these links.

A better understanding of the role that nutritional factors play in aetiology will further understand and may, in due course, contribute to developing interventional strategies to augment care and improve symptoms for patients living with this complex and multifactorial neurodegenerative disease.

Acknowledgements

We gratefully acknowledge the input of Stephanie Drake who illustrated Fig. 1 on behalf of the authors.

Financial Support

All authors receive salary support from the Gatsby Foundation.

Conflict of Interest

None.

Authorship

The authors were solely responsible for all aspects of preparation of the present review.

Footnotes

The online version of this article has been updated since original publication. A notice detailing the changes has also been published at https://doi.org/10.1017/S0029665122000787.

References

Parkinson, J (2002) An essay on the shaking palsy. 1817. J Neuropsychiatry Clin Neurosci 14, 223236, discussion 222.CrossRefGoogle ScholarPubMed
de Lau, LM & Breteler, MM (2006) Epidemiology of Parkinson's disease. Lancet Neurol 5, 525535.CrossRefGoogle ScholarPubMed
Chaudhuri, KR, Healy, DG & Schapira, AH (2006) Non-motor symptoms of Parkinson's disease: diagnosis and management. Lancet Neurol 5, 235245.CrossRefGoogle ScholarPubMed
Hughes, AJ, Daniel, SE, Kilford, L et al. (1992) Accuracy of clinical diagnosis of idiopathic Parkinson's disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry 55, 181184.CrossRefGoogle ScholarPubMed
Suchowersky, O, Reich, S, Perlmutter, J et al. (2006) Practice parameter: diagnosis and prognosis of new onset Parkinson disease (an evidence-based review) [RETIRED]. Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 66, 968975.CrossRefGoogle Scholar
Hoehn, MM & Yahr, MD (1967) Parkinsonism: onset, progression and mortality. Neurology 17, 427442.CrossRefGoogle ScholarPubMed
Goetz, CG, Tilley, BC, Shaftman, SR et al. (2008) Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord 23, 21292170.CrossRefGoogle ScholarPubMed
MacMahon, DG & Thomas, S (1998) Practical approach to quality of life in Parkinson's disease: the nurse's role. J Neurol 245(Suppl 1), S19S22.CrossRefGoogle ScholarPubMed
Schrag, A, Horsfall, L, Walters, K et al. (2015) Prediagnostic presentations of Parkinson's disease in primary care: a case-control study. Lancet Neurol 14, 5764.CrossRefGoogle ScholarPubMed
Kaiserova, M, Grambalova, Z, Kurcova, S et al. (2021) Premotor Parkinson's disease: overview of clinical symptoms and current diagnostic methods. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 165, 103112.CrossRefGoogle ScholarPubMed
Braak, H, Del Tredici, K, Rüb, U et al. (2003) Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging 24, 197211.CrossRefGoogle ScholarPubMed
Braak, H, de Vos, RA, Bohl, J et al. (2006) Gastric alpha-synuclein immunoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Parkinson's disease-related brain pathology. Neurosci Lett 396, 6772.CrossRefGoogle ScholarPubMed
Hawkes, CH, Del Tredici, K & Braak, H (2007) Parkinson's disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol 33, 599614.CrossRefGoogle ScholarPubMed
Lill, CM, Roehr, JT, McQueen, MB et al. (2012) Comprehensive research synopsis and systematic meta-analyses in Parkinson's disease genetics: the PDGene database. PLoS Genet 8, e1002548.CrossRefGoogle ScholarPubMed
Vázquez-Vélez, GE & Zoghbi, HY (2021) Parkinson's disease genetics and pathophysiology. Annu Rev Neurosci 44, 87108.CrossRefGoogle ScholarPubMed
Bellou, V, Belbasis, L, Tzoulaki, I et al. (2016) Environmental risk factors and Parkinson's disease: an umbrella review of meta-analyses. Parkinsonism Relat Disord 23, 19.CrossRefGoogle ScholarPubMed
Jacobs, BM, Belete, D, Bestwick, J et al. (2020) Parkinson's disease determinants, prediction and gene–environment interactions in the UK Biobank. J Neurol Neurosurg Psychiatry 91, 10461054.CrossRefGoogle ScholarPubMed
Trist, BG, Hare, DJ & Double, KL (2019) Oxidative stress in the aging substantia nigra and the etiology of Parkinson's disease. Aging Cell 18, e13031.CrossRefGoogle ScholarPubMed
Subramaniam, SR & Chesselet, MF (2013) Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Prog Neurobiol 106–107, 1732.CrossRefGoogle ScholarPubMed
Puspita, L, Chung, SY & Shim, JW (2017) Oxidative stress and cellular pathologies in Parkinson's disease. Mol Brain 10, 53.CrossRefGoogle ScholarPubMed
Zhang, SM, Hernán, MA, Chen, H et al. (2002) Intakes of vitamins E and C, carotenoids, vitamin supplements, and PD risk. Neurology 59, 11611169.CrossRefGoogle Scholar
Hughes, KC, Gao, X, Kim, IY et al. (2016) Intake of antioxidant vitamins and risk of Parkinson's disease. Mov Disord 31, 19091914.CrossRefGoogle ScholarPubMed
Yang, F, Wolk, A, Håkansson, N et al. (2017) Dietary antioxidants and risk of Parkinson's disease in two population-based cohorts. Mov Disord 32, 16311636.CrossRefGoogle ScholarPubMed
Etminan, M, Gill, SS & Samii, A (2005) Intake of vitamin E, vitamin C, and carotenoids and the risk of Parkinson's disease: a meta-analysis. Lancet Neurol 4, 362365.CrossRefGoogle ScholarPubMed
Takeda, A, Nyssen, OP, Syed, A et al. (2014) Vitamin A and carotenoids and the risk of Parkinson's disease: a systematic review and meta-analysis. Neuroepidemiology 42, 2538.CrossRefGoogle ScholarPubMed
Selhub, J (2002) Folate, vitamin B12 and vitamin B6 and one carbon metabolism. J Nutr Health Aging 6, 3942.Google ScholarPubMed
Coppedè, F (2021) One-carbon epigenetics and redox biology of neurodegeneration. Free Radic Biol Med 170, 1933.CrossRefGoogle ScholarPubMed
Murray, LK & Jadavji, NM (2019) The role of one-carbon metabolism and homocysteine in Parkinson's disease onset, pathology and mechanisms. Nutr Res Rev 32, 218230.CrossRefGoogle ScholarPubMed
de Lau, LM, Koudstaal, PJ, Witteman, JC et al. (2006) Dietary folate, vitamin B12, and vitamin B6 and the risk of Parkinson disease. Neurology 67, 315318.CrossRefGoogle ScholarPubMed
Chen, H, Zhang, SM, Schwarzschild, MA et al. (2004) Folate intake and risk of Parkinson's disease. Am J Epidemiol 160, 368375.CrossRefGoogle ScholarPubMed
Knekt, P, Kilkkinen, A, Rissanen, H et al. (2010) Serum vitamin D and the risk of Parkinson disease. Arch Neurol 67, 808811.CrossRefGoogle ScholarPubMed
Larsson, SC, Singleton, AB, Nalls, MA et al. (2017) No clear support for a role for vitamin D in Parkinson's disease: a Mendelian randomization study. Mov Disord 32, 12491252.CrossRefGoogle ScholarPubMed
Evatt, ML, Delong, MR, Khazai, N et al. (2008) Prevalence of vitamin D insufficiency in patients with Parkinson disease and Alzheimer disease. Arch Neurol 65, 13481352.CrossRefGoogle ScholarPubMed
Ding, H, Dhima, K, Lockhart, KC et al. (2013) Unrecognized vitamin D3 deficiency is common in Parkinson disease: Harvard biomarker study. Neurology 81, 15311537.CrossRefGoogle ScholarPubMed
Hiller, AL, Murchison, CF, Lobb, BM et al. (2018) A randomized, controlled pilot study of the effects of vitamin D supplementation on balance in Parkinson's disease: does age matter? PLoS One 13, e0203637e0203637.CrossRefGoogle ScholarPubMed
Sato, Y, Honda, Y & Iwamoto, J (2007) Risedronate and ergocalciferol prevent hip fracture in elderly men with Parkinson disease. Neurology 68, 911915.Google ScholarPubMed
Sato Y, Honda Y & Iwamoto J (2016) Erratum: Risedronate and ergocalciferol prevent hip fracture in elderly men with Parkinson disease (Neurology (2007) 68 (911–915) DOI: 10⋅1212/01.wnl.0000257089⋅50476⋅92). Neurology 87, 239.Google Scholar
Sato, Y, Honda, Y, Iwamoto, J et al. (2005) Abnormal bone and calcium metabolism in immobilized Parkinson's disease patients. Mov Disord 20, 15981603.CrossRefGoogle ScholarPubMed
Sato Y, Honda Y, Iwamoto J et al. (2019) Retracted: Abnormal bone and calcium metabolism in immobilized Parkinson's disease patients (Movement Disorders, (2005), 20, 12, (1598–1603), 10.1002/mds.20658). Movement Disorders 34, 156.Google Scholar
Sato, Y, Honda, Y, Kaji, M et al. (2002) RETRACTED: Amelioration of osteoporosis by menatetrenone in elderly female Parkinson's disease patients with vitamin D deficiency. Bone 31, 114118.CrossRefGoogle ScholarPubMed
Hiller, A (2021) Response to the retraction of papers by Yoshihiro Sato – a review of vitamin D and Parkinson's disease. Maturitas 148, 54.CrossRefGoogle Scholar
Fullard, ME & Duda, JE (2020) A review of the relationship between vitamin D and Parkinson disease symptoms. Front Neurol 11, 454454.CrossRefGoogle ScholarPubMed
Zhang, H-J, Zhang, J-R, Mao, C-J et al. (2019) Relationship between 25-hydroxyvitamin D, bone density, and Parkinson's disease symptoms. Acta Neurol Scand 140, 274280.CrossRefGoogle ScholarPubMed
Chen, H, Zhang, SM, Hernán, MA et al. (2003) Dietary intakes of fat and risk of Parkinson's disease. Am J Epidemiol 157, 10071014.CrossRefGoogle ScholarPubMed
Dong, J, Beard, JD, Umbach, DM et al. (2014) Dietary fat intake and risk for Parkinson's disease. Mov Disord 29, 16231630.CrossRefGoogle ScholarPubMed
Kyrozis, A, Ghika, A, Stathopoulos, P et al. (2013) Dietary and lifestyle variables in relation to incidence of Parkinson's disease in Greece. Eur J Epidemiol 28, 6777.CrossRefGoogle Scholar
de Lau, LM, Bornebroek, M, Witteman, JC et al. (2005) Dietary fatty acids and the risk of Parkinson disease: the Rotterdam study. Neurology 64, 20402045.CrossRefGoogle ScholarPubMed
Abbott, RD, Ross, GW, White, LR et al. (2003) Environmental, life-style, and physical precursors of clinical Parkinson's disease: recent findings from the Honolulu-Asia aging study. J Neurol 250(Suppl 3), Iii30Iii39.CrossRefGoogle ScholarPubMed
Tan, LC, Methawasin, K, Tan, EK et al. (2016) Dietary cholesterol, fats and risk of Parkinson's disease in the Singapore Chinese health study. J Neurol Neurosurg Psychiatry 87, 8692.Google ScholarPubMed
Kamel, F, Goldman, SM, Umbach, DM et al. (2014) Dietary fat intake, pesticide use, and Parkinson's disease. Parkinsonism Relat Disord 20, 8287.CrossRefGoogle ScholarPubMed
Wang, A, Lin, Y, Wu, Y et al. (2015) Macronutrients intake and risk of Parkinson's disease: a meta-analysis. Geriatr Gerontol Int 15, 606616.CrossRefGoogle ScholarPubMed
Zhang, Y, Chen, J, Qiu, J et al. (2016) Intakes of fish and polyunsaturated fatty acids and mild-to-severe cognitive impairment risks: a dose-response meta-analysis of 21 cohort studies. Am J Clin Nutr 103, 330340.CrossRefGoogle ScholarPubMed
Ross, GW, Abbott, RD, Petrovitch, H et al. (2000) Association of coffee and caffeine intake with the risk of Parkinson disease. JAMA 283, 26742679.CrossRefGoogle ScholarPubMed
Kim, IY, O'Reilly É, J, Hughes, KC et al. (2017) Differences in Parkinson's disease risk with caffeine intake and postmenopausal hormone use. J Parkinsons Dis 7, 677684.CrossRefGoogle ScholarPubMed
Ascherio, A, Zhang, SM, Hernán, MA et al. (2001) Prospective study of caffeine consumption and risk of Parkinson's disease in men and women. Ann Neurol 50, 5663.CrossRefGoogle ScholarPubMed
Ascherio, A, Weisskopf, MG, O'Reilly, EJ et al. (2004) Coffee consumption, gender, and Parkinson's disease mortality in the cancer prevention study II cohort: the modifying effects of estrogen. Am J Epidemiol 160, 977984.CrossRefGoogle ScholarPubMed
Ascherio, A, Chen, H, Schwarzschild, MA et al. (2003) Caffeine, postmenopausal estrogen, and risk of Parkinson's disease. Neurology 60, 790795.CrossRefGoogle ScholarPubMed
Sääksjärvi, K, Knekt, P, Lundqvist, A 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, 329337.CrossRefGoogle ScholarPubMed
Palacios, N, Gao, X, McCullough, ML et al. (2012) Caffeine and risk of Parkinson's disease in a large cohort of men and women. Mov Disord 27, 12761282.CrossRefGoogle Scholar
Tan, LC, Koh, WP, Yuan, JM et al. (2008) Differential effects of black versus green tea on risk of Parkinson's disease in the Singapore Chinese health study. Am J Epidemiol 167, 553560.CrossRefGoogle ScholarPubMed
Hu, G, Bidel, S, Jousilahti, P et al. (2007) Coffee and tea consumption and the risk of Parkinson's disease. Mov Disord 22, 22422248.CrossRefGoogle ScholarPubMed
Tan, EK, Chua, E, Fook-Chong, SM et al. (2007) Association between caffeine intake and risk of Parkinson's disease among fast and slow metabolizers. Pharmacogenet Genomics 17, 10011005.CrossRefGoogle ScholarPubMed
Paganini-Hill, A (2001) Risk factors for Parkinson's disease: the leisure world cohort study. Neuroepidemiology 20, 118124.CrossRefGoogle ScholarPubMed
Wirdefeldt, K, Gatz, M, Pawitan, Y et al. (2005) Risk and protective factors for Parkinson's disease: a study in Swedish twins. Ann Neurol 57, 2733.CrossRefGoogle ScholarPubMed
Fall, PA, Fredrikson, M, Axelson, O et al. (1999) Nutritional and occupational factors influencing the risk of Parkinson's disease: a case-control study in southeastern Sweden. Mov Disord 14, 2837.3.0.CO;2-O>CrossRefGoogle ScholarPubMed
Benedetti, MD, Bower, JH, Maraganore, DM et al. (2000) Smoking, alcohol, and coffee consumption preceding Parkinson's disease: a case-control study. Neurology 55, 13501358.CrossRefGoogle ScholarPubMed
Checkoway, H, Powers, K, Smith-Weller, T et al. (2002) Parkinson's disease risks associated with cigarette smoking, alcohol consumption, and caffeine intake. Am J Epidemiol 155, 732738.CrossRefGoogle ScholarPubMed
Ragonese, P, Salemi, G, Morgante, L et al. (2003) A case-control study on cigarette, alcohol, and coffee consumption preceding Parkinson's disease. Neuroepidemiology 22, 297304.CrossRefGoogle ScholarPubMed
Tanaka, K, Miyake, Y, Fukushima, W et al. (2011) Intake of Japanese and Chinese teas reduces risk of Parkinson's disease. Parkinsonism Relat Disord 17, 446450.CrossRefGoogle ScholarPubMed
Liu, R, Guo, X, Park, Y et al. (2012) Caffeine intake, smoking, and risk of Parkinson disease in men and women. Am J Epidemiol 175, 12001207.CrossRefGoogle ScholarPubMed
Qi, H & Li, S (2014) Dose–response meta-analysis on coffee, tea and caffeine consumption with risk of Parkinson's disease. Geriatr Gerontol Int 14, 430439.CrossRefGoogle ScholarPubMed
Hong, CT, Chan, L & Bai, C-H (2020) The effect of caffeine on the risk and progression of Parkinson's disease: a meta-analysis. Nutrients 12, 1860.CrossRefGoogle ScholarPubMed
Moccia, M, Erro, R, Picillo, M et al. (2016) Caffeine consumption and the 4-year progression of de novo Parkinson's disease. Parkinsonism Relat Disord 32, 116119.CrossRefGoogle ScholarPubMed
Kandinov, B, Giladi, N & Korczyn, AD (2009) Smoking and tea consumption delay onset of Parkinson's disease. Parkinsonism Relat Disord 15, 4146.CrossRefGoogle ScholarPubMed
Scott, NW, Macleod, AD & Counsell, CE (2016) Motor complications in an incident Parkinson's disease cohort. Eur J Neurol 23, 304312.CrossRefGoogle Scholar
Bakshi, R, Macklin, EA, Hung, AY et al. (2020) Associations of lower caffeine intake and plasma urate levels with idiopathic Parkinson's disease in the Harvard biomarkers study. J Parkinsons Dis 10, 505510.CrossRefGoogle ScholarPubMed
Torti, M, Vacca, L & Stocchi, F (2018) Istradefylline for the treatment of Parkinson's disease: is it a promising strategy? Expert Opin Pharmacother 19, 18211828.CrossRefGoogle ScholarPubMed
Hughes, KC, Gao, X, Kim, IY et al. (2017) Intake of dairy foods and risk of Parkinson disease. Neurology 89, 4652.CrossRefGoogle ScholarPubMed
Chen, H, O'Reilly, E, McCullough, ML et al. (2007) Consumption of dairy products and risk of Parkinson's disease. Am J Epidemiol 165, 9981006.CrossRefGoogle ScholarPubMed
Chen, H, Zhang, SM, Hernán, MA et al. (2002) Diet and Parkinson's disease: a potential role of dairy products in men. Ann Neurol 52, 793801.CrossRefGoogle ScholarPubMed
Park, M, Ross, GW, Petrovitch, H et al. (2005) Consumption of milk and calcium in midlife and the future risk of Parkinson disease. Neurology 64, 10471051.CrossRefGoogle ScholarPubMed
Jiang, W, Ju, C, Jiang, H et al. (2014) Dairy foods intake and risk of Parkinson's disease: a dose-response meta-analysis of prospective cohort studies. Eur J Epidemiol 29, 613619.CrossRefGoogle ScholarPubMed
Zhang, X, Chen, X, Xu, Y et al. (2021) Milk consumption and multiple health outcomes: umbrella review of systematic reviews and meta-analyses in humans. Nutr Metab (Lond) 18, 7.CrossRefGoogle ScholarPubMed
Chrysohoou, C, Panagiotakos, DB, Pitsavos, C et al. (2004) Adherence to the Mediterranean diet attenuates inflammation and coagulation process in healthy adults: the ATTICA study. J Am Coll Cardiol 44, 152158.CrossRefGoogle Scholar
Sofi, F, Cesari, F, Abbate, R et al. (2008) Adherence to Mediterranean diet and health status: meta-analysis. Br Med J 337, a1344.CrossRefGoogle ScholarPubMed
Sofi, F, Abbate, R, Gensini, GF et al. (2010) Accruing evidence on benefits of adherence to the Mediterranean diet on health: an updated systematic review and meta-analysis. Am J Clin Nutr 92, 11891196.CrossRefGoogle Scholar
Gu, Y, Brickman, AM, Stern, Y et al. (2015) Mediterranean diet and brain structure in a multiethnic elderly cohort. Neurology 85, 17441751.CrossRefGoogle Scholar
Gao, X, Chen, H, Fung, TT et al. (2007) Prospective study of dietary pattern and risk of Parkinson disease. Am J Clin Nutr 86, 14861494.CrossRefGoogle ScholarPubMed
Anastasiou, CA, Yannakoulia, M, Kosmidis, MH et al. (2017) Mediterranean diet and cognitive health: initial results from the hellenic longitudinal investigation of ageing and diet. PLoS One 12, e0182048.CrossRefGoogle ScholarPubMed
Gardener, H & Caunca, MR (2018) Mediterranean diet in preventing neurodegenerative diseases. Curr Nutr Rep 7, 1020.CrossRefGoogle ScholarPubMed
Molsberry, S, Bjornevik, K, Hughes, KC et al. (2020) Diet pattern and prodromal features of Parkinson disease. Neurology 95, e2095e2108.CrossRefGoogle ScholarPubMed
Maraki, MI, Yannakoulia, M, Stamelou, M et al. (2019) Mediterranean diet adherence is related to reduced probability of prodromal Parkinson's disease. Mov Disord 34, 4857.CrossRefGoogle ScholarPubMed
Yin, W, Löf, M, Pedersen, NL et al. (2021) Mediterranean dietary pattern at middle age and risk of Parkinson's disease: a Swedish cohort study. Mov Disord 36, 255260.CrossRefGoogle ScholarPubMed
Metcalfe-Roach, A, Yu, AC, Golz, E et al. (2021) MIND and Mediterranean diets associated with later onset of Parkinson's disease. Mov Disord 36, 977984.CrossRefGoogle ScholarPubMed
Agarwal, P, Wang, Y, Buchman, AS et al. (2018) MIND diet associated with reduced incidence and delayed progression of ParkinsonismA in old age. J Nutr Health Aging 22, 12111215.CrossRefGoogle ScholarPubMed
Włodarek, D (2019) Role of ketogenic diets in neurodegenerative diseases (Alzheimer's disease and Parkinson's disease). Nutrients 11, 169.CrossRefGoogle Scholar
Okubo, H, Miyake, Y, Sasaki, S et al. (2012) Dietary patterns and risk of Parkinson's disease: a case-control study in Japan. Eur J Neurol 19, 681688.CrossRefGoogle ScholarPubMed
Sharma, JC & Turton, J (2012) Olfaction, dyskinesia and profile of weight change in Parkinson's disease: identifying neurodegenerative phenotypes. Parkinsonism Relat Disord 18, 964970.CrossRefGoogle ScholarPubMed
Chen, H, Zhang, SM, Hernán, MA et al. (2003) Weight loss in Parkinson's disease. Ann Neurol 53, 676679.CrossRefGoogle ScholarPubMed
Ragonese, P, D'Amelio, M, Callari, G et al. (2008) Body mass index does not change before Parkinson's disease onset. Eur J Neurol 15, 965968.CrossRefGoogle Scholar
van der Marck, MA, Dicke, HC, Uc, EY et al. (2012) Body mass index in Parkinson's disease: a meta-analysis. Parkinsonism Relat Disord 18, 263267.CrossRefGoogle ScholarPubMed
Yong, VW, Tan, YJ, Ng, YD et al. (2020) Progressive and accelerated weight and body fat loss in Parkinson's disease: a three-year prospective longitudinal study. Parkinsonism Relat Disord 77, 2835.CrossRefGoogle ScholarPubMed
Sheard, JM, Ash, S, Mellick, GD et al. (2013) Malnutrition in a sample of community-dwelling people with Parkinson's disease. PLoS One 8, e53290.Google Scholar
Cheshire, WP Jr & Wszolek, ZK (2005) Body mass index is reduced early in Parkinson's disease. Parkinsonism Relat Disord 11, 3538.CrossRefGoogle ScholarPubMed
Barichella, M, Marczewska, A, Vairo, A et al. (2003) Is underweightness still a major problem in Parkinson's disease patients? Eur J Clin Nutr 57, 543547.CrossRefGoogle Scholar
Morales-Briceño, H, Cervantes-Arriaga, A, Rodríguez-Violante, M et al. (2012) Overweight is more prevalent in patients with Parkinson's disease. Arq Neuropsiquiatr 70, 843846.CrossRefGoogle ScholarPubMed
Kim, HJ, Oh, ES, Lee, JH et al. (2012) Relationship between changes of body mass index (BMI) and cognitive decline in Parkinson's disease (PD). Arch Gerontol Geriatr 55, 7072.CrossRefGoogle Scholar
Park, K, Oeda, T, Kohsaka, M et al. (2018) Low body mass index and life prognosis in Parkinson's disease. Parkinsonism Relat Disord 55, 8185.CrossRefGoogle ScholarPubMed
Cumming, K, Macleod, AD, Myint, PK et al. (2017) Early weight loss in parkinsonism predicts poor outcomes: evidence from an incident cohort study. Neurology 89, 22542261.CrossRefGoogle ScholarPubMed
Ma, K, Xiong, N, Shen, Y et al. (2018) Weight loss and malnutrition in patients with Parkinson's disease: current knowledge and future prospects. Front Aging Neurosci 10, 1.CrossRefGoogle ScholarPubMed
Ross, GW, Abbott, RD, Petrovitch, H et al. (2012) Pre-motor features of Parkinson's disease: the Honolulu-Asia aging study experience. Parkinsonism Relat Disord 18, S199S202.CrossRefGoogle ScholarPubMed
Abbott, RD, Ross, GW, Petrovitch, H et al. (2007) Bowel movement frequency in late-life and incidental Lewy bodies. Mov Disord 22, 15811586.CrossRefGoogle ScholarPubMed
Barichella, M, Cereda, E, Faierman, SA et al. (2020) Resting energy expenditure in Parkinson's disease patients under dopaminergic treatment. Nutr Neurosci, 110. doi: 10.1080/1028415X.2020.1745427.Google ScholarPubMed
Toth, MJ, Fishman, PS & Poehlman, ET (1997) Free-living daily energy expenditure in patients with Parkinson's disease. Neurology 48, 8891.CrossRefGoogle ScholarPubMed
Tuite, PJ, Maxwell, RE, Ikramuddin, S et al. (2005) Weight and body mass index in Parkinson's disease patients after deep brain stimulation surgery. Parkinsonism Relat Disord 11, 247252.CrossRefGoogle ScholarPubMed
Strowd, RE, Cartwright, MS, Passmore, LV et al. (2010) Weight change following deep brain stimulation for movement disorders. J Neurol 257, 12931297.CrossRefGoogle ScholarPubMed
Steinhardt, J, Münte, TF, Schmid, SM et al. (2020) A systematic review of body mass gain after deep brain stimulation of the subthalamic nucleus in patients with Parkinson's disease. Obes Rev 21, e12955.CrossRefGoogle ScholarPubMed
Kelly, MJ, Baig, F, Hu, MT et al. (2020) Spectrum of impulse control behaviours in Parkinson's disease: pathophysiology and management. J Neurol Neurosurg Psychiatry 91, 703711.CrossRefGoogle ScholarPubMed
de Chazeron, I, Durif, F, Lambert, C et al. (2021) A case–control study investigating food addiction in Parkinson patients. Sci Rep 11, 10934.CrossRefGoogle ScholarPubMed
de Chazeron, I, Durif, F, Chereau-Boudet, I et al. (2019) Compulsive eating behaviors in Parkinson's disease. Eat Weight Disord 24, 421429.CrossRefGoogle ScholarPubMed
Lorefält, B, Granérus, AK & Unosson, M (2006) Avoidance of solid food in weight losing older patients with Parkinson's disease. J Clin Nurs 15, 14041412.CrossRefGoogle ScholarPubMed
Meyers, C, Amick, MA & Friedman, JH (2010) Ice cream preference in Parkinson's disease. Med Health R I 93, 9192.Google ScholarPubMed
Palavra, NC, Lubomski, M, Flood, VM et al. (2021) Increased added sugar consumption is common in Parkinson's disease. Front Nutr 8, 628845.CrossRefGoogle ScholarPubMed
Wolz, M, Kaminsky, A, Löhle, M et al. (2009) Chocolate consumption is increased in Parkinson's disease. Results from a self-questionnaire study. J Neurol 256, 488492.CrossRefGoogle ScholarPubMed
Ådén, E, Carlsson, M, Poortvliet, E et al. (2011) Dietary intake and olfactory function in patients with newly diagnosed Parkinson's disease: a case-control study. Nutr Neurosci 14, 2531.CrossRefGoogle ScholarPubMed
Drouot, X, Moutereau, S, Lefaucheur, JP et al. (2011) Low level of ventricular CSF orexin-A is not associated with objective sleepiness in PD. Sleep Med 12, 936937.CrossRefGoogle Scholar
Chieffi, S, Carotenuto, M, Monda, V et al. (2017) Orexin system: the key for a healthy life. Front Physiol 8, 357.CrossRefGoogle ScholarPubMed
Fiszer, U, Michałowska, M, Baranowska, B et al. (2010) Leptin and ghrelin concentrations and weight loss in Parkinson's disease. Acta Neurol Scand 121, 230236.CrossRefGoogle ScholarPubMed
Unger, MM, Möller, JC, Mankel, K et al. (2011) Postprandial ghrelin response is reduced in patients with Parkinson's disease and idiopathic REM sleep behaviour disorder: a peripheral biomarker for early Parkinson's disease? J Neurol 258, 982990.CrossRefGoogle ScholarPubMed
Rahnemayan, S, Mirghafourvand, M, Fathalizadeh, A et al. (2021) Leptin levels in patients with Parkinson's disease: a systematic review and meta-analysis. Clin Nutr ESPEN 41, 104109.CrossRefGoogle ScholarPubMed
Park, J-S, Davis, RL & Sue, CM (2018) Mitochondrial dysfunction in Parkinson's disease: new mechanistic insights and therapeutic perspectives. Curr Neurol Neurosci Rep 18, 2121.CrossRefGoogle ScholarPubMed
Sheard, JM, Ash, S, Silburn, PA et al. (2011) Prevalence of malnutrition in Parkinson's disease: a systematic review. Nutr Rev 69, 520532.CrossRefGoogle ScholarPubMed
Yang, T, Zhan, Z, Zhang, L et al. (2020) Prevalence and risk factors for malnutrition in patients With Parkinson's disease. Front Neurol 11, 17.CrossRefGoogle ScholarPubMed
Paul, BS, Singh, T, Paul, G et al. (2019) Prevalence of malnutrition in Parkinson's disease and correlation with gastrointestinal symptoms. Ann Indian Acad Neurol 22, 447452.Google ScholarPubMed
Beaudart, C, Sanchez-Rodriguez, D, Locquet, M et al. (2019) Malnutrition as a strong predictor of the onset of sarcopenia. Nutrients 11, 113.CrossRefGoogle ScholarPubMed
Cruz-Jentoft, AJ, Kiesswetter, E, Drey, M et al. (2017) Nutrition, frailty, and sarcopenia. Aging Clin Exp Res 29, 4348.CrossRefGoogle Scholar
Gómez-Gómez, ME & Zapico, SC (2019) Frailty, cognitive decline, neurodegenerative diseases and nutrition interventions. Int J Mol Sci 20, 118.CrossRefGoogle ScholarPubMed
Morley, JE, Baumgartner, RN, Roubenoff, R et al. (2001) Sarcopenia. J Lab Clin Med 137, 231243.CrossRefGoogle ScholarPubMed
Cruz-Jentoft, AJ, Baeyens, JP, Bauer, JM et al. (2010) Sarcopenia: European consensus on definition and diagnosis: report of the European working group on sarcopenia in older people. Age Ageing 39, 412423.CrossRefGoogle ScholarPubMed
Cruz-Jentoft, AJ, Bahat, G, Bauer, J et al. (2019) Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing 48, 1631.CrossRefGoogle ScholarPubMed
Studenski, SA, Peters, KW, Alley, DE et al. (2014) The FNIH sarcopenia project: rationale, study description, conference recommendations, and final estimates. J Gerontol A Biol Sci Med Sci 69, 547558.CrossRefGoogle ScholarPubMed
Fielding, RA, Vellas, B, Evans, WJ et al. (2011) Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International working group on sarcopenia. J Am Med Dir Assoc 12, 249256.CrossRefGoogle ScholarPubMed
Burgos, R, Bretón, I, Cereda, E et al. (2018) ESPEN guideline clinical nutrition in neurology. Clin Nutr 37, 354396.CrossRefGoogle ScholarPubMed
Barichella, M, Pinelli, G, Iorio, L et al. (2016) Sarcopenia and dynapenia in patients with parkinsonism. J Am Med Dir Assoc 17, 640646.CrossRefGoogle ScholarPubMed
Ozer, FF, Akın, S, Gultekin, M et al. (2020) Sarcopenia, dynapenia, and body composition in Parkinson's disease: are they good predictors of disability?: a case-control study. Neurol Sci 41, 313320.CrossRefGoogle ScholarPubMed
da Luz, MCL, Bezerra, GKA, Asano, AGC et al. (2021) Determinant factors of sarcopenia in individuals with Parkinson's disease. Neurol Sci 42, 979985.CrossRefGoogle ScholarPubMed
Tan, YJ, Lim, SY, Yong, VW et al. (2021) Osteoporosis in Parkinson's disease: relevance of distal radius dual-energy X-ray absorptiometry (DXA) and sarcopenia. J Clin Densitom 24, 351361.CrossRefGoogle ScholarPubMed
Chen, LK, Liu, LK, Woo, J et al. (2014) Sarcopenia in Asia: consensus report of the Asian working group for sarcopenia. J Am Med Dir Assoc 15, 95101.CrossRefGoogle ScholarPubMed
Vetrano, DL, Pisciotta, MS, Laudisio, A et al. (2018) Sarcopenia in Parkinson disease: comparison of different criteria and association with disease severity. J Am Med Dir Assoc 19, 523527.CrossRefGoogle ScholarPubMed
Torsney, KM, Noyce, AJ, Doherty, KM et al. (2014) Bone health in Parkinson's disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 85, 11591166.CrossRefGoogle ScholarPubMed
Liu, B, Chen, G, Yu, Z et al. (2021) Bone mineral density and related scores in Parkinson's disease: a systematic review and meta-analysis. World Neurosurg 146, e1202e1218.CrossRefGoogle ScholarPubMed
Tan, L, Wang, Y, Zhou, L et al. (2014) Parkinson's disease and risk of fracture: a meta-analysis of prospective cohort studies. PLoS One 9, e94379.Google ScholarPubMed
Hely, MA, Reid, WG, Adena, MA et al. (2008) The Sydney multicenter study of Parkinson's disease: the inevitability of dementia at 20 years. Mov Disord 23, 837844.CrossRefGoogle ScholarPubMed
Bhattacharya, RK, Dubinsky, RM, Lai, SM et al. (2012) Is there an increased risk of hip fracture in Parkinson's disease? A nationwide inpatient sample. Mov Disord 27, 14401442.CrossRefGoogle ScholarPubMed
Genever, RW, Downes, TW & Medcalf, P (2005) Fracture rates in Parkinson's disease compared with age- and gender-matched controls: a retrospective cohort study. Age Ageing 34, 2124.CrossRefGoogle ScholarPubMed
Schneider, JL, Fink, HA, Ewing, SK et al. (2008) The association of Parkinson's disease with bone mineral density and fracture in older women. Osteoporosis Int 19, 10931097.CrossRefGoogle ScholarPubMed
Fink, HA, Kuskowski, MA, Taylor, BC et al. (2008) Association of Parkinson's disease with accelerated bone loss, fractures and mortality in older men: the osteoporotic fractures in Men (MrOS) study. Osteoporosis Int 19, 12771282.CrossRefGoogle ScholarPubMed
Schini, M, Vilaca, T, Poku, E et al. (2020) The risk of hip and non-vertebral fractures in patients with Parkinson's disease and parkinsonism: a systematic review and meta-analysis. Bone 132, 115173.CrossRefGoogle ScholarPubMed
Lee, CK, Choi, SK, Shin, DA et al. (2018) Parkinson's disease and the risk of osteoporotic vertebral compression fracture: a nationwide population-based study. Osteoporosis Int 29, 11171124.CrossRefGoogle ScholarPubMed
Kalilani, L, Asgharnejad, M, Palokangas, T et al. (2016) Comparing the incidence of falls/fractures in Parkinson's disease patients in the US population. PLoS One 11, e0161689.CrossRefGoogle ScholarPubMed
Sleeman, I, Che, ZC & Counsell, C (2016) Risk of fracture amongst patients with Parkinson's disease and other forms of Parkinsonism. Parkinsonism Relat Disord 29, 6065.CrossRefGoogle ScholarPubMed
Dennison, EM, Compston, JE, Flahive, J et al. (2012) Effect of co-morbidities on fracture risk: findings from the global longitudinal study of osteoporosis in women (GLOW). Bone 50, 12881293.CrossRefGoogle Scholar
Coomber, R, Alshameeri, Z, Masia, AF et al. (2017) Hip fractures and Parkinson's disease: a case series. Injury 48, 27302735.CrossRefGoogle ScholarPubMed
Chen, YY, Cheng, PY, Wu, SL et al. (2012) Parkinson's disease and risk of hip fracture: an 8-year follow-up study in Taiwan. Parkinsonism Relat Disord 18, 506509.CrossRefGoogle ScholarPubMed
Mühlenfeld, N, Söhling, N, Marzi, I et al. (2021) Fractures in Parkinson's disease: injury patterns, hospitalization, and therapeutic aspects. Eur J Trauma Emerg Surg 47, 573580.CrossRefGoogle ScholarPubMed
Walker, RW, Chaplin, A, Hancock, RL et al. (2013) Hip fractures in people with idiopathic Parkinson's disease: incidence and outcomes. Mov Disord 28, 334340.CrossRefGoogle ScholarPubMed
Crego-Vita, D, Sanchez-Perez, C, Gomez-Rico, JA et al. (2017) Intracapsular hip fractures in the elderly. Do we know what is important? Injury 48, 695700.CrossRefGoogle ScholarPubMed
Abou-Raya, S, Helmii, M & Abou-Raya, A (2009) Bone and mineral metabolism in older adults with Parkinson's disease. Age Ageing 38, 675680.CrossRefGoogle ScholarPubMed
van den Bos, F, Speelman, AD, van Nimwegen, M et al. (2013) Bone mineral density and vitamin D status in Parkinson's disease patients. J Neurol 260, 754760.Google ScholarPubMed
Peterson, AL, Mancini, M & Horak, FB (2013) The relationship between balance control and vitamin D in Parkinson's disease-a pilot study. Mov Disord 28, 11331137.CrossRefGoogle ScholarPubMed
Tarakad, A & Jankovic, J (2017) Anosmia and ageusia in Parkinson's disease. Int Rev Neurobiol 133, 541556.CrossRefGoogle ScholarPubMed
Shah, M, Deeb, J, Fernando, M et al. (2009) Abnormality of taste and smell in Parkinson's disease. Parkinsonism Relat Disord 15, 232237.CrossRefGoogle ScholarPubMed
Andersson, I & Sidenvall, B (2001) Case studies of food shopping, cooking and eating habits in older women with Parkinson's disease. J Adv Nurs 35, 6978.CrossRefGoogle ScholarPubMed
Masala, C, Loy, F, Piras, R et al. (2020) Effect of olfactory and gustatory dysfunction and motor symptoms on body weight in patients with Parkinson's disease. Brain Sci 10, 218.CrossRefGoogle ScholarPubMed
Roos, DS, Oranje, OJM, Freriksen, AFD et al. (2018) Flavor perception and the risk of malnutrition in patients with Parkinson's disease. J Neural Transm 125, 925930.CrossRefGoogle ScholarPubMed
Kalf, JG, de Swart, BJ, Bloem, BR et al. (2012) Prevalence of oropharyngeal dysphagia in Parkinson's disease: a meta-analysis. Parkinsonism Relat Disord 18, 311315.CrossRefGoogle ScholarPubMed
Simons, JA (2017) Swallowing dysfunctions in Parkinson's disease. Int Rev Neurobiol 134, 12071238.CrossRefGoogle ScholarPubMed
Umemoto, G, Fujioka, S, Iwasa, Y et al. (2021) Impact of progression of Parkinson's disease on swallowing ability and oral environment. Parkinsons Dis 2021, 55715565571556.Google ScholarPubMed
Hanaoka, A & Kashihara, K (2009) Increased frequencies of caries, periodontal disease and tooth loss in patients with Parkinson's disease. J Clin Neurosci 16, 12791282.CrossRefGoogle ScholarPubMed
Coon, EA & Laughlin, RS (2012) Burning mouth syndrome in Parkinson's disease: dopamine as cure or cause? J Headache Pain 13, 255257.CrossRefGoogle ScholarPubMed
Ou, R, Wei, Q, Hou, Y et al. (2021) Facial tremor in patients with Parkinson's disease: prevalence, determinants and impacts on disease progression. BMC Neurol 21, 86.CrossRefGoogle ScholarPubMed
Carneiro, D, das Graças Wanderley de Sales Coriolano, M, Belo, LR et al. (2014) Quality of life related to swallowing in Parkinson's disease. Dysphagia 29, 578582.CrossRefGoogle ScholarPubMed
Pennington, S, Snell, K, Lee, M et al. (2010) The cause of death in idiopathic Parkinson's disease. Parkinsonism Relat Disord 16, 434437.CrossRefGoogle ScholarPubMed
Li, X, Feng, X, Jiang, Z et al. (2021) Association of small intestinal bacterial overgrowth with Parkinson's disease: a systematic review and meta-analysis. Gut Pathog 13, 25.CrossRefGoogle ScholarPubMed
Boulos, C, Yaghi, N, El Hayeck, R et al. (2019) Nutritional risk factors, Microbiota and Parkinson's disease: what is the current evidence? Nutrients 11, 124.CrossRefGoogle ScholarPubMed
Bures, J, Cyrany, J, Kohoutova, D et al. (2010) Small intestinal bacterial overgrowth syndrome. World J Gastroenterol 16, 29782990.CrossRefGoogle ScholarPubMed
Hasuike, Y, Endo, T, Koroyasu, M et al. (2020) Bile acid abnormality induced by intestinal dysbiosis might explain lipid metabolism in Parkinson's disease. Med Hypotheses 134, 109436.CrossRefGoogle ScholarPubMed
Dardiotis, E, Tsouris, Z, Mentis, AA et al. (2018) H. pylori and Parkinson's disease: meta-analyses including clinical severity. Clin Neurol Neurosurg 175, 1624.CrossRefGoogle ScholarPubMed
Bai, F & Li, X (2021) Association of Helicobacter pylori treatment with Parkinsonism and related disorders: a systematic review and meta-analysis. Life Sci 281, 119767.CrossRefGoogle ScholarPubMed
Sakakibara, R (2021) Gastrointestinal dysfunction in movement disorders. Neurol Sci 42, 13551365.CrossRefGoogle ScholarPubMed
Picillo, M, Palladino, R, Erro, R et al. (2021) The PRIAMO study: age- and sex-related relationship between prodromal constipation and disease phenotype in early Parkinson's disease. J Neurol 268, 448454.CrossRefGoogle ScholarPubMed
Camacho, M, Macleod, AD, Maple-Grødem, J et al. (2021) Early constipation predicts faster dementia onset in Parkinson's disease. NPJ Parkinsons Dis 7, 45.CrossRefGoogle ScholarPubMed
Ohlsson, B & Englund, E (2019) Atrophic myenteric and submucosal neurons are observed in Parkinson's disease. Parkinsons Dis 2019, 7935820.Google ScholarPubMed
Yu, D, Ramsey, FV, Norton, WF et al. (2017) The burdens, concerns, and quality of life of patients with gastroparesis. Dig Dis Sci 62, 879893.CrossRefGoogle ScholarPubMed
Doi, H, Sakakibara, R, Sato, M et al. (2012) Plasma levodopa peak delay and impaired gastric emptying in Parkinson's disease. J Neurol Sci 319, 8688.CrossRefGoogle ScholarPubMed
Fávaro-Moreira, NC, Krausch-Hofmann, S, Matthys, C et al. (2016) Risk factors for malnutrition in older adults: a systematic review of the literature based on longitudinal data. Adv Nutr 7, 507522.CrossRefGoogle ScholarPubMed
Evidence based practice guidelines for the nutritional management of malnutrition in adult patients across the continuum of care (2019). Nutr Diet 66, S1S34.CrossRefGoogle Scholar
Sheard, JM, Ash, S, Silburn, PA et al. (2013) Nutritional status in Parkinson's disease patients undergoing deep brain stimulation surgery: a pilot study. J Nutr Health Aging 17, 148151.CrossRefGoogle ScholarPubMed
Wang, G, Wan, Y, Cheng, Q et al. (2010) Malnutrition and associated factors in Chinese patients with Parkinson's disease: results from a pilot investigation. Parkinsonism Relat Disord 16, 119123.CrossRefGoogle ScholarPubMed
Athlin, E, Norberg, A, Axelsson, K et al. (1989) Aberrant eating behavior in elderly parkinsonian patients with and without dementia: analysis of video-recorded meals. Res Nurs Health 12, 4151.CrossRefGoogle ScholarPubMed
Fagerberg, P, Klingelhoefer, L, Bottai, M et al. (2020) Lower energy intake among advanced vs. early Parkinson's disease patients and healthy controls in a clinical lunch setting: a cross-sectional study. Nutrients 12, 2109.CrossRefGoogle Scholar
De Lucia, N, Peluso, S, Esposito, M et al. (2020) Frontal defect contribution to decreasing of body mass index in Parkinson's disease patients. J Clin Neurosci 72, 229232.CrossRefGoogle ScholarPubMed
Jung, D, De Gagne, JC, Lee, H et al. (2021) Factors associated with eating performance in older adults with dementia in long-term care facilities: a cross-sectional study. BMC Geriatr 21, 365.CrossRefGoogle ScholarPubMed
Kaiser, MJ, Bauer, JM, Rämsch, C et al. (2010) Frequency of malnutrition in older adults: a multinational perspective using the mini nutritional assessment. J Am Geriatr Soc 58, 17341738.CrossRefGoogle ScholarPubMed
Liu, W, Jao, YL & Williams, K (2017) The association of eating performance and environmental stimulation among older adults with dementia in nursing homes: a secondary analysis. Int J Nurs Stud 71, 7079.CrossRefGoogle ScholarPubMed
Reijnders, JS, Ehrt, U, Weber, WE et al. (2008) A systematic review of prevalence studies of depression in Parkinson's disease. Mov Disord 23, 183189, quiz 313.CrossRefGoogle ScholarPubMed
Kim, SR, Chung, SJ & Yoo, SH (2016) Factors contributing to malnutrition in patients with Parkinson's disease. Int J Nurs Pract 22, 129137.CrossRefGoogle ScholarPubMed
Wills, AM, Li, R, Pérez, A et al. (2017) Predictors of weight loss in early treated Parkinson's disease from the NET-PD LS-1 cohort. J Neurol 264, 17461753.CrossRefGoogle ScholarPubMed
Artaud, F, Lee, PC, Mangone, G et al. (2020) Longitudinal association between dopamine agonists and weight in Parkinson's disease. Parkinsonism Relat Disord 80, 158164.CrossRefGoogle ScholarPubMed
Fabbri, M, Zibetti, M, Beccaria, L et al. (2019) Levodopa/carbidopa intestinal gel infusion and weight loss in Parkinson's disease. Eur J Neurol 26, 490496.CrossRefGoogle ScholarPubMed
Muhlack, S, Kinkel, M, Herrman, L et al. (2017) Levodopa, placebo and rotigotine change biomarker levels for oxidative stress. Neurol Res 39, 381386.CrossRefGoogle ScholarPubMed
Taher, J, Naranian, T, Poon, YY et al. (2021) Vitamins and infusion of levodopa-carbidopa intestinal gel. Can J Neurol Sci, 110. doi: 10.1017/cjn.2021.78.Google ScholarPubMed
Balestrino, R, Baroncini, D, Fichera, M et al. (2017) Weight gain after subthalamic nucleus deep brain stimulation in Parkinson's disease is influenced by dyskinesias' reduction and electrodes' position. Neurol Sci 38, 21232129.CrossRefGoogle ScholarPubMed
Arabia, G, Zappia, M, Bosco, D et al. (2002) Body weight, levodopa pharmacokinetics and dyskinesia in Parkinson's disease. Neurol Sci 23(Suppl 2), S53S54.CrossRefGoogle ScholarPubMed
Sharma, JC, Macnamara, L, Hasoon, M et al. (2006) Cascade of levodopa dose and weight-related dyskinesia in Parkinson's disease (LD-WD-PD cascade). Parkinsonism Relat Disord 12, 499505.CrossRefGoogle Scholar
Zappia, M, Crescibene, L, Arabia, G et al. (2002) Body weight influences pharmacokinetics of levodopa in Parkinson's disease. Clin Neuropharmacol 25, 7982.CrossRefGoogle ScholarPubMed
Adams, F, Boschmann, M, Lobsien, E et al. (2008) Influences of levodopa on adipose tissue and skeletal muscle metabolism in patients with idiopathic Parkinson's disease. Eur J Clin Pharmacol 64, 863870.CrossRefGoogle ScholarPubMed
Ceravolo, R, Cossu, G, Bandettini di Poggio, M et al. (2013) Neuropathy and levodopa in Parkinson's disease: evidence from a multicenter study. Mov Disord 28, 13911397.CrossRefGoogle ScholarPubMed
British Dietetic Association. Best practice guidance for dietitians on the nutritional management of Parkinson's.Google Scholar
Figure 0

Fig. 1. Nutrition-related issues faced by patients with Parkinson's disease.

Figure 1

Fig. 2. Putative representation of the dynamic and interacting factors that impact nutritional status in Parkinson's disease. GI, gastrointestinal; M & T, MacMahon and Thomas(8); H & Y, Hoehn and Yahr(6).

Figure 2

Table 1. Common medications and their effects on nutritional status