Neurology International Open 2017; 01(01): E28-E35
DOI: 10.1055/s-0043-102389
Original Article
© Georg Thieme Verlag KG Stuttgart · New York

Hypothesis of Ascension in Idiopathic Parkinson’s Disease

L. Klingelhoefer
1   Department of Neurology, Technical University Dresden, Dresden
,
H. Reichmann
1   Department of Neurology, Technical University Dresden, Dresden
› Author Affiliations
Further Information

Correspondence

Dr. med. Lisa Klingelhoefer
Department of Neurology
Technical University Dresden
Fetscherstraße 74
01307 Dresden

Publication History

Publication Date:
06 April 2017 (online)

 

Abstract

Different clinical stages are observed in idiopathic Parkinson’s disease (PD). Non-motor symptoms define in particular the prodromal period of PD whereas primary motor symptoms such as bradykinesia with rigidity, resting tremor or postural instability are mandatory for the diagnosis of PD. Important non-motor symptoms are olfactory dysfunction, constipation, depression and sleep disturbances. Corresponding to the clinical course of PD, the Braak staging system postulates that the neuropathological process of PD starts in the enteric nervous system (ENS) of the gut and in the olfactory bulb. From there, Parkinson pathology spreads by transsynaptic cell-to-cell transfer via the sympathetic and parasympathetic nervous system in a rostrocranial direction. If the central nervous system is reached, typical neuropathological changes of PD with selective degeneration of dopaminergic neurons of the Substantia nigra pars compacta, the formation of Lewy bodies, reactive gliosis and progressive central neurodegeneration appear. Evidence of clinical, pathological and animal studies supporting these hypotheses are summarised in this review article. α-synuclein as PD-specific pathology was found in the olfactory bulb, the ENS, the submandibular gland, the intermediolateral nucleus of the spinal cord and the dorsal motor nucleus of the vagus nerve. In an animal model, in which mice are treated with the pesticide rotenone chronically and intragastrically, we could almost completely reproduce the typical pathological and clinical features of PD as well as their development in a chronological and regional sequence.


#

Introduction

Patients with idiopathic Parkinson’s disease (PD) show motor and non-motor disease-related symptoms. Although this is globally accepted, the diagnosis of PD according to the UK Brain Bank criteria [1] can only be made if the combination of the motor cardinal symptoms of bradykinesia with at least one other symptom, namely muscular rigidity, rest tremor or postural instability, are clinically evident. There are first approaches to diagnosis that include non-motor complaints occurring even before the motor symptoms. The Movement Disorders Society (MDS) has established new clinical diagnostic criteria for PD. While motor symptoms continue to be regarded as constituting the core of the disease, non-motor symptoms have also been included as important criteria [2]. Meanwhile, the proposed scientific criteria of the MDS for prodromal PD are based primarily on non-motor symptoms [3]. Prodromal PD is the disease phase in which early signs of Parkinson’s-specific neurodegeneration are detectable, while the diagnosis of PD cannot be made as motor symptoms are not yet fully developed. Thus, hyposmia [4] [5], sleep disturbances, in particular REM (rapid eye movement) sleep disturbance disorder [6] [7], autonomous dysfunctions, in particular constipation [8] [9], and psychiatric symptoms such as depression and anxiety disorders [10] [11] occur many years before the appearance of the first motor symptoms. This correlates well with the presumed and partially confirmed hypotheses of ascension of the underlying neurodegenerative changes. There is evidence that, in the context of the gut-brain connection via the vagal nerve of the enteric nervous system (ENS) and additionally, by means of the olfactory bulb, ascension of the Parkinson-specific pathology occurs in the form of pathological α-synuclein. On reaching the central nervous system (CNS), the main features of the Parkinsonian pathology are initiated, namely selective degeneration of the dopaminergic neurons in the substantia nigra pars compacta, formation of Lewy bodies, reactive gliosis and progressive neurodegeneration. The underlying clinical, pathological, and animal studies that support these hypotheses are presented in this review article.


#

Gastrointestinal Symptoms and Olfactory Disorder as Prodromal Non-motor Symptoms of Idiopathic Parkinson’s Disease

On average, one-third of all patients report having gastrointestinal symptoms; these occur in all phases of PD [12] [13] [14]. With a prevalence rate of 28–80%, constipation is one of the most common non-motor complaints in PD [15] [16] [17] [18] [19], and up to 6 times more common in patients with PD compared to healthy controls of comparable age and sex [8] [15] [20] [21]. Studies have also shown that constipation precedes the development of PD and is a major risk factor for this disease [8] [22] [23] [24]. In the Honolulu Heart program, 6 790 men (without PD at the time of inclusion in the study) were controlled for up to 24 years regarding the incidence of PD [22]. After an average of 12 years, 96 men developed PD. After controlling for various factors that can affect constipation, such as age, nicotine and caffeine consumption, men who did not have even one bowel movement a day had a 2.7-fold, 4.1-fold, and 4.5-fold higher risk of developing Parkinsonian syndrome than men who had one, 2 or more than 2 bowel movements a day. In another case-control study, it was shown that the incidence of constipation was higher in people who developed PD than in those who did not (incidence risk rate: 2.01) and this already 10 years before the diagnosis of PD was made [11]. A recent study using positron emission tomography showed a significantly lower concentration of acethycholinesterase in the small intestine of patients with PD compared to healthy control subjects [25]. This finding, however, did not correlate with the severity of constipation. Thus, constipation is not only an early non-motor manifestation, but at the same time also a risk factor of PD. The cause is thought to be neurodegeneration of the ENS and also CNS degeneration in the advanced stage of the disease [26]. It is also known that patients with PD have a markedly reduced olfactory function compared to controls and that this affects both the perception of threshold odours and the discrimination and identification of odours [21] [27]. This olfactory disorder is detectable in up to 90% of patients with PD and is also a prodromal non-motor symptom of PD [28] [29].


#

Pathological Progression of Parkinson’s Disease

The pathognomic histology of PD is Lewy bodies in neurons, Lewy neurites in dendrites and axons, loss of catecholaminergic neurons in the locus coeruleus, and loss of dopaminergic neurons in the substantia nigra. Lewy bodies and Lewy neurites are intracytoplasmatic protein aggregates, which consist mainly of α-synuclein [30]. Pathological studies suggest that there is a specific temporal and spatial pattern in the spread of neuropathological changes in PD [31] [32]. Braak and colleagues developed a system for classifying the degree of pathology (Braak stages) for Parkinson’s disease, which correlated very well with the clinical symptom presentation in the different phases of disease [33] [34] [35], and over the years was adapted to the latest state of knowledge [36]. According to Braak stages currently in use, the neuropathological process of Parkinson’s disease begins in the olfactory bulb, in the ENS, in the intermediolateral nucleus (IML) of the spinal cord, and in the dorsal motor nucleus of the vagus nerve (DMNV) [36] [37] [38] [39]. The detection of α-synuclein in the olfactory bulb is associated with a neuropathologically confirmed diagnosis of advanced PD and with dementia with Lewy bodies (DLB) with a sensitivity and specificity of more than 90%, and also correlates with the extent of synucleinopathy in other brain regions [40]. Lewy bodies and Lewy neurites were also found in the peripheral nervous system of the intestine and sympathetic and parasympathetic ganglia [41] [42] [43] [44]. These pathological changes have been observed in Auerbach’s Plexus and Meissner’s plexus of the ENS and in the olfactory bulb, both in patients with diagnosed PD but also in people who did not noticeably demonstrate the classic motor symptoms of PD or the classic pathological changes in the CNS [41] [45] [46] [47] [48]. In autopsy studies of patients with PD, α-synuclein was found in the ENS in 50–100% of cases and in 0–52% of cases in autopsy studies of control subjects; in in vivo studies, α-synuclein was found in the ENS in 66–100% of patients with Parkinson’s disease and in 0–8% of control subjects [49] [50]. In contrast, phosphorylated α-synuclein was detected in 23–93% of patients with Parkinson’s, but not in control subjects [51] [52]. The analysis of the Danish National Pathology Registry showed that Lewy bodies were detectable in the gastrointestinal tract of patients with PD up to 20 years before the development of Parkinsonian syndrome [53]. Furthermore, in this study, the detection of phosphorylated α-synuclein was significantly higher in these patients compared to controls [53]. Other smaller case series also demonstrated phosphorylated α- synuclein in the gastrointestinal tract in patients in the prodromal phase of PD [54] and dementia with Lewy bodies [55] [56]. In addition, in autopsies of individuals of advanced age who had suffered from constipation but had shown no clinical symptoms of Parkinson’s disease or dementia prior to their death, incidental Lewy bodies in the substantia nigra and the locus coeruleus were found, and this was associated with a reduced neuron density in the substantia nigra [57] [58]. The inclusion of both the olfactory and the gastrointestinal system in the neurodegenerative process of PD is explained by a “dual-hit” mechanism [59] [60]. Thus, a neurotrophic pathogen is inhaled and/or swallowed, which then initiates the pathological process. This is followed by an anterograde progression of the pathology of the olfactory system in the temporal lobes or retrograde progression from the gastrointestinal system via the sympathetic and parasympathetic nervous system to the brain stem ([Fig. 1]) [35] [61] [62]. This neurodegenerative process appears to spread only in anatomically connected structures in the caudorostral direction and, after reaching the brain stem, spread further to cortical regions and the spinal cord [38]. The motor symptoms that clinically define PD only occur after a 60–80% loss of dopaminergic neurons of the substantia nigra and, correspondingly, the dysfunction of the nigrostriatal dopaminergic control circuits [63]. In the further course of the disease, involvement of higher cortical structures is revealed, often by the presence of dementia [64].

Zoom Image
Fig. 1 a Schematic drawing of the brain and the gastrointestinal system with a dual-hit of the olfactory system and the gastrointestinal tract via inhalation/ingestion (red thread) with rostrocranial ascension of Parkinson’s pathology (red arrows) via sympathetic and parasympathetic nervous system (vagus nerve as green lines) reaching the central nervous system. Manifestation of non-motor symptoms with corresponding Braak stage (black arrows to corresponding anatomical structure). b Histologic intestinal section with A: mucosa, B: submucosa, c ring muscle, d longitudinal muscle and the enteric nervous system with E: Meissner’s plexus and F: Auerbach’s plexus. c α-synuclein inclusion similar to Lewy body (appearing red by immunohistochemical staining after proteinase K digestion) as Parkinson pathology.

Of course, there are also findings that cannot be clearly classified into Braak stages, raising questions about the validity of the classification system. Thus, retrospective autopsy studies have shown that in up to half of the examined patients, the pathological findings do not conform to the Braak stages [65] [66] [67] [68] [69] [70]. So, for example, it was reported that in 7% of patients with PD, no α-synuclein was detectable in DMNV despite the detection of α-synuclein in the substantia nigra and in the cortex [65]. In another study, this result was confirmed in 8.3% of patients with PD [66]. Also, phosphorylated α-synuclein was shown in the ENS, particularly of older healthy control subjects, and not at all, or only slightly in younger control subjects [71] [72]. This suggests that the phosphorylation of α-synuclein and the accumulation of phosphorylated α-synuclein might also be a part of the process of aging [71] [73]. Furthermore, the relationship between PD-specific pathology and impaired gastrointestinal motility in patients with PD is not really understood. In studies that looked for Lewy pathology in the ENS in patients with Lewy body disease, α-synuclein could be detected in the ENS of almost every patient with PD [51] [74] but neither neuronal loss of Auerbach’s plexus nor a change in the neurochemical composition could be demonstrated in patients with PD compared to healthy controls [48] [75]. On the other hand, a 15% neuronal loss per ganglion of Meissner’s plexus [76] and a loss of dopaminergic neurons of Auerbach’s plexus [77] were described in colonic biopsies of patients with PD from other centres. Neurochemical changes such as low concentrations of glutathione and prostaglandin [78] and high expression of glial fibrillary acidic protein and Sox 10 [79] in patients with PD suggest dysregulation and loss of enteric glial cells [79] [80] [81]. Thus, a positive correlation between gastrointestinal permeability, which is regulated by enteric glial cells, on the one hand, and the intestinal amount of α-synuclein in untreated patients, on the other, was found in the early phase of PD [82]. Enteric glial cells may thus be involved in the initiation and progression of PD in the context of gastrointestinal disorder [83]. The extent of glial markers decreases with prolonged duration of PD, suggesting a stronger role of glial cells at the onset of disease and a decrease with time [79]. Thus, the extent of enteric neuronal loss and neurochemical changes in the different autopsy studies in patients with advanced PD could be explained by the inclusion of enteric glial cells and their different behaviour during the progression of PD [75] [76] [77].

Each classification system will have weak points and deviating results should always be expected. Thus, the distribution of Lewy bodies in PD might be more diverse than hitherto suspected. However, the different study results could also be due to the different cohorts and techniques used as well as the detection of different types of α-synuclein or the inclusion of patients with incidental Lewy-body disease (ILBD) and not with PD [84] [85] [86] [87]. An ILBD is defined by the presence of cerebral Lewy bodies in autopsies of individuals without evidence of a neurodegenerative disease before their death [88] [89]. It is assumed that 5–24% of the elderly population have ILDB [69] [84] [90] and could correspond to the preclinical stage of synucleinopathy [91]. Nevertheless, detection of Lewy bodies is essential for the pathological diagnosis of PD, whereas it does not appear to be specific for prodromal PD [84].


#

Evidence for Ascension Hypothesis using Animal Models for Parkinsons Disease

Currently, there is no animal model that fully maps the temporal and spatial spread of Parkinson pathology corresponding to the Braak stages. Some animal models may present clinical symptoms such as hypokinesia and symptomatic effects of levodopa and dopamine agonists, while other pathological changes such as the loss of tyrosine hydroxylase-positive and dopaminergic neurons in the substantia nigra, the loss of tyrosine hydroxylase-positive neurons in the ENS or α- synuclein inclusions in the substantia nigra and in the ENS. In the different animal models generated by toxins, not only the nature of the toxin (for example, MPTP, the pesticide rotenone or herbicide paraquat) [92] [93] [94] but also the route of administration (systemic, local) [95] [96] [97] plays an essential role. Pathogenic environmental factors must be able to overcome physiological defence mechanisms, such as the mucous membranes of the nasopharyngeal and gastrointestinal system, before they can be absorbed. Thus, the administration route of the toxin is crucial for triggering the pathology of PD in a realistic and natural form. The chronic intranasal administration of MPTP induces motor symptoms in mice, a reduction in the striatal dopamine content and the loss of tyrosine hydroxylase in the substantia nigra and in the striatum [96]. While chronic inhalation of rotenone did not produce any clinical or pathological changes typical of PD in mice and rats [96], systemic administration induced Parkinson’s-typical pathological changes with nigrostriatal neurodegeneration and formation of α-synuclein inclusions similar to Lewy bodies [95] [98]. Furthermore, a degeneration of CNS structures could be detected, which is not usually associated with the PD and this process also showed no progression [98]. Pan-Montojo et al. [97] applied low doses of rotenone in wild-type mice in a chronic intragastric fashion (by means of a gastric tube) with the aim of inducing neuropathological changes typical of Parkinson’s. Rotenone was administered at such low doses that it had only a local effect on the ENS, but not a systemic effect. In mice treated for 1.5 months, typical Parkinsonian neuropathological changes occurred, such as α-synuclein aggregation in the ENS, IML and DMNV, while changes in the substantia nigra or motor symptoms were not observed. Both α-synuclein aggregation and a loss of dopaminergic neurons in the substantia nigra pars compacta as well as motor symptoms could be demonstrated in mice treated for 3 months. In this mouse model, a temporal and spatial progression of the Parkinsonian pathology corresponding to the Braak stages and the clinical symptom presentation in patients with PD could be simulated. Inden et al. [99] have also shown that chronic oral administration of rotenone in mice resulted in selective nigrostriatal dopaminergic neurodegeneration, motor symptoms, and cytoplasmic accumulation of α-synuclein in dopaminergic neurons. Interestingly, the loss of dopaminergic neurons in the substantia nigra pars compacta occurred asymmetrically, corresponding to the presentation in humans with PD. Tasselli et al. [100] were able to show the neurodegeneration of the substantia nigra in mice after 4 weeks of chronic oral rotenone treatment, but no change in gastrointestinal motility or neuropathological changes of the ENS could be detected. What is certain is that the administration of rotenone leads to an accumulation of α-synuclein [95] [101] [102] [103] [104]


#

Accumulated α-Synuclein as a Pathological Correlate of Parkinson’s Disease

Mitochondrial dysfunction caused by a complex I defect [105] [106] [107], oxidative stress [108], inflammation and protein deficiency [109] [110] are crucial mechanisms in the pathogenesis of PD. Environmental toxins such as rotenone cause exocytosis and release of α-synuclein from enteric and sympathetic neurons into the extracellular space [111] [112] [113]. The released α-synuclein can be absorbed by neurons and retrograded to the soma [114] [115]. In addition to transmission from neuron to neuron, transmission to non-neuronal cells is possible. Transmitted α-synuclein can trigger aggregation of endogenous α-synuclein in neurons [116]. This accumulated α-synuclein affects the mitochondrial complex I activity, reduces mitochondrial function, and causes oxidative stress in the neurons [108] [117] [118]. Different species of neurons react with different sensitivity to the loss of mitochondrial complex I activity. Thus, the combination of rotenone and endogenous dopamine results in selective toxicity of dopaminergic neurons in cell cultures from the mesencephalon of mice and rats and in the substantia nigra pars compacta of mice [95] [119]. Central dopaminergic neurons also appear to have a higher sensitivity to the accumulation of intracellular α-synuclein than, for example, neurons of the DMNV and the IML [97]. These results show that the dopaminergic neurons of the substantia nigra have intrinsic sensitivity to complex I defects. Furthermore, a proinflammatory effect of α-synuclein might exacerbate the degeneration of the dopaminergic neurons. Thus extracellular α-synuclein leads to the release of inflammatory factors such as cytokines and activates microglia, which in turn leads to an inflammatory reaction [120] [121] [122]. Furthermore, a study showed that the combination of low-dose rotenone with an inflammogen works synergistically and leads to a selective degeneration of dopaminergic neurons, which shows that inflammatory changes can increase neurodegeneration [110]. The accumulation of Lewy bodies is, however, not restricted to the dopaminergic neurons of the substantia nigra, but also occurs in other dopaminergic, glutamatergic, noradrenergic, serotonergic, histaminergic, and cholinergic neurons. This is quite in agreeement with the clinically variable phenotypic presentation of the PD, especially with regard to non-motor subtypes [51] [123] [124] [125]. Finally, cells exposed to neuron-mediated α-synuclein show signs of apoptosis, such as the decay of the cell nucleus and caspase 3 activation [117] [126].


#

Ascension of Pathology from ENS to CNS

It is postulated that the Parkinsonian pathology propagates in the form of accumulated α-synuclein by means of sympathetic and parasympathetic nerves from the ENS to the CNS. α-synuclein is released from neurons into the extracellular space where it is either free or associated with exosomes [127] and can be taken up via endocytosis from neighbouring neurons and neuronal precursor cells [116] [126]. In a mouse model with Parkinson-like pathology, it was shown that when α-synuclein was transferred to transplanted neuronal precursor cells where it accumulated and formed inclusion bodies similar to Lewy bodies [126] [128]. Also, in autopsy studies of patients with PD who had received fetal mesencephalic tissue transplants, accumulated α-synuclein could be detected in the transplanted neurons, suggesting that there must have been a transfer of α-synuclein from the “host” to the transplant neurons [129] [130]. In a mouse model, the progress of Parkinsonian-like pathology could be stopped by interrupting the connection between the enteric neurons and the sympathetic or parasympathetic neurons [113]. Thus, hemivagotomy or partial sympathectomy carried out before the occurrence of motor symptoms delayed the occurrence of these symptoms, but not the occurrence of gastrointestinal symptoms, in rotenone-treated and operated mice compared to rotenone-treated but non-operated mice [31]. Furthermore, the amount of accumulated α-synuclein in choline acetyltransferase (ChAT)-positive neurons was significantly greater in the DMNV contralateral to the hemivagotomy compared to the DMNV ipsilateral to the hemivagotomy [113] in the rotonone-treated and hemivagotomized mice. This asymmetry could not be explained by neuronal cell death in the ipsilateral DMNV, which can be a consequence of hemivagotomy [131]. Thus, hemivagotomy seems to stop the progression of Parkinsonian-like pathology in the sense of transmission of accumulated α-synuclein from the ENS via the vagal nerve to the DMNV [113]. These results are supported by another study, which also showed an active axonal transport of α-synuclein from the intestine to the DMNV via the vagus nerve in a time-dependent fashion after injection of various forms of α-synuclein into the intestinal wall in rats [132]. In contrast to other studies, the transport of α-synuclein did not lead to neuronal cell death, and exogenous α-synuclein could not be demonstrated in the DMNV or the substantia nigra [132]. Whether this is a dose-dependent effect of the amount of α-synuclein injected, whether a certain incubation time is required after injection of α-synuclein into the intestinal wall, or whether exogenous α-synuclein failed to initiate aggregation of endogenous α-synuclein in neurons, is currently not clear [116] [133].

A large epidemiological study supports the findings of these animal model studies [134]. Here it was shown that patients who underwent truncal vagotomy, but not superselective, had a low risk for the development of PD than the general population. The incidence of PD in vagotomized persons with a follow-up of more than 20 years was 0.65 per 1 000 person-years, compared to 1.28 per 1 000 person-years in adapted controls who had no vagotomy (adapted for the year of birth, sex, so-called index date, which means that the control person had no vagotomy after the vagotomy date of the corresponding Parkinsonian patient). These results demonstrate the long prodromal phase of PD and the vagus nerve as a rostrocranial pathway of the Parkinsonian pathology to the CNS. In the animal model, vagotomized mice showed a lower dopaminergic neuronal cell death in the substantia nigra pars compacta ipsilateral to the vagotomy side than mice that were not vagotomised after 4 months of chronic gastric rotenone treatment [113]. Soon after Parkinson’s pathology reaches the DMNV, it appears to spread further to the CNS. In summary, the spread of Parkinsonian pathology appears to be due to an intact synaptic linkage. Thus, only neurons with synaptic connection to the ENS show pathological changes after gastric rotenone treatment [97].


#

Summary

Non-motor symptoms, in particular olfactory disturbances and constipation, appear many years ahead of Parkinson’s disease with motor symptoms; these becomes clinically manifest only when there is a degeneration of 60–80% of the dopaminergic neurons in the substantia nigra. In addition to these non-motor symptoms, the pathological process runs its course decades before the clinical manifestation of the PD according to the UK Brain Bank criteria. The olfactory and gastrointestinal systems can serve as contact points to the environment as an entry site for pathogenic substances via inhalation and swallowing. The Braak stages postulate that the pathological process of PD begins in the olfactory bulb and in the ENS, and in the form of α-synuclein, spreads rostrocranially by transsyptic cell-to-cell transport mechanism via the sympathetic and parasympathetic nervous system over the DMNV and IML to the CNS. Thus, PD follows clinically and also pathologically a specific temporal and spatial sequence. Epidemiological, clinical, in particular autopsy studies and animal model studies have confirmed decisive aspects of the Braak stages. However, there are also conflicting study results such as the loss of nerve cells in the ENS as part of the pathological process, the role of enteric glial cells and the gastrointestinal microbiome, as well as different results concerning neurochemical changes in the gastrointestinal tract and the relationship between constipation and neuropathological changes. Furthermore, the Lewy body pathology is binding for the diagnosis of PD but not specific for idiopathic PD in the prodromal phase, since even incidental Lewy pathology could be demonstrated especially in the elderly, and of course α-synuclein in other synucleinopathies. This should be kept in mind in discussions about detection of α-synuclein in biospies, in particular from the gastrointestinal tract, as a promising biomarker for PD. Currently, lacking adequate specificity and sensitivity, it has no diagnostic use in clinical routine in suspected PD. In the future, however, testing for phosphorylated α-synuclein in tissue samples could be carried out in the course of routine gastrointestinal screening in combination with controlling for the presence of non-motor symptoms of PD as a clinical test battery for staging risk of possible PD. This would enable, on the one hand, investigation of neuroprotective therapies and, on the other, defining additional predisposing factors for PD that influence the progress rate or the course of the disease and the phenotypic variability of the PD.


#
#

Conflict of Interest

The authors declare they have no conflict of interest.

Acknowledgment

We would like to thank Dr. J. Schultz-Schaeffer, Head of Prion and Dementia Research, National Reference Centre for Spongiform Encephalopathies, University Hospital Göttingen, Germany, for the histological sections ([Fig. 1b,c]).

  • References

  • 1 Hughes AJ, Daniel SE, Kilford L. et al. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry 1992; 55: 181-184
  • 2 Postuma RB, Berg D, Stern M. et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord 2015; 30: 1591-601
  • 3 Berg D, Postuma RB, Adler CH. et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord 2015; 30: 1600-1611
  • 4 Haehner A, Hummel T, Reichmann H. Olfactory dysfunction as a diagnostic marker for Parkinson’s disease. Expert Rev Neurother 2009; 9: 1773-1779
  • 5 Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol 2012; 8: 329-339
  • 6 Iranzo A, Molinuevo JL, Santamaria J. et al. Rapid-eye-movement sleep behaviour disorder as an early marker for a neurodegenerative disorder: a descriptive study. Lancet Neurol 2006; 5: 572-577
  • 7 Stiasny-Kolster K, Doerr Y, Moller JC. et al. Combination of ‘idiopathic’ REM sleep behaviour disorder and olfactory dysfunction as possible indicator for alpha-synucleinopathy demonstrated by dopamine transporter FP-CIT-SPECT. Brain 2005; 128: 126-137
  • 8 Cersosimo MG, Raina GB, Pecci C. et al. Gastrointestinal manifestations in Parkinson’s disease: prevalence and occurrence before motor symptoms. J Neurol 2013; 260: 1332-1338
  • 9 Jost WH. Gastrointestinal dysfunction in Parkinson’s disease. J Neurol Sci 2010; 289: 69-73
  • 10 Reichmann H, Schneider C, Lohle M. Non-motor features of Parkinson’s disease: depression and dementia. Parkinsonism Relat Disord 2009; 15 (Suppl. 03) S87-S92
  • 11 Schrag A, Horsfall L, Walters K. et al. Prediagnostic presentations of Parkinson’s disease in primary care: a case-control study. Lancet Neurol 2015; 14: 57-64
  • 12 Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol 2003; 2: 107-116
  • 13 Rodriguez-Violante M, Cervantes-Arriaga A, Villar-Velarde A. et al. Prevalence of non-motor dysfunction among Parkinson’s disease patients from a tertiary referral center in Mexico City. Clin Neurol Neurosurg 2010; 112: 883-885
  • 14 Martinez-Martin P. The importance of non-motor disturbances to quality of life in Parkinson’s disease. J Neurol Sci 2011; 310: 12-16
  • 15 Chaudhuri KR, Martinez-Martin P, Schapira AH. et al. International multicenter pilot study of the first comprehensive self-completed nonmotor symptoms questionnaire for Parkinson’s disease: the NMSQuest study. Mov Disord 2006; 21: 916-923
  • 16 Martinez-Martin P, Schapira AH, Stocchi F. et al. Prevalence of nonmotor symptoms in Parkinson’s disease in an international setting; study using nonmotor symptoms questionnaire in 545 patients. Mov Disord 2007; 22: 1623-1629
  • 17 Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Parkinsonism Relat Disord 2011; 17: 10-15
  • 18 Jost WH. Gastrointestinal motility problems in patients with Parkinson’s disease. Effects of antiparkinsonian treatment and guidelines for management. Drugs Aging 1997; 10: 249-258
  • 19 Barone P, Antonini A, Colosimo C. et al. The PRIAMO study: A multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson’s disease. Mov Disord 2009; 24: 1641-1649
  • 20 Edwards LL, Pfeiffer RF, Quigley EM. et al. Gastrointestinal symptoms in Parkinson’s disease. Mov Disord 1991; 6: 151-156
  • 21 Mollenhauer B, Trautmann E, Sixel-Doring F. et al. Nonmotor and diagnostic findings in subjects with de novo Parkinson disease of the DeNoPa cohort. Neurology 2013; 81: 1226-1234
  • 22 Abbott RD, Petrovitch H, White LR. et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology 2001; 57: 456-462
  • 23 Savica R, Carlin JM, Grossardt BR. et al. Medical records documentation of constipation preceding Parkinson disease: A case-control study. Neurology 2009; 73: 1752-1758
  • 24 Adams-Carr KL, Bestwick JP, Shribman S et al. Constipation preceding Parkinson’s disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2015
  • 25 Gjerloff T, Fedorova T, Knudsen K. et al. Imaging acetylcholinesterase density in peripheral organs in Parkinson’s disease with 11C-donepezil PET. Brain 2015; 138: 653-663
  • 26 Cersosimo MG, Benarroch EE. Neural control of the gastrointestinal tract: implications for Parkinson disease. Mov Disord 2008; 23: 1065-1075
  • 27 Tissingh G, Berendse HW, Bergmans P. et al. Loss of olfaction in de novo and treated Parkinson’s disease: possible implications for early diagnosis. Mov Disord 2001; 16: 41-46
  • 28 Chaudhuri KR, Healy DG, Schapira AH. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol 2006; 5: 235-245
  • 29 Ponsen MM, Stoffers D, Booij J. et al. Idiopathic hyposmia as a preclinical sign of Parkinson’s disease. Ann Neurol 2004; 56: 173-181
  • 30 Spillantini MG, Schmidt ML, Lee VM. et al. Alpha-synuclein in Lewy bodies. Nature 1997; 388: 839-840
  • 31 Gaspar P, Gray F. Dementia in idiopathic Parkinson’s disease. A neuropathological study of 32 cases. Acta Neuropathol 1984; 64: 43-52
  • 32 Braak H, Braak E, Yilmazer D. et al. Pattern of brain destruction in Parkinson’s and Alzheimer’s diseases. J Neural Transm 1996; 103: 455-490
  • 33 Braak H, Del Tredici K, Rub U. et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 2003; 24: 197-211
  • 34 Braak H, Ghebremedhin E, Rub U. et al. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res 2004; 318: 121-134
  • 35 Del Tredici K, Rub U, De Vos RA. et al. Where does parkinson disease pathology begin in the brain?. J Neuropathol Exp Neurol 2002; 61: 413-426
  • 36 Del Tredici K, Braak H. Review: Sporadic Parkinson’s disease: development and distribution of alpha-synuclein pathology. Neuropathol Appl Neurobiol 2016; 42: 33-50
  • 37 Del Tredici K, Braak H. Spinal cord lesions in sporadic Parkinson’s disease. Acta Neuropathol 2012; 124: 643-664
  • 38 Braak H, Sastre M, Bohl JR. et al. Parkinson’s disease: lesions in dorsal horn layer I, involvement of parasympathetic and sympathetic pre- and postganglionic neurons. Acta Neuropathol 2007; 113: 421-429
  • 39 Bloch A, Probst A, Bissig H. et al. Alpha-synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects. Neuropathol Appl Neurobiol 2006; 32: 284-295
  • 40 Beach TG, White 3rd CL, Hladik CL. et al. Olfactory bulb alpha-synucleinopathy has high specificity and sensitivity for Lewy body disorders. Acta Neuropathol 2009; 117: 169-174
  • 41 Braak H, de Vos RA, Bohl J. et al. Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci Lett 2006; 396: 67-72
  • 42 Del Tredici K, Hawkes CH, Ghebremedhin E. et al. Lewy pathology in the submandibular gland of individuals with incidental Lewy body disease and sporadic Parkinson’s disease. Acta Neuropathol 2010; 119: 703-713
  • 43 Orimo S, Amino T, Itoh Y. et al. Cardiac sympathetic denervation precedes neuronal loss in the sympathetic ganglia in Lewy body disease. Acta Neuropathol 2005; 109: 583-588
  • 44 Minguez-Castellanos A, Chamorro CE, Escamilla-Sevilla F. et al. Do alpha-synuclein aggregates in autonomic plexuses predate Lewy body disorders?: a cohort study. Neurology 2007; 68: 2012-2018
  • 45 Wakabayashi K, Takahashi H, Ohama E. et al. Parkinson’s disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol 1990; 79: 581-583
  • 46 Wakabayashi K, Takahashi H, Takeda S. et al. Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol 1988; 76: 217-221
  • 47 Kupsky WJ, Grimes MM, Sweeting J. et al. Parkinson’s disease and megacolon: concentric hyaline inclusions (Lewy bodies) in enteric ganglion cells. Neurology 1987; 37: 1253-1255
  • 48 Lebouvier T, Chaumette T, Damier P. et al. Pathological lesions in colonic biopsies during Parkinson’s disease. Gut 2008; 57: 1741-1743
  • 49 Gold A, Turkalp ZT, Munoz DG. Enteric alpha-synuclein expression is increased in Parkinson’s disease but not Alzheimer’s disease. Mov Disord 2013; 28: 237-240
  • 50 Shannon KM, Keshavarzian A, Mutlu E. et al. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov Disord 2012; 27: 709-715
  • 51 Beach TG, Adler CH, Sue LI. et al. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol 2010; 119: 689-702
  • 52 Pouclet H, Lebouvier T, Coron E. et al. A comparison between rectal and colonic biopsies to detect Lewy pathology in Parkinson’s disease. Neurobiol Dis 2012; 45: 305-309
  • 53 Stokholm MG, Danielsen EH, Hamilton-Dutoit SJ. et al. Pathological alpha-synuclein in gastrointestinal tissues from prodromal Parkinson disease patients. Ann Neurol 2016; 79: 940-949
  • 54 Hilton D, Stephens M, Kirk L. et al. Accumulation of alpha-synuclein in the bowel of patients in the pre-clinical phase of Parkinson’s disease. Acta Neuropathol 2014; 127: 235-241
  • 55 Shannon KM, Keshavarzian A, Dodiya HB. et al. Is alpha-synuclein in the colon a biomarker for premotor Parkinson’s disease? Evidence from 3 cases. Mov Disord 2012; 27: 716-719
  • 56 Ito S, Takao M, Hatsuta H. et al. Alpha-synuclein immunohistochemistry of gastrointestinal and biliary surgical specimens for diagnosis of Lewy body disease. Int J Clin Exp Pathol 2014; 7: 1714-1723
  • 57 Abbott RD, Ross GW, Petrovitch H. et al. Bowel movement frequency in late-life and incidental Lewy bodies. Mov Disord 2007; 22: 1581-1586
  • 58 Petrovitch H, Abbott RD, Ross GW. et al. Bowel movement frequency in late-life and substantia nigra neuron density at death. Mov Disord 2009; 24: 371-376
  • 59 Reichmann H. View point: etiology in Parkinson’s disease. Dual hit or spreading intoxication. J Neurol Sci 2011; 310: 9-11
  • 60 Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol 2007; 33: 599-614
  • 61 Braak H, Rub U, Gai WP. et al. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm 2003; 110: 517-536
  • 62 Braak H, Del Tredici K. Neuroanatomy and pathology of sporadic Parkinson’s disease. Adv Anat Embryol Cell Biol 2009; 201: 1-119
  • 63 Albin RL, Young AB, Penney JB. The functional anatomy of basal ganglia disorders. Trends Neurosci 1989; 12: 366-375
  • 64 Klingelhoefer L, Reichmann H. Dementia – the real problem for patients with Parkinson’s disease. Basal Ganglia 2014; 4: 9-13
  • 65 Kalaitzakis ME, Graeber MB, Gentleman SM. et al. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease: a critical analysis of alpha-synuclein staging. Neuropathol Appl Neurobiol 2008; 34: 284-295
  • 66 Attems J, Jellinger KA. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease. Neuropathol Appl Neurobiol 2008; 34: 466-467
  • 67 Zaccai J, Brayne C, McKeith I. et al. Patterns and stages of alpha-synucleinopathy: Relevance in a population-based cohort. Neurology 2008; 70: 1042-1048
  • 68 Parkkinen L, Pirttila T, Alafuzoff I. Applicability of current staging/categorization of alpha-synuclein pathology and their clinical relevance. Acta Neuropathol 2008; 115: 399-407
  • 69 Jellinger KA. Lewy body-related alpha-synucleinopathy in the aged human brain. J Neural Transm 2004; 111: 1219-1235
  • 70 Jellinger KA. Alpha-synuclein pathology in Parkinson’s and Alzheimer’s disease brain: incidence and topographic distribution – a pilot study. Acta Neuropathol 2003; 106: 191-201
  • 71 Bottner M, Zorenkov D, Hellwig I. et al. Expression pattern and localization of alpha-synuclein in the human enteric nervous system. Neurobiol Dis 2012; 48: 474-480
  • 72 Visanji NP, Marras C, Kern DS. et al. Colonic mucosal a-synuclein lacks specificity as a biomarker for Parkinson disease. Neurology 2015; 84: 609-616
  • 73 Muntane G, Ferrer I, Martinez-Vicente M. Alpha-synuclein phosphorylation and truncation are normal events in the adult human brain. Neuroscience 2012; 200: 106-119
  • 74 Gelpi E, Navarro-Otano J, Tolosa E. et al. Multiple organ involvement by alpha-synuclein pathology in Lewy body disorders. Mov Disord 2014; 29: 1010-1018
  • 75 Annerino DM, Arshad S, Taylor GM. et al. Parkinson’s disease is not associated with gastrointestinal myenteric ganglion neuron loss. Acta Neuropathol 2012; 124: 665-680
  • 76 Lebouvier T, Neunlist M. Bruley des Varannes S et al. Colonic biopsies to assess the neuropathology of Parkinson’s disease and its relationship with symptoms. PLoS One 2010; 5: e12728
  • 77 Singaram C, Ashraf W, Gaumnitz EA. et al. Dopaminergic defect of enteric nervous system in Parkinson’s disease patients with chronic constipation. Lancet 1995; 346: 861-864
  • 78 De Giorgio R, Giancola F, Boschetti E. et al. Enteric glia and neuroprotection: basic and clinical aspects. Am J Physiol Gastrointest Liver Physiol 2012; 303: G887-G893
  • 79 Devos D, Lebouvier T, Lardeux B. et al. Colonic inflammation in Parkinson’s disease. Neurobiol Dis 2013; 50: 42-48
  • 80 Clairembault T, Kamphuis W, Leclair-Visonneau L. et al. Enteric GFAP expression and phosphorylation in Parkinson’s disease. J Neurochem 2014; 130: 805-815
  • 81 Bassotti G, Villanacci V, Maurer CA. et al. The role of glial cells and apoptosis of enteric neurones in the neuropathology of intractable slow transit constipation. Gut 2006; 55: 41-46
  • 82 Forsyth CB, Shannon KM, Kordower JH. et al. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS One 2011; 6: e28032
  • 83 Clairembault T, Leclair-Visonneau L, Neunlist M. et al. Enteric glial cells: New players in Parkinson’s disease?. Mov Disord 2015; 30: 494-498
  • 84 Halliday G, McCann H, Shepherd C. Evaluation of the Braak hypothesis: how far can it explain the pathogenesis of Parkinson’s disease?. Expert Rev Neurother 2012; 12: 673-686
  • 85 Parkkinen L, Soininen H, Laakso M. et al. Alpha-synuclein pathology is highly dependent on the case selection. Neuropathol Appl Neurobiol 2001; 27: 314-325
  • 86 Antunes L, Frasquilho S, Ostaszewski M. et al. Similar alpha-Synuclein staining in the colon mucosa in patients with Parkinson’s disease and controls. Mov Disord 2016; 31: 1567-1570
  • 87 Schneider SA, Boettner M, Alexoudi A. et al. Can we use peripheral tissue biopsies to diagnose Parkinson’s disease? A review of the literature. Eur J Neurol 2016; 23: 247-261
  • 88 Parkkinen L, Kauppinen T, Pirttila T. et al. Alpha-synuclein pathology does not predict extrapyramidal symptoms or dementia. Ann Neurol 2005; 57: 82-91
  • 89 Forno LS. Concentric hyalin intraneuronal inclusions of Lewy type in the brains of elderly persons (50 incidental cases): relationship to parkinsonism. J Am Geriatr Soc 1969; 17: 557-575
  • 90 Saito Y, Ruberu NN, Sawabe M. et al. Lewy body-related alpha-synucleinopathy in aging. J Neuropathol Exp Neurol 2004; 63: 742-749
  • 91 Dickson DW, Fujishiro H, DelleDonne A. et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol 2008; 115: 437-444
  • 92 Bove J, Prou D, Perier C. et al. Toxin-induced models of Parkinson’s disease. NeuroRx 2005; 2: 484-494
  • 93 McDowell K, Chesselet MF. Animal models of the non-motor features of Parkinson’s disease. Neurobiol Dis 2012; 46: 597-606
  • 94 Hisahara S, Shimohama S. Toxin-induced and genetic animal models of Parkinson’s disease. Parkinsons Dis 2010; 2011: 951709
  • 95 Betarbet R, Sherer TB, MacKenzie G. et al. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 2000; 3: 1301-1306
  • 96 Rojo AI, Cavada C, de Sagarra MR. et al. Chronic inhalation of rotenone or paraquat does not induce Parkinson’s disease symptoms in mice or rats. Exp Neurol 2007; 208: 120-126
  • 97 Pan-Montojo F, Anichtchik O, Dening Y. et al. Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS One 2010; 5: e8762
  • 98 Hoglinger GU, Feger J, Prigent A. et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J Neurochem 2003; 84: 491-502
  • 99 Inden M, Kitamura Y, Takeuchi H. et al. Neurodegeneration of mouse nigrostriatal dopaminergic system induced by repeated oral administration of rotenone is prevented by 4-phenylbutyrate, a chemical chaperone. J Neurochem 2007; 101: 1491-1504
  • 100 Tasselli M, Chaumette T, Paillusson S. et al. Effects of oral administration of rotenone on gastrointestinal functions in mice. Neurogastroenterol Motil 2013; 25: e183-e193
  • 101 Sherer TB, Kim JH, Betarbet R. et al. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol 2003; 179: 9-16
  • 102 Silva BA, Einarsdottir O, Fink AL. et al. Biophysical Characterization of alpha-Synuclein and Rotenone Interaction. Biomolecules 2013; 3: 703-732
  • 103 Yuan YH, Yan WF, Sun JD. et al. The molecular mechanism of rotenone-induced alpha-synuclein aggregation: Emphasizing the role of the calcium/GSK3beta pathway. Toxicol Lett 2015; 233: 163-171
  • 104 Chorfa A, Lazizzera C, Betemps D et al. A variety of pesticides trigger in vitro alpha-synuclein accumulation, a key event in Parkinson’s disease. Arch Toxicol 2014
  • 105 Schapira AH. Disease modification in Parkinson’s disease. Lancet Neurol 2004; 3: 362-368
  • 106 Mizuno Y, Ohta S, Tanaka M. et al. Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochem Biophys Res Commun 1989; 163: 1450-1455
  • 107 Franco-Iborra S, Vila M, Perier C. The parkinson disease mitochondrial hypothesis: where are ee at?. Neuroscientist 2016; 22: 266-277
  • 108 Jenner P. Oxidative stress in Parkinson’s disease. Ann Neurol 2003; 53 (Suppl. 03) S26-S36 discussion S36–S38
  • 109 McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord 2004; 10 (Suppl. 01) S3-S7
  • 110 Gao HM, Hong JS, Zhang W. et al. Synergistic dopaminergic neurotoxicity of the pesticide rotenone and inflammogen lipopolysaccharide: relevance to the etiology of Parkinson’s disease. J Neurosci 2003; 23: 1228-1236
  • 111 Alvarez-Erviti L, Seow Y, Schapira AH. et al. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol Dis 2011; 42: 360-367
  • 112 Hasegawa T, Konno M, Baba T. et al. The AAA-ATPase VPS4 regulates extracellular secretion and lysosomal targeting of alpha-synuclein. PLoS One 2011; 6: e29460
  • 113 Pan-Montojo F, Schwarz M, Winkler C. et al. Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice. Sci Rep 2012; 2: 898
  • 114 Luk KC, Kehm V, Carroll J. et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 2012; 338: 949-953
  • 115 Lee HJ, Suk JE, Bae EJ. et al. Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. Int J Biochem Cell Biol 2008; 40: 1835-1849
  • 116 Angot E, Steiner JA, Lema Tome CM. et al. Alpha-synuclein cell-to-cell transfer and seeding in grafted dopaminergic neurons in vivo. PLoS One 2012; 7: e39465
  • 117 Braidy N, Gai WP, Xu YH. et al. Alpha-synuclein transmission and mitochondrial toxicity in primary human foetal enteric neurons in vitro. Neurotox Res 2014; 25: 170-182
  • 118 Esteves AR, Arduino DM, Silva DF. et al. Mitochondrial Dysfunction: The Road to Alpha-Synuclein Oligomerization in PD. Parkinsons Dis 2011; 2011: 693761
  • 119 Choi WS, Palmiter RD, Xia Z. Loss of mitochondrial complex I activity potentiates dopamine neuron death induced by microtubule dysfunction in a Parkinson’s disease model. J Cell Biol 2011; 192: 873-882
  • 120 Klegeris A, Giasson BI, Zhang H. et al. Alpha-synuclein and its disease-causing mutants induce ICAM-1 and IL-6 in human astrocytes and astrocytoma cells. FASEB J 2006; 20: 2000-2008
  • 121 McGeer PL, McGeer EG. The alpha-synuclein burden hypothesis of Parkinson disease and its relationship to Alzheimer disease. Exp Neurol 2008; 212: 235-238
  • 122 Hunot S, Dugas N, Faucheux B. et al. FcepsilonRII/CD23 is expressed in Parkinson’s disease and induces, in vitro, production of nitric oxide and tumor necrosis factor-alpha in glial cells. J Neurosci 1999; 19: 3440-3447
  • 123 Dickson DW, Braak H, Duda JE. et al. Neuropathological assessment of Parkinson’s disease: refining the diagnostic criteria. Lancet Neurol 2009; 8: 1150-1157
  • 124 Jellinger KA. Neuropathobiology of non-motor symptoms in Parkinson disease. J Neural Transm (Vienna) 2015; 122: 1429-1440
  • 125 Sauerbier A, Jenner P, Todorova A. et al. Non motor subtypes and Parkinson’s disease. Parkinsonism Relat Disord 2016; 22 (Suppl. 01) S41-S46
  • 126 Desplats P, Lee HJ, Bae EJ. et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA 2009; 106: 13010-13015
  • 127 Danzer KM, Kranich LR, Ruf WP. et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener 2012; 7: 42
  • 128 Brundin P, Li JY, Holton JL. et al. Research in motion: the enigma of Parkinson’s disease pathology spread. Nat Rev Neurosci 2008; 9: 741-745
  • 129 Li JY, Englund E, Holton JL. et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med 2008; 14: 501-503
  • 130 Kordower JH, Brundin P. Lewy body pathology in long-term fetal nigral transplants: is Parkinson’s disease transmitted from one neural system to another?. Neuropsychopharmacology 2009; 34: 254
  • 131 Ling EA, Wong WC, Yick TY. et al. Ultrastructural changes in the dorsal motor nucleus of monkey following bilateral cervical vagotomy. J Neurocytol 1986; 15: 1-15
  • 132 Holmqvist S, Chutna O, Bousset L. et al. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol 2014; 128: 805-820
  • 133 Hardy J. Expression of normal sequence pathogenic proteins for neurodegenerative disease contributes to disease risk: ’permissive templating’ as a general mechanism underlying neurodegeneration. Biochem Soc Trans 2005; 33: 578-581
  • 134 Svensson E, Horvath-Puho E, Thomsen RW. et al. Vagotomy and subsequent risk of Parkinson’s disease. Ann Neurol 2015; 78: 522-529

Correspondence

Dr. med. Lisa Klingelhoefer
Department of Neurology
Technical University Dresden
Fetscherstraße 74
01307 Dresden

  • References

  • 1 Hughes AJ, Daniel SE, Kilford L. et al. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry 1992; 55: 181-184
  • 2 Postuma RB, Berg D, Stern M. et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord 2015; 30: 1591-601
  • 3 Berg D, Postuma RB, Adler CH. et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord 2015; 30: 1600-1611
  • 4 Haehner A, Hummel T, Reichmann H. Olfactory dysfunction as a diagnostic marker for Parkinson’s disease. Expert Rev Neurother 2009; 9: 1773-1779
  • 5 Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol 2012; 8: 329-339
  • 6 Iranzo A, Molinuevo JL, Santamaria J. et al. Rapid-eye-movement sleep behaviour disorder as an early marker for a neurodegenerative disorder: a descriptive study. Lancet Neurol 2006; 5: 572-577
  • 7 Stiasny-Kolster K, Doerr Y, Moller JC. et al. Combination of ‘idiopathic’ REM sleep behaviour disorder and olfactory dysfunction as possible indicator for alpha-synucleinopathy demonstrated by dopamine transporter FP-CIT-SPECT. Brain 2005; 128: 126-137
  • 8 Cersosimo MG, Raina GB, Pecci C. et al. Gastrointestinal manifestations in Parkinson’s disease: prevalence and occurrence before motor symptoms. J Neurol 2013; 260: 1332-1338
  • 9 Jost WH. Gastrointestinal dysfunction in Parkinson’s disease. J Neurol Sci 2010; 289: 69-73
  • 10 Reichmann H, Schneider C, Lohle M. Non-motor features of Parkinson’s disease: depression and dementia. Parkinsonism Relat Disord 2009; 15 (Suppl. 03) S87-S92
  • 11 Schrag A, Horsfall L, Walters K. et al. Prediagnostic presentations of Parkinson’s disease in primary care: a case-control study. Lancet Neurol 2015; 14: 57-64
  • 12 Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol 2003; 2: 107-116
  • 13 Rodriguez-Violante M, Cervantes-Arriaga A, Villar-Velarde A. et al. Prevalence of non-motor dysfunction among Parkinson’s disease patients from a tertiary referral center in Mexico City. Clin Neurol Neurosurg 2010; 112: 883-885
  • 14 Martinez-Martin P. The importance of non-motor disturbances to quality of life in Parkinson’s disease. J Neurol Sci 2011; 310: 12-16
  • 15 Chaudhuri KR, Martinez-Martin P, Schapira AH. et al. International multicenter pilot study of the first comprehensive self-completed nonmotor symptoms questionnaire for Parkinson’s disease: the NMSQuest study. Mov Disord 2006; 21: 916-923
  • 16 Martinez-Martin P, Schapira AH, Stocchi F. et al. Prevalence of nonmotor symptoms in Parkinson’s disease in an international setting; study using nonmotor symptoms questionnaire in 545 patients. Mov Disord 2007; 22: 1623-1629
  • 17 Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Parkinsonism Relat Disord 2011; 17: 10-15
  • 18 Jost WH. Gastrointestinal motility problems in patients with Parkinson’s disease. Effects of antiparkinsonian treatment and guidelines for management. Drugs Aging 1997; 10: 249-258
  • 19 Barone P, Antonini A, Colosimo C. et al. The PRIAMO study: A multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson’s disease. Mov Disord 2009; 24: 1641-1649
  • 20 Edwards LL, Pfeiffer RF, Quigley EM. et al. Gastrointestinal symptoms in Parkinson’s disease. Mov Disord 1991; 6: 151-156
  • 21 Mollenhauer B, Trautmann E, Sixel-Doring F. et al. Nonmotor and diagnostic findings in subjects with de novo Parkinson disease of the DeNoPa cohort. Neurology 2013; 81: 1226-1234
  • 22 Abbott RD, Petrovitch H, White LR. et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology 2001; 57: 456-462
  • 23 Savica R, Carlin JM, Grossardt BR. et al. Medical records documentation of constipation preceding Parkinson disease: A case-control study. Neurology 2009; 73: 1752-1758
  • 24 Adams-Carr KL, Bestwick JP, Shribman S et al. Constipation preceding Parkinson’s disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2015
  • 25 Gjerloff T, Fedorova T, Knudsen K. et al. Imaging acetylcholinesterase density in peripheral organs in Parkinson’s disease with 11C-donepezil PET. Brain 2015; 138: 653-663
  • 26 Cersosimo MG, Benarroch EE. Neural control of the gastrointestinal tract: implications for Parkinson disease. Mov Disord 2008; 23: 1065-1075
  • 27 Tissingh G, Berendse HW, Bergmans P. et al. Loss of olfaction in de novo and treated Parkinson’s disease: possible implications for early diagnosis. Mov Disord 2001; 16: 41-46
  • 28 Chaudhuri KR, Healy DG, Schapira AH. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol 2006; 5: 235-245
  • 29 Ponsen MM, Stoffers D, Booij J. et al. Idiopathic hyposmia as a preclinical sign of Parkinson’s disease. Ann Neurol 2004; 56: 173-181
  • 30 Spillantini MG, Schmidt ML, Lee VM. et al. Alpha-synuclein in Lewy bodies. Nature 1997; 388: 839-840
  • 31 Gaspar P, Gray F. Dementia in idiopathic Parkinson’s disease. A neuropathological study of 32 cases. Acta Neuropathol 1984; 64: 43-52
  • 32 Braak H, Braak E, Yilmazer D. et al. Pattern of brain destruction in Parkinson’s and Alzheimer’s diseases. J Neural Transm 1996; 103: 455-490
  • 33 Braak H, Del Tredici K, Rub U. et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 2003; 24: 197-211
  • 34 Braak H, Ghebremedhin E, Rub U. et al. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res 2004; 318: 121-134
  • 35 Del Tredici K, Rub U, De Vos RA. et al. Where does parkinson disease pathology begin in the brain?. J Neuropathol Exp Neurol 2002; 61: 413-426
  • 36 Del Tredici K, Braak H. Review: Sporadic Parkinson’s disease: development and distribution of alpha-synuclein pathology. Neuropathol Appl Neurobiol 2016; 42: 33-50
  • 37 Del Tredici K, Braak H. Spinal cord lesions in sporadic Parkinson’s disease. Acta Neuropathol 2012; 124: 643-664
  • 38 Braak H, Sastre M, Bohl JR. et al. Parkinson’s disease: lesions in dorsal horn layer I, involvement of parasympathetic and sympathetic pre- and postganglionic neurons. Acta Neuropathol 2007; 113: 421-429
  • 39 Bloch A, Probst A, Bissig H. et al. Alpha-synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects. Neuropathol Appl Neurobiol 2006; 32: 284-295
  • 40 Beach TG, White 3rd CL, Hladik CL. et al. Olfactory bulb alpha-synucleinopathy has high specificity and sensitivity for Lewy body disorders. Acta Neuropathol 2009; 117: 169-174
  • 41 Braak H, de Vos RA, Bohl J. et al. Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci Lett 2006; 396: 67-72
  • 42 Del Tredici K, Hawkes CH, Ghebremedhin E. et al. Lewy pathology in the submandibular gland of individuals with incidental Lewy body disease and sporadic Parkinson’s disease. Acta Neuropathol 2010; 119: 703-713
  • 43 Orimo S, Amino T, Itoh Y. et al. Cardiac sympathetic denervation precedes neuronal loss in the sympathetic ganglia in Lewy body disease. Acta Neuropathol 2005; 109: 583-588
  • 44 Minguez-Castellanos A, Chamorro CE, Escamilla-Sevilla F. et al. Do alpha-synuclein aggregates in autonomic plexuses predate Lewy body disorders?: a cohort study. Neurology 2007; 68: 2012-2018
  • 45 Wakabayashi K, Takahashi H, Ohama E. et al. Parkinson’s disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol 1990; 79: 581-583
  • 46 Wakabayashi K, Takahashi H, Takeda S. et al. Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol 1988; 76: 217-221
  • 47 Kupsky WJ, Grimes MM, Sweeting J. et al. Parkinson’s disease and megacolon: concentric hyaline inclusions (Lewy bodies) in enteric ganglion cells. Neurology 1987; 37: 1253-1255
  • 48 Lebouvier T, Chaumette T, Damier P. et al. Pathological lesions in colonic biopsies during Parkinson’s disease. Gut 2008; 57: 1741-1743
  • 49 Gold A, Turkalp ZT, Munoz DG. Enteric alpha-synuclein expression is increased in Parkinson’s disease but not Alzheimer’s disease. Mov Disord 2013; 28: 237-240
  • 50 Shannon KM, Keshavarzian A, Mutlu E. et al. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov Disord 2012; 27: 709-715
  • 51 Beach TG, Adler CH, Sue LI. et al. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol 2010; 119: 689-702
  • 52 Pouclet H, Lebouvier T, Coron E. et al. A comparison between rectal and colonic biopsies to detect Lewy pathology in Parkinson’s disease. Neurobiol Dis 2012; 45: 305-309
  • 53 Stokholm MG, Danielsen EH, Hamilton-Dutoit SJ. et al. Pathological alpha-synuclein in gastrointestinal tissues from prodromal Parkinson disease patients. Ann Neurol 2016; 79: 940-949
  • 54 Hilton D, Stephens M, Kirk L. et al. Accumulation of alpha-synuclein in the bowel of patients in the pre-clinical phase of Parkinson’s disease. Acta Neuropathol 2014; 127: 235-241
  • 55 Shannon KM, Keshavarzian A, Dodiya HB. et al. Is alpha-synuclein in the colon a biomarker for premotor Parkinson’s disease? Evidence from 3 cases. Mov Disord 2012; 27: 716-719
  • 56 Ito S, Takao M, Hatsuta H. et al. Alpha-synuclein immunohistochemistry of gastrointestinal and biliary surgical specimens for diagnosis of Lewy body disease. Int J Clin Exp Pathol 2014; 7: 1714-1723
  • 57 Abbott RD, Ross GW, Petrovitch H. et al. Bowel movement frequency in late-life and incidental Lewy bodies. Mov Disord 2007; 22: 1581-1586
  • 58 Petrovitch H, Abbott RD, Ross GW. et al. Bowel movement frequency in late-life and substantia nigra neuron density at death. Mov Disord 2009; 24: 371-376
  • 59 Reichmann H. View point: etiology in Parkinson’s disease. Dual hit or spreading intoxication. J Neurol Sci 2011; 310: 9-11
  • 60 Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol 2007; 33: 599-614
  • 61 Braak H, Rub U, Gai WP. et al. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm 2003; 110: 517-536
  • 62 Braak H, Del Tredici K. Neuroanatomy and pathology of sporadic Parkinson’s disease. Adv Anat Embryol Cell Biol 2009; 201: 1-119
  • 63 Albin RL, Young AB, Penney JB. The functional anatomy of basal ganglia disorders. Trends Neurosci 1989; 12: 366-375
  • 64 Klingelhoefer L, Reichmann H. Dementia – the real problem for patients with Parkinson’s disease. Basal Ganglia 2014; 4: 9-13
  • 65 Kalaitzakis ME, Graeber MB, Gentleman SM. et al. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease: a critical analysis of alpha-synuclein staging. Neuropathol Appl Neurobiol 2008; 34: 284-295
  • 66 Attems J, Jellinger KA. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease. Neuropathol Appl Neurobiol 2008; 34: 466-467
  • 67 Zaccai J, Brayne C, McKeith I. et al. Patterns and stages of alpha-synucleinopathy: Relevance in a population-based cohort. Neurology 2008; 70: 1042-1048
  • 68 Parkkinen L, Pirttila T, Alafuzoff I. Applicability of current staging/categorization of alpha-synuclein pathology and their clinical relevance. Acta Neuropathol 2008; 115: 399-407
  • 69 Jellinger KA. Lewy body-related alpha-synucleinopathy in the aged human brain. J Neural Transm 2004; 111: 1219-1235
  • 70 Jellinger KA. Alpha-synuclein pathology in Parkinson’s and Alzheimer’s disease brain: incidence and topographic distribution – a pilot study. Acta Neuropathol 2003; 106: 191-201
  • 71 Bottner M, Zorenkov D, Hellwig I. et al. Expression pattern and localization of alpha-synuclein in the human enteric nervous system. Neurobiol Dis 2012; 48: 474-480
  • 72 Visanji NP, Marras C, Kern DS. et al. Colonic mucosal a-synuclein lacks specificity as a biomarker for Parkinson disease. Neurology 2015; 84: 609-616
  • 73 Muntane G, Ferrer I, Martinez-Vicente M. Alpha-synuclein phosphorylation and truncation are normal events in the adult human brain. Neuroscience 2012; 200: 106-119
  • 74 Gelpi E, Navarro-Otano J, Tolosa E. et al. Multiple organ involvement by alpha-synuclein pathology in Lewy body disorders. Mov Disord 2014; 29: 1010-1018
  • 75 Annerino DM, Arshad S, Taylor GM. et al. Parkinson’s disease is not associated with gastrointestinal myenteric ganglion neuron loss. Acta Neuropathol 2012; 124: 665-680
  • 76 Lebouvier T, Neunlist M. Bruley des Varannes S et al. Colonic biopsies to assess the neuropathology of Parkinson’s disease and its relationship with symptoms. PLoS One 2010; 5: e12728
  • 77 Singaram C, Ashraf W, Gaumnitz EA. et al. Dopaminergic defect of enteric nervous system in Parkinson’s disease patients with chronic constipation. Lancet 1995; 346: 861-864
  • 78 De Giorgio R, Giancola F, Boschetti E. et al. Enteric glia and neuroprotection: basic and clinical aspects. Am J Physiol Gastrointest Liver Physiol 2012; 303: G887-G893
  • 79 Devos D, Lebouvier T, Lardeux B. et al. Colonic inflammation in Parkinson’s disease. Neurobiol Dis 2013; 50: 42-48
  • 80 Clairembault T, Kamphuis W, Leclair-Visonneau L. et al. Enteric GFAP expression and phosphorylation in Parkinson’s disease. J Neurochem 2014; 130: 805-815
  • 81 Bassotti G, Villanacci V, Maurer CA. et al. The role of glial cells and apoptosis of enteric neurones in the neuropathology of intractable slow transit constipation. Gut 2006; 55: 41-46
  • 82 Forsyth CB, Shannon KM, Kordower JH. et al. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS One 2011; 6: e28032
  • 83 Clairembault T, Leclair-Visonneau L, Neunlist M. et al. Enteric glial cells: New players in Parkinson’s disease?. Mov Disord 2015; 30: 494-498
  • 84 Halliday G, McCann H, Shepherd C. Evaluation of the Braak hypothesis: how far can it explain the pathogenesis of Parkinson’s disease?. Expert Rev Neurother 2012; 12: 673-686
  • 85 Parkkinen L, Soininen H, Laakso M. et al. Alpha-synuclein pathology is highly dependent on the case selection. Neuropathol Appl Neurobiol 2001; 27: 314-325
  • 86 Antunes L, Frasquilho S, Ostaszewski M. et al. Similar alpha-Synuclein staining in the colon mucosa in patients with Parkinson’s disease and controls. Mov Disord 2016; 31: 1567-1570
  • 87 Schneider SA, Boettner M, Alexoudi A. et al. Can we use peripheral tissue biopsies to diagnose Parkinson’s disease? A review of the literature. Eur J Neurol 2016; 23: 247-261
  • 88 Parkkinen L, Kauppinen T, Pirttila T. et al. Alpha-synuclein pathology does not predict extrapyramidal symptoms or dementia. Ann Neurol 2005; 57: 82-91
  • 89 Forno LS. Concentric hyalin intraneuronal inclusions of Lewy type in the brains of elderly persons (50 incidental cases): relationship to parkinsonism. J Am Geriatr Soc 1969; 17: 557-575
  • 90 Saito Y, Ruberu NN, Sawabe M. et al. Lewy body-related alpha-synucleinopathy in aging. J Neuropathol Exp Neurol 2004; 63: 742-749
  • 91 Dickson DW, Fujishiro H, DelleDonne A. et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol 2008; 115: 437-444
  • 92 Bove J, Prou D, Perier C. et al. Toxin-induced models of Parkinson’s disease. NeuroRx 2005; 2: 484-494
  • 93 McDowell K, Chesselet MF. Animal models of the non-motor features of Parkinson’s disease. Neurobiol Dis 2012; 46: 597-606
  • 94 Hisahara S, Shimohama S. Toxin-induced and genetic animal models of Parkinson’s disease. Parkinsons Dis 2010; 2011: 951709
  • 95 Betarbet R, Sherer TB, MacKenzie G. et al. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 2000; 3: 1301-1306
  • 96 Rojo AI, Cavada C, de Sagarra MR. et al. Chronic inhalation of rotenone or paraquat does not induce Parkinson’s disease symptoms in mice or rats. Exp Neurol 2007; 208: 120-126
  • 97 Pan-Montojo F, Anichtchik O, Dening Y. et al. Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS One 2010; 5: e8762
  • 98 Hoglinger GU, Feger J, Prigent A. et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J Neurochem 2003; 84: 491-502
  • 99 Inden M, Kitamura Y, Takeuchi H. et al. Neurodegeneration of mouse nigrostriatal dopaminergic system induced by repeated oral administration of rotenone is prevented by 4-phenylbutyrate, a chemical chaperone. J Neurochem 2007; 101: 1491-1504
  • 100 Tasselli M, Chaumette T, Paillusson S. et al. Effects of oral administration of rotenone on gastrointestinal functions in mice. Neurogastroenterol Motil 2013; 25: e183-e193
  • 101 Sherer TB, Kim JH, Betarbet R. et al. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol 2003; 179: 9-16
  • 102 Silva BA, Einarsdottir O, Fink AL. et al. Biophysical Characterization of alpha-Synuclein and Rotenone Interaction. Biomolecules 2013; 3: 703-732
  • 103 Yuan YH, Yan WF, Sun JD. et al. The molecular mechanism of rotenone-induced alpha-synuclein aggregation: Emphasizing the role of the calcium/GSK3beta pathway. Toxicol Lett 2015; 233: 163-171
  • 104 Chorfa A, Lazizzera C, Betemps D et al. A variety of pesticides trigger in vitro alpha-synuclein accumulation, a key event in Parkinson’s disease. Arch Toxicol 2014
  • 105 Schapira AH. Disease modification in Parkinson’s disease. Lancet Neurol 2004; 3: 362-368
  • 106 Mizuno Y, Ohta S, Tanaka M. et al. Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochem Biophys Res Commun 1989; 163: 1450-1455
  • 107 Franco-Iborra S, Vila M, Perier C. The parkinson disease mitochondrial hypothesis: where are ee at?. Neuroscientist 2016; 22: 266-277
  • 108 Jenner P. Oxidative stress in Parkinson’s disease. Ann Neurol 2003; 53 (Suppl. 03) S26-S36 discussion S36–S38
  • 109 McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord 2004; 10 (Suppl. 01) S3-S7
  • 110 Gao HM, Hong JS, Zhang W. et al. Synergistic dopaminergic neurotoxicity of the pesticide rotenone and inflammogen lipopolysaccharide: relevance to the etiology of Parkinson’s disease. J Neurosci 2003; 23: 1228-1236
  • 111 Alvarez-Erviti L, Seow Y, Schapira AH. et al. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol Dis 2011; 42: 360-367
  • 112 Hasegawa T, Konno M, Baba T. et al. The AAA-ATPase VPS4 regulates extracellular secretion and lysosomal targeting of alpha-synuclein. PLoS One 2011; 6: e29460
  • 113 Pan-Montojo F, Schwarz M, Winkler C. et al. Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice. Sci Rep 2012; 2: 898
  • 114 Luk KC, Kehm V, Carroll J. et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 2012; 338: 949-953
  • 115 Lee HJ, Suk JE, Bae EJ. et al. Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. Int J Biochem Cell Biol 2008; 40: 1835-1849
  • 116 Angot E, Steiner JA, Lema Tome CM. et al. Alpha-synuclein cell-to-cell transfer and seeding in grafted dopaminergic neurons in vivo. PLoS One 2012; 7: e39465
  • 117 Braidy N, Gai WP, Xu YH. et al. Alpha-synuclein transmission and mitochondrial toxicity in primary human foetal enteric neurons in vitro. Neurotox Res 2014; 25: 170-182
  • 118 Esteves AR, Arduino DM, Silva DF. et al. Mitochondrial Dysfunction: The Road to Alpha-Synuclein Oligomerization in PD. Parkinsons Dis 2011; 2011: 693761
  • 119 Choi WS, Palmiter RD, Xia Z. Loss of mitochondrial complex I activity potentiates dopamine neuron death induced by microtubule dysfunction in a Parkinson’s disease model. J Cell Biol 2011; 192: 873-882
  • 120 Klegeris A, Giasson BI, Zhang H. et al. Alpha-synuclein and its disease-causing mutants induce ICAM-1 and IL-6 in human astrocytes and astrocytoma cells. FASEB J 2006; 20: 2000-2008
  • 121 McGeer PL, McGeer EG. The alpha-synuclein burden hypothesis of Parkinson disease and its relationship to Alzheimer disease. Exp Neurol 2008; 212: 235-238
  • 122 Hunot S, Dugas N, Faucheux B. et al. FcepsilonRII/CD23 is expressed in Parkinson’s disease and induces, in vitro, production of nitric oxide and tumor necrosis factor-alpha in glial cells. J Neurosci 1999; 19: 3440-3447
  • 123 Dickson DW, Braak H, Duda JE. et al. Neuropathological assessment of Parkinson’s disease: refining the diagnostic criteria. Lancet Neurol 2009; 8: 1150-1157
  • 124 Jellinger KA. Neuropathobiology of non-motor symptoms in Parkinson disease. J Neural Transm (Vienna) 2015; 122: 1429-1440
  • 125 Sauerbier A, Jenner P, Todorova A. et al. Non motor subtypes and Parkinson’s disease. Parkinsonism Relat Disord 2016; 22 (Suppl. 01) S41-S46
  • 126 Desplats P, Lee HJ, Bae EJ. et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA 2009; 106: 13010-13015
  • 127 Danzer KM, Kranich LR, Ruf WP. et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener 2012; 7: 42
  • 128 Brundin P, Li JY, Holton JL. et al. Research in motion: the enigma of Parkinson’s disease pathology spread. Nat Rev Neurosci 2008; 9: 741-745
  • 129 Li JY, Englund E, Holton JL. et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med 2008; 14: 501-503
  • 130 Kordower JH, Brundin P. Lewy body pathology in long-term fetal nigral transplants: is Parkinson’s disease transmitted from one neural system to another?. Neuropsychopharmacology 2009; 34: 254
  • 131 Ling EA, Wong WC, Yick TY. et al. Ultrastructural changes in the dorsal motor nucleus of monkey following bilateral cervical vagotomy. J Neurocytol 1986; 15: 1-15
  • 132 Holmqvist S, Chutna O, Bousset L. et al. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol 2014; 128: 805-820
  • 133 Hardy J. Expression of normal sequence pathogenic proteins for neurodegenerative disease contributes to disease risk: ’permissive templating’ as a general mechanism underlying neurodegeneration. Biochem Soc Trans 2005; 33: 578-581
  • 134 Svensson E, Horvath-Puho E, Thomsen RW. et al. Vagotomy and subsequent risk of Parkinson’s disease. Ann Neurol 2015; 78: 522-529

Zoom Image
Fig. 1 a Schematic drawing of the brain and the gastrointestinal system with a dual-hit of the olfactory system and the gastrointestinal tract via inhalation/ingestion (red thread) with rostrocranial ascension of Parkinson’s pathology (red arrows) via sympathetic and parasympathetic nervous system (vagus nerve as green lines) reaching the central nervous system. Manifestation of non-motor symptoms with corresponding Braak stage (black arrows to corresponding anatomical structure). b Histologic intestinal section with A: mucosa, B: submucosa, c ring muscle, d longitudinal muscle and the enteric nervous system with E: Meissner’s plexus and F: Auerbach’s plexus. c α-synuclein inclusion similar to Lewy body (appearing red by immunohistochemical staining after proteinase K digestion) as Parkinson pathology.