involvement of the peripheral nervous system in

12
REVIEW Involvement of the peripheral nervous system in synucleinopathies, tauopathies and other neurodegenerative proteinopathies of the brain Koichi Wakabayashi Fumiaki Mori Kunikazu Tanji Satoshi Orimo Hitoshi Takahashi Received: 25 March 2010 / Revised: 29 May 2010 / Accepted: 29 May 2010 / Published online: 9 June 2010 Ó Springer-Verlag 2010 Abstract Involvement of the peripheral nervous system (PNS) is relatively common in some neurodegenerative proteinopathies of the brain and may be pathogenetically and diagnostically important. In Parkinson’s disease, neu- ronal a-synuclein aggregates are distributed throughout the nervous system, including the central nervous system (CNS), sympathetic ganglia, enteric nervous system, car- diac and pelvic plexuses, submandibular gland, adrenal medulla and skin. The pathological process may target the PNS and CNS at the same time. In multiple system atro- phy, numerous glial cytoplasmic inclusions composed of filamentous a-synuclein are widely distributed in the CNS, while a-synuclein accumulation is minimal in the sympa- thetic ganglia and is restricted to neurons. Neurofibrillary tangles can occur in the sympathetic and spinal ganglia in tauopathy, although they appear to develop independently of cerebral Alzheimer’s disease pathology. In amyotrophic lateral sclerosis, neuronal loss with TDP-43-positive neu- ronal cytoplasmic inclusions in the spinal ganglia is more frequent than previously thought. Peripheral ganglia and visceral organs are also involved in polyglutamine dis- eases. Further elucidation and characterization of PNS lesions will have implications for intravital biopsy diagnosis in neurodegenerative proteinopathy, particularly in Parkinson’s disease. Keywords a-Synuclein Á Enteric nervous system Á Lewy body Á Parkinson’s disease Á Tauopathy Á TDP-43 Introduction Common cellular and molecular mechanisms including protein aggregation and inclusion body formation are involved in many neurodegenerative diseases [82, 135]. a-Synuclein is a major component of Lewy bodies (LBs) in Parkinson’s disease (PD) and dementia with LBs (DLB) [176] as well as in neuronal and glial cytoplasmic inclu- sions (GCIs) in multiple system atrophy (MSA) [170]. Tau is a principal component of neurofibrillary and glial tangles in tauopathies [77]. Expanded polyglutamine repeats are found in intranuclear and cytoplasmic inclusions in hereditary neurodegeneration, including Huntington’s disease [178]. Recently, TDP-43 was identified as a com- ponent of ubiquitinated inclusions in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration [8, 106]. PD is traditionally considered as a movement disorder with hallmark lesions in the brainstem pigmented nuclei (substantia nigra and locus ceruleus). However, accumu- lating evidence suggests that non-motor complications (cognitive, psychiatric, autonomic, sleep and sensory disorders) are also common in PD [86, 91]. Pathological changes occur in widespread regions of the central and peripheral nervous systems (PNS) in this disease [21, 40, 41]. Furthermore, primary glial involvement (‘‘gliodegen- eration’’) can be observed in a-synucleinopathy [29, 177] as well as in tauopathy [31, 58]. K. Wakabayashi (&) Á F. Mori Á K. Tanji Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan e-mail: [email protected] S. Orimo Department of Neurology, Kanto Central Hospital, Tokyo, Japan H. Takahashi Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan 123 Acta Neuropathol (2010) 120:1–12 DOI 10.1007/s00401-010-0706-x

Upload: others

Post on 08-May-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Involvement of the peripheral nervous system in

REVIEW

Involvement of the peripheral nervous systemin synucleinopathies, tauopathies and otherneurodegenerative proteinopathies of the brain

Koichi Wakabayashi • Fumiaki Mori •

Kunikazu Tanji • Satoshi Orimo • Hitoshi Takahashi

Received: 25 March 2010 / Revised: 29 May 2010 / Accepted: 29 May 2010 / Published online: 9 June 2010

� Springer-Verlag 2010

Abstract Involvement of the peripheral nervous system

(PNS) is relatively common in some neurodegenerative

proteinopathies of the brain and may be pathogenetically

and diagnostically important. In Parkinson’s disease, neu-

ronal a-synuclein aggregates are distributed throughout the

nervous system, including the central nervous system

(CNS), sympathetic ganglia, enteric nervous system, car-

diac and pelvic plexuses, submandibular gland, adrenal

medulla and skin. The pathological process may target the

PNS and CNS at the same time. In multiple system atro-

phy, numerous glial cytoplasmic inclusions composed of

filamentous a-synuclein are widely distributed in the CNS,

while a-synuclein accumulation is minimal in the sympa-

thetic ganglia and is restricted to neurons. Neurofibrillary

tangles can occur in the sympathetic and spinal ganglia in

tauopathy, although they appear to develop independently

of cerebral Alzheimer’s disease pathology. In amyotrophic

lateral sclerosis, neuronal loss with TDP-43-positive neu-

ronal cytoplasmic inclusions in the spinal ganglia is more

frequent than previously thought. Peripheral ganglia and

visceral organs are also involved in polyglutamine dis-

eases. Further elucidation and characterization of PNS

lesions will have implications for intravital biopsy

diagnosis in neurodegenerative proteinopathy, particularly

in Parkinson’s disease.

Keywords a-Synuclein � Enteric nervous system �Lewy body � Parkinson’s disease � Tauopathy � TDP-43

Introduction

Common cellular and molecular mechanisms including

protein aggregation and inclusion body formation are

involved in many neurodegenerative diseases [82, 135].

a-Synuclein is a major component of Lewy bodies (LBs) in

Parkinson’s disease (PD) and dementia with LBs (DLB)

[176] as well as in neuronal and glial cytoplasmic inclu-

sions (GCIs) in multiple system atrophy (MSA) [170]. Tau

is a principal component of neurofibrillary and glial tangles

in tauopathies [77]. Expanded polyglutamine repeats are

found in intranuclear and cytoplasmic inclusions in

hereditary neurodegeneration, including Huntington’s

disease [178]. Recently, TDP-43 was identified as a com-

ponent of ubiquitinated inclusions in amyotrophic lateral

sclerosis (ALS) and frontotemporal lobar degeneration

[8, 106].

PD is traditionally considered as a movement disorder

with hallmark lesions in the brainstem pigmented nuclei

(substantia nigra and locus ceruleus). However, accumu-

lating evidence suggests that non-motor complications

(cognitive, psychiatric, autonomic, sleep and sensory

disorders) are also common in PD [86, 91]. Pathological

changes occur in widespread regions of the central and

peripheral nervous systems (PNS) in this disease [21, 40,

41]. Furthermore, primary glial involvement (‘‘gliodegen-

eration’’) can be observed in a-synucleinopathy [29, 177]

as well as in tauopathy [31, 58].

K. Wakabayashi (&) � F. Mori � K. Tanji

Department of Neuropathology, Institute of Brain Science,

Hirosaki University Graduate School of Medicine,

5 Zaifu-cho, Hirosaki 036-8562, Japan

e-mail: [email protected]

S. Orimo

Department of Neurology, Kanto Central Hospital,

Tokyo, Japan

H. Takahashi

Department of Pathology, Brain Research Institute,

University of Niigata, Niigata, Japan

123

Acta Neuropathol (2010) 120:1–12

DOI 10.1007/s00401-010-0706-x

Page 2: Involvement of the peripheral nervous system in

Inclusion bodies are considered to be related to neuronal

degeneration, which does not imply that the inclusions are

always fatal to the neurons [10, 158]. Recent studies have

suggested that oligomers and protofibrils are cytotoxic, and

that inclusions may represent a cytoprotective mechanism

in neurodegenerative proteinopathy [43, 135, 160]. How-

ever, the number of LBs in patients with mild to moderate

loss of neurons in the substantia nigra is higher than in

those with severe neuronal depletion, thereby suggesting

that LB-containing neurons may be dying neurons [176].

The present article reviews neuropathological data

concerning the PNS involvement in neurodegenerative

disorders of the brain. Particular focus is given to abnormal

protein accumulation and inclusion body formation outside

the central nervous system (CNS).

Parkinson’s disease and dementia with Lewy bodies

The histological hallmark of PD and DLB is neuronal

a-synuclein aggregates called LBs and Lewy neurites. To

date, more than 70 molecules have been identified in LBs

[176], in which phosphorylated a-synuclein is a major

constituent [49]. It is now known that the substantia nigra is

not the only or the first brain region involved in PD [34].

Braak et al. [22] proposed a pathological staging scheme

for PD, in which early a-synuclein pathology is present in

the dorsal vagal nucleus and in the olfactory bulb. This

staging system characterizes a progression from the dorsal

vagal nucleus (stage 1), through the pontine tegmentum

(stage 2), into the midbrain and neostriatum (stage 3), and

then the basal prosencephalon and mesocortex (stage 4),

and finally through the neocortex (stages 5 and 6) [17, 22,

102]. Braak PD stages 1–3 correspond to incidental LB

disease, which is considered to represent presymptomatic

PD and/or DLB [1, 11, 35, 39, 134]. Saito et al. [136] also

proposed a pathological staging scheme for the progression

of LB disease (PD and DLB). Accumulation of a-synuclein

is also found both in astrocytes and oligodendrocytes in the

brain of patients with PD and DLB [25, 166].

LBs are widely distributed in the brain of patients with

PD [114] and DLB [80] mentioned above. a-Synuclein

abnormality is found in 10% of pigmented neurons in the

substantia nigra and more than 50% of those in the locus

ceruleus [101]. LBs are also found in the spinal cord,

including the intermediolateral nucleus, dorsal horn and

sacral autonomic nucleus [12, 14, 24, 76, 115, 125, 168].

Except for olfactory structures and spinal dorsal horn,

sensory components of the nervous system are relatively

spared. LBs are also found in the PNS even in the early

stage of PD (described below). The widespread distribution

of Lewy pathology may correspond to a variety of motor

and non-motor symptoms of PD [40, 91].

Sympathetic ganglia

LBs in the sympathetic ganglia were first observed by

Herzog [57]. In the ganglia, LBs occur mainly in the nerve

cell processes (Fig. 1a, b), and the majority of LB-con-

taining processes are unmyelinated axons [45, 171].

Although paravertebral and celiac sympathetic ganglia are

the predilection sites for LBs [37, 169], no LBs are found

in the dorsal root ganglia.

The degenerative process of the sympathetic ganglia has

been classified into three categories [117]. In the early

stage, a few LBs are observed in the ganglia but the

number of neurons and immunoreactivity for tyrosine

hydroxylase (TH), a rate-limiting enzyme of the catechol-

amine synthesizing pathway, is well preserved. In the

middle stage, many LBs are found in the ganglia but the

number of neurons appears to be normal on H&E stained

sections. Interestingly, a significant number of neuronal

somata (about 20–30% of neurons) is TH-immunonegative.

In the advanced stage, there is apparent neuronal loss in the

ganglia on H&E stained or TH immunostained sections.

The number of LBs is decreased compared with the middle

stage [117].

Parasympathetic nervous system

The largest source of preganglionic parasympathetic fibers

is the dorsal vagal nucleus, which supplies all the thoracic

and abdominal viscera except those in the pelvic region

[128]. In the cranial region, the postganglionic fibers derive

from ciliary, pterygopalatine, submandibular and otic

ganglia [128]. The occurrence of LBs has been reported in

the ciliary [7] and submandibular ganglia [153]. Recently,

Del Tredici et al. [33] have shown that Lewy pathology

occurs in the submandibular glands and in anatomically

related structures (superior cervical ganglia, cervical sym-

pathetic trunk and peripheral vagal nerve) in PD (9/9 cases)

and incidental LB disease (2/3 cases) but not in MSA or

controls. The presence of Lewy pathology in the autonomic

nerves and/or ganglia innervating the submandibular

glands might be related to the reduced salivary secretion

that accompanies PD [33].

Enteric nervous system

Gastrointestinal dysfunction is a common feature of PD

[1, 27, 97, 129], and constipation is frequent in DLB [59].

The occurrence of LBs in the enteric nervous system (ENS)

of PD patients was first reported by Qualman et al. [133]

who recognized the inclusions in the myenteric plexus of

the esophagus of 1 patient and the colon of another out of

22 patients with PD. They also found similar inclusions in

the esophagus in two out of eight patients with esophageal

2 Acta Neuropathol (2010) 120:1–12

123

Page 3: Involvement of the peripheral nervous system in

achalasia. Kupsky et al. [85] reported the occurrence of

LBs in the myenteric and submucosal plexuses of the

surgically resected colon and biopsied rectum of a patient

with PD and megacolon. Wakabayashi et al. [175] dem-

onstrated that LBs were distributed widely in the myenteric

and submucosal plexuses from the upper esophagus to the

Fig. 1 Lewy pathology in the peripheral nervous system in Parkin-

son’s disease. a Intracytoplasmic (arrows) and intraneuritic Lewy

bodies (arrowheads) in the sympathetic ganglia. b a-Synuclein-

immunoreactive neuritic structures in the sympathetic ganglia.

c Lewy body in the submucosal plexus of the rectum (arrowhead).

d Lewy bodies in the myenteric plexus of the lower esophagus.

e, f Lewy bodies in the cardiac sympathetic nerves. g Lewy bodies in

the adrenal medulla (arrowheads). h a-Synuclein-immunoreactive

neuritic structures in the subcutaneous tissue (arrowheads). a, c, ghematoxylin–eosin, b, d, f, h a-synuclein immunostain, e tyrosine

hydroxylase immunostain. Bars 20 lm

Acta Neuropathol (2010) 120:1–12 3

123

Page 4: Involvement of the peripheral nervous system in

rectum and were most frequent and numerous in the

myenteric plexus of the lower esophagus (Fig. 1c, d).

Recently, Beach et al. [12] have shown that there is a

rostrocaudal gradient of decreasing frequency and density

of phosphorylated a-synuclein histopathology in the gas-

trointestinal tract in LB disorders. The submandibular

gland and lower esophagus have the greatest involvement

and the colon and rectum the lowest.

A similarity has been noticed between the distribution of

LBs and that of monoaminergic neurons in the CNS [114].

In addition, intrinsic neurons immunoreactive for TH exist

in the human ENS [172]. Interestingly, these neurons are

most frequent and numerous in the myenteric plexus of the

lower esophagus, which is the site where most LBs occur in

PD [12, 175]. However, LB-containing TH-immunoreac-

tive neurons have been rarely encountered in PD. In the

gastrointestinal tract, most LBs were found in the vasoac-

tive intestinal polypeptide (VIP)-immunoreactive processes

[173]. The majority of VIP-immunoreactive processes in

the ENS are intrinsic in origin. In addition, VIP is a cho-

linergic co-transmitter in the intrinsic innervation of the

human gastrointestinal tract [6]. However, there was no

significant decrease in the number of cholinergic or VIP

neurons in PD [145] as well as in MPTP-treated mice and

monkeys [5, 28]. The number of TH-immunoreactive

neurons has been reported to be relatively preserved in PD

patients [145] and decreased in MPTP monkeys [28]. In

contrast, the number of dopaminergic neurons in the

colonic myenteric plexus was markedly reduced in patients

with advanced PD [145]. Loss of dopaminergic neurons

was also found in MPTP mice [5]. The discrepancy

between the frequency of LBs and the severity of VIP

neuron loss may imply that Lewy neurites cause dysfunc-

tional axonal transport but do not cause neuronal cell death.

In the CNS, early sites of Lewy pathology are the

olfactory bulb and dorsal vagal nucleus [22, 34, 141].

Several investigators proposed that a neurotropic pathogen

enters the brain via two routes (1) anterogradely, via

olfactory pathway; and (2) retrogradely, via gastric nerve

plexus and preganglionic nerve fibers of the dorsal vagal

nucleus [20, 23, 55, 88]. Abbott et al. [1] have shown that

constipation could be one of the earliest markers of the

beginning of PD processes. This is supported by the find-

ings that LBs can occur in the ENS in elderly individuals

without LBs in the CNS [175] and that a-synuclein

accumulation has been seen in the gastric plexus in Braak

PD stage 2 [20]. Interestingly, a-synuclein aggregates in

the ENS precede the CNS and cardiac autonomic abnor-

malities in transgenic mice containing PD-associated

a-synuclein gene mutations [84]. It is possible that the

pathological process of PD targets the ENS even before

lower brainstem nuclei become involved. Lebouvier et al.

[87] performed routine colonic biopsies in five PD patients

complaining of constipation and five age-matched control

patients, and immunohistochemical staining revealed

phosphorylated a-synuclein-immunoreactive neurites in

the submucosal plexus in four out of five PD patients.

Colonic biopsies may be useful to detect LB-type degen-

eration in PD.

Recently, several studies have shown that PD patients

who had long-term survival of transplanted fetal nigral

tissue (11–16 years) developed LBs in grafted neurons [79,

89]. This suggests that Lewy pathology could be induced in

cells by prion-like transmissible agents [2, 23, 116]. The

mucosa of foregut receives the efferent fibers from the

dorsal vagal nucleus. Moreover, a link between Helico-

bacter pylori infection and PD has been suggested [3, 42].

If the postulated pathogen could penetrate the gastric

mucosa, it ascends retrogradely in preganglionic para-

sympathetic fibers of the vagus nerve to the medulla

oblongata [20]. This possibility deserves further

investigation.

Heart

The sympathetic neurons in the cervical and upper thoracic

ganglia are the origin of the postganglionic fibers inner-

vating the heart, namely, the cardiac sympathetic nerves.

The heart also receives parasympathetic cholinergic fibers

from the dorsal vagal nucleus. The density of sympathetic

nerves is six times higher than that of cholinergic fibers in

the anterior wall of the left ventricle [73]. In PD, LBs are

found in the ganglia located in the interatrial groove (atrial

ganglia) and in the nerve fibers around the coronary arteries

and in the myocardium (Fig. 1e, f) [169]. LBs are also

found in the heart of patients with incidental LB disease

[62].

Reduced cardiac uptake of meta-iodobenzylguanidine

(MIBG), a physiological analog of noradrenaline, on [123I]

MIBG myocardial scintigraphy has been reported in

patients with LB disorders [118]. Decreased cardiac uptake

of MIBG in LB disease reflects loss of cardiac sympathetic

nerves in the ventricular wall [4], which precedes the neu-

ronal loss in the sympathetic ganglia [117]. The depletion

of sympathetic nerves involves not only the ventricles, but

also the atria and the conduction system [51]. Accumula-

tion of a-synuclein in the distal axons of the cardiac

sympathetic nervous system precedes that in neuronal

somata or neurites in the paravertebral sympathetic ganglia

and heralds centripetal degeneration of the cardiac sym-

pathetic nerves in PD [123]. Cardiac sympathetic

denervation begins in the early disease process of PD [51,

122]. Cardiac sympathetic denervation and a-synuclein

pathology increase with disease duration and severity [48].

Similar degeneration occurs in PARK4 (familial PD linked

to SNCA duplication) [51, 124], but not in PARK2 (familial

4 Acta Neuropathol (2010) 120:1–12

123

Page 5: Involvement of the peripheral nervous system in

PD linked to parkin mutations without LBs) [119].

Reduced cardiac uptake of MIBG is a potential biomarker

for the presence of LBs [118]. Interestingly, reduced car-

diac uptake of MIBG is also reported in idiopathic REM

sleep behavior disorder [99]. However, it is uncertain

whether idiopathic REM sleep behavior disorder is a forme

fruste of LB disease or not.

Recently, Miki et al. [95] reported the presence of LBs

and Lewy neurites solely in the cardiac sympathetic nerves

and stellate ganglia of a non-parkinsonian young adult at

autopsy. This singular finding is not consistent with the

‘‘dual-hit’’ hypothesis mentioned above [55].

Pelvic organs

In the pelvic plexus, LBs are found in the ganglia near the

urinary bladder and accessory male genital organs [169] as

well as in the ovary [12]. Wakabayashi et al. [169] have

reported that LBs were found in the pelvic plexus in 11 out

of 30 patients with PD. Minguez-Castellanos et al. [98]

examined surgical specimens from 100 patients without

known neurodegenerative disorders, who ranged in age

from 44 to 84 years and reported that a-synuclein aggre-

gates were found in the abdominopelvic autonomic

plexuses in 9% of the whole sample but were more com-

mon in vesicoprostatic (26.1%) than in digestive tract

specimens (3.9%). Although the most widely accepted

mechanisms of urinary and sexual dysfunctions in PD are

thought to be related to nerve cell loss in the CNS [13, 94],

Lewy pathology in the pelvic plexus may also play a role in

these conditions.

Adrenal gland

LBs occur in the nerve plexuses around the adrenal gland

and in the ganglion cells in the adrenal medulla (Fig. 1g)

[169, 174]. These ganglion cells are observed singly or in

small groups and are sympathetic in nature. Fumimura et al.

[50] reported that the accumulation of a-synuclein was

found in the adrenal gland in 207 (26.4%) of 783 cases

ranging in age from 48 to 104 years, with 1 case solely in the

adrenal gland. LBs in the adrenal medulla are easily dif-

ferentiated from adrenal bodies [36] observed in the

noradrenaline-containing chromaffin cells because the latter

are positive for periodic acid-Schiff but immunonegative for

ubiquitin and a-synuclein [149]. Interestingly, the number

of adrenal bodies in LB disease is significantly higher than

that in controls, and correlated with disease duration [149].

Other visceral organs

Recently, Beach et al. [12] have shown that Lewy pathol-

ogy occurs in multiple visceral organs including the

respiratory tract (larynx and bronchus), endocrine system

(pancreas and parathyroid gland) and genitourinary tract in

Lewy body disorders.

Skin

Ikemura et al. [61] prospectively examined skin samples

from the abdominal wall and upper limb in 279 consecu-

tively autopsied patients ranging in age from 52 to

104 years (mean 80.8 years) and showed that a-synuclein

immunoreactivity was present in the dermis in 20 (23.5%)

of 85 patients with Lewy pathology in the CNS. The latter

authors also retrospectively examined the abdominal skin

in 142 patients with Lewy pathology in the CNS and

demonstrated that the sensitivity of Lewy pathology in the

skin was 70% in PD and 40% in DLB. Dabby et al. [30]

performed 4-mm punch biopsies from the lower limb in 22

patients with PD and 19 controls using anti-PGP 9.5 anti-

body as a panneuronal marker. They reported that

cutaneous autonomic innervation was decreased in PD

compared to controls and that the denervation scores were

significantly correlated with disease duration in PD.

Nolano et al. [113] also performed 3-mm punch skin

biopsies from the fingertip, thigh and distal leg in 18

patients with PD and 30 controls and demonstrated

significant loss of epidermal nerve fibers in PD patients.

Recently, Miki et al. [96] immunohistochemically exam-

ined cutaneous tissue obtained from 6-mm punch biopsies

in 20 clinically diagnosed PD patients using phosphory-

lated a-synuclein immunohistochemistry. Abnormal

a-synuclein accumulation was found in the dermis of the

thoracic wall but not in the lower limb in 2 (10%) of 20

patients with PD (Fig. 1h). This suggests that skin biopsy

may not be suitable for the diagnosis of PD.

Pure autonomic failure and Lewy body dysphagia

From clinical and neuropathological viewpoints, primary

autonomic failure is divided into three types: pure auto-

nomic failure (PAF) without other neurological conditions,

autonomic failure with PD and autonomic failure with

MSA. In PAF, the main pathological findings are neuronal

loss in the intermediolateral column of the spinal cord and

sympathetic ganglia and the occurrence of LBs in the

brainstem pigmented nuclei, sympathetic ganglia, myen-

teric plexus, epicardium, adrenal medulla and urinary

bladder [7, 52, 71, 162]. Pigmented neurons in the sub-

stantia nigra and locus ceruleus are well preserved in PAF.

Cardiac sympathetic denervation has been observed in PAF

[121]. Skin biopsy revealed a-synuclein accumulation in

the skin nerve fibers in a patient with PAF [144]. Kauf-

mann et al. [72] reported two cases present with PAF; one

Acta Neuropathol (2010) 120:1–12 5

123

Page 6: Involvement of the peripheral nervous system in

developed PD 20 years after onset of the disease and the

other developed DLB 4 years later. Similar cases have

been reported by several investigators [67, 182]. PAF is an

extreme variant of LB disorders, in which LB-type

degeneration may begin outside the CNS. Given the dis-

tribution and severity of Lewy pathology, PAF and a

significant proportion of PD may represent a ‘‘bottom-up’’

variant of LB disease, whereas cerebral type LB disease

[81] represents a ‘‘top-down’’ variant of LB disease.

The presence of LBs in autonomic structures could

explain isolated progressive dysphagia without extrapyra-

midal symptoms, i.e., LB dysphagia [66, 83]. Postmortem

examination has shown that neuronal loss with LBs was

chiefly restricted to the dorsal vagal nucleus in patients

with LB dysphagia [66, 83].

It is important to note that PAF and LB dysphagia are not

incompatible with early stages (or an early phase) of PD.

Multiple system atrophy

Multiple system atrophy is a sporadic adult-onset neuro-

degenerative disease, which is characterized by

striatonigral degeneration, olivopontocerebellar atrophy

and preganglionic autonomic lesions in any combination.

The histological hallmark is the presence of GCIs [105,

127] that contain many substances, including ubiquitin,

a-synuclein and p25a [170, 177]. The incidence of GCIs is

correlated with the severity of neuronal loss in the olivo-

pontocerebellar system as well as in the striatonigral

system [126]. Fibrillary inclusions are also found in the

neuronal somata, axons and nucleus [9, 107, 108, 164].

Severe atrophy of the frontal or temporal lobes has also

been reported in some cases of MSA [78, 131, 143]. The

primary motor cortex and spinal anterior horn are also

involved in MSA [148, 155, 161]. Thus, the neuropathol-

ogy of MSA is more extensive than previously thought.

The PNS is also affected in MSA. Filamentous aggre-

gates of a-synuclein are found in neurons in the

sympathetic ganglia [107]. Sone et al. [146] reported that

a-synuclein-immunoreactive neuronal inclusions were

found in the sympathetic ganglia in 11 out of 26 cases with

MSA and that the mean disease duration in cases with

a-synuclein inclusions was significantly longer than those

without. Although a-synuclein immunoreactivity is present

in the cytoplasm of Schwann cells [100], abnormal accu-

mulation of a-synuclein has not been reported in the PNS

glial cells. Sural nerve biopsy from patients with MSA

shows a reduction of unmyelinated fibers (sensory afferent

fibers and postganglionic sympathetic fibers) by 23% [68].

Mild degeneration of cardiac sympathetic nerves can occur

in MSA [120]. Thus, no portion of the nervous system

appears to be spared in MSA.

Tauopathy

Tauopathies comprise a group of neurodegenerative dis-

orders that share accumulation of phosphorylated tau in

selected vulnerable neurons and glial cells in the brain,

including Alzheimer’s disease (AD) [16, 19, 60, 104],

progressive supranuclear palsy (PSP) [64, 147], cortico-

basal degeneration (CBD) [38, 54], argyrophilic grain

disease [18, 44, 138] and Pick disease [77]. The spinal cord

is also involved in the disease process of AD [137], PSP

[63, 70, 75], CBD [65] and Guam parkinsonism-dementia

complex [93].

The occurrence of neurofibrillary tangles (NFTs) in the

PNS was first reported in the upper cervical ganglia in a

76-year-old man by Kawasaki et al. [74], who also dem-

onstrated that the NFTs are ultrastructurally composed of

paired helical filaments. To date, 10 autopsy cases with

NFTs in the paravertebral and prevertebral (celiac) sym-

pathetic ganglia have been reported [15, 74, 154, 163, 165].

All of these patients were over the age of 60 (age range

61–96 years; mean 78.8 years) and concomitant AD

lesions (many NFTs and senile plaques in the brain) were

found in only three cases. In addition, no NFTs were found

in the sympathetic ganglia of 27 patients with AD [140]

and in 20 patients with AD [165]. Shankle et al. [142]

examined the myenteric plexuses of 18 patients with AD

using an anti-tau antibody (Alz-50) and found no NFTs in

the plexuses. These findings suggest that NFTs in the

sympathetic ganglia develop independently of AD.

Nishimura et al. [112] reported that NFTs were found in

the spinal ganglia in two out of five patients with PSP. The

two patients’ ages were 76 and 71 years at death. Although

NFTs present in the CNS in PSP patients are ultrastructur-

ally composed of 15-nm-wide straight tubules [152, 157],

the NFTs observed in the spinal ganglia were composed of

paired helical filaments. However, immunohistochemical

study using antibodies against phosphorylation-dependent

and -independent tau has shown that no NFTs were detected

in the spinal ganglia of 8 patients with PSP and of 20

patients with AD [165]. The occurrence of NFTs in the

spinal ganglia may be unrelated to the occurrence of NFTs

in the CNS.

NFTs have not been reported in the visceral autonomic

nervous system in human subjects. Interestingly, however,

phosphorylated tau is deposited in the myenteric plexus in

aged Fischer 344 rats [130].

TDP-43 proteinopathy

TDP-43 is a major component of ubiquitinated inclusions

in ALS and frontotemporal lobar degeneration with or

without motor neuron disease [8, 106]. Thus, these

6 Acta Neuropathol (2010) 120:1–12

123

Page 7: Involvement of the peripheral nervous system in

neurodegenerative disorders comprise a new disease con-

cept, namely that of ‘‘TDP-43 proteinopathy.’’ TDP-43

immunohistochemistry has revealed overt inclusions of

filamentous structures (skein-like inclusions) or compact,

round morphology (round inclusions) in motor and non-

motor neurons in TDP-43 proteinopathy [32, 103]. In

addition, TDP-43-positive inclusions occur both in oligo-

dendrocytes and astrocytes in a considerably wider area in

the CNS [109–111]. TDP-43 deposited in TDP-43 pro-

teinopathy lesions is phosphorylated [53].

Although an absence of sensory disturbances is a

negative neurological sign in sporadic ALS, occasional

reports describing sensory signs and symptoms have been

published [92, 156]. Brownell et al. [26] have reported

that posterior column lesions were observed in 5 out of 45

cases of sporadic ALS. In addition, rare instances of

sporadic ALS with severe involvement of the posterior

column and spinal sensory ganglia have been reported

[167]. However, some of these lesions have been con-

sidered an incidental complication due to spondylotic

myelopathy or circulatory disturbance. Recent studies

have revealed that abnormal accumulation of TDP-43

occurs in the dorsal root ganglia but not in the peripheral

sympathetic ganglia in patients with sporadic ALS [110].

Nishihira et al. [110] found mild neuronal loss in the

dorsal root ganglia in 10 out of 35 patients with sporadic

ALS, two of whom showed TDP-43-positive neuronal

cytoplasmic inclusions in the ganglia. TDP-43-immuno-

reactive LB-like inclusions have also been described in

the dorsal root ganglia in a patient with ALS after long-

term survival on a respirator [111]. Sensory involvement

may occur with greater frequency in sporadic ALS than

previously thought.

TDP-43 is a nuclear transcription factor and TDP-43

immunohistochemistry shows diffuse nuclear staining in

unaffected neurons. However, in neurons with well-formed

inclusions, such as skein-like and round inclusions, nuclear

TDP-43 immunoreactivity is absent. Recently, Suzuki et al.

[150] performed a quantitative immunohistochemical study

of TDP-43 in the skin from patients with ALS (n = 15) and

control subjects (n = 15). The proportion and optical den-

sity of TDP-43-positive nuclei in the epidermis were

significantly higher in ALS than in controls. In addition,

there was a significant positive relationship between the

nuclear staining and disease duration in ALS patients. TDP-

43 protein in the plasma [46, 47] and cerebrospinal fluid

[69] may be a potential biomarker of TDP-43 proteinopa-

thy, and skin biopsy may be useful for the diagnosis of ALS.

Bunina bodies are small round eosinophilic inclusions

observable in the lower motor neurons in ALS. They are

thought to originate from the endoplasmic reticulum [151,

159]. Piao et al. [132] have reported the presence of Bunina

bodies in 88 out of 102 patients with ALS (86.3%).

However, such inclusions have not hitherto been reported

in the PNS.

Polyglutamine disease

The expansion of a CAG repeat that codes for polyglutamine

is a common gene mutation in hereditary neurodegenerative

disorders [178]. Recent immunohistochemical studies of

human and animal models of polyglutamine diseases have

shown that neuronal and glial nuclear abnormalities extend

to multiple central and peripheral areas of the nervous sys-

tem, far beyond the affected regions that have been reported

in conventional pathological studies [56, 180]. Immuno-

histochemistry for expanded polyglutamine stretches have

demonstrated that neuronal intranuclear inclusions are

present in the dorsal root ganglia and paravertebral and

celiac sympathetic ganglia in spinocerebellar ataxia type 3

(Machado-Joseph disease) [179], but not in dentatorubral-

pallidoluysian atrophy [181] or spinal and bulbar muscular

atrophy [90]. Moreover, formation of polyglutamine inclu-

sions have been reported in non-CNS tissue in a mouse

model of Huntington’s disease [139] and patients with spinal

and bulbar muscular atrophy [90].

Conclusions

Neurodegenerative proteinopathies are multisystem disor-

ders including the PNS. Further elucidation and

characterization of the PNS lesions will provide implica-

tions for intravital biopsy diagnosis in neurodegenerative

proteinopathy, particularly in LB disease. Future studies

are necessary to identify novel biomarkers and proteins for

the diagnosis and for assessing disease progression of

neurodegenerative disorders.

Acknowledgments This work has been supported by Grants-in-Aid

20300123 (K.W.), 20591361 (F.M.) and 20590335 (K.T.) for Scien-

tific Research from the Ministry of Education, Culture, Sports,

Science, and Technology, Japan, a Grant for Hirosaki University

Institutional Research (K.W.), and a Grant-in-Aid for Studies on the

Development of Diagnostic Technique and Therapies for Lewy Body

Disease, the Ministry of Health, Labour and Welfare, Japan (K.W.).

References

1. Abbott RD, Ross GW, Petrovitch H et al (2007) Bowel move-

ment frequency in late-life and incidental Lewy bodies. Mov

Disord 22:1581–1586

2. Aguzzi A (2009) Cell biology: beyond the prion principle.

Nature 459:924–925

3. Altschuler E (1996) Gastric Helicobacter pylori infection as a

cause of idiopathic Parkinson disease and non-arteric anterior

optic ischemic neuropathy. Med Hypotheses 47:413–414

Acta Neuropathol (2010) 120:1–12 7

123

Page 8: Involvement of the peripheral nervous system in

4. Amino T, Orimo S, Itoh Y, Takahashi A, Uchihara T, Mizusawa

H (2005) Profound cardiac sympathetic denervation occurs in

Parkinson disease. Brain Pathol 15:29–34

5. Anderson G, Noorian AR, Taylor G et al (2007) Loss of enteric

dopaminergic neurons and associated changes in colon motility

in an MPTP mouse model of Parkinson’s disease. Exp Neurol

207:4–12

6. Anlauf M, Schafer MK, Eiden L, Weihe E (2003) Chemical

coding of the human gastrointestinal nervous system: choliner-

gic, VIPergic, and catecholaminergic phenotypes. J Comp

Neurol 459:90–111

7. Arai K, Kato N, Kawashima K, Hattori T (2000) Pure autonomic

failure in association with human a-synucleinopathy. Neurosci

Lett 296:171–173

8. Arai T, Hasegawa M, Akiyama H et al (2006) TDP-43 is a

component of ubiquitin-positive tau-negative inclusions in

frontotemporal lobar degeneration and amyotrophic lateral

sclerosis. Biochem Biophys Res Commun 351:602–611

9. Arima K, Ueda K, Sunohara N et al (1998) NACP/a-synuclein

immunoreactivity in fibrillary components of neuronal and oli-

godendroglial cytoplasmic inclusions in the pontine nuclei in

multiple system atrophy. Acta Neuropathol 96:439–444

10. Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S

(2004) Inclusion body formation reduces levels of mutant hun-

tingtin and the risk of neuronal death. Nature 431:805–810

11. Beach TG, Adler CH, Sue LI et al (2008) Reduced striatal

tyrosine hydroxylase in incidental Lewy body disease. Acta

Neuropathol 115:445–451

12. Beach TG, Adler CH, Sue LI et al (2010) Multi-organ distri-

bution of phosphorylated a-synuclein histopathology in subjects

with Lewy body disorders. Acta Neuropathol 119:689–702

13. Blackett H, Walker R, Wood B (2009) Urinary dysfunction in

Parkinson’s disease: a review. Parkinsonism Relat Disord 15:81–87

14. Bloch A, Probst A, Bissig H, Adams H, Tolnay M (2006)

a-Synuclein pathology of the spinal and peripheral autonomic

nervous system in neurologically unimpaired elderly subjects.

Neuropathol Appl Neurobiol 32:284–295

15. Bohl J, Ulbricht D, Steinmetz H (1997) Neurofibrillary tangles

in peripheral autonomic ganglion cells. In: Iqbal K, Winblad B,

Nishimura T, Takeda M, Wisniewski HM (eds) Alzheimer’s

disease: biology. Diagnosis and therapeutics. Wiley, Chichester,

pp 281–287

16. Braak H, Alafuzoff I, Arzberger T et al (2006) Staging of

Alzheimer disease-associated neurofibrillary pathology using

paraffin sections and immunocytochemistry. Acta Neuropathol

112:389–404

17. Braak H, Bohl JR, Muller CM et al (2006) The staging proce-

dure for the inclusion body pathology associated with sporadic

Parkinson’s disease reconsidered. Mov Disord 21:2042–2051

18. Braak H, Braak E (1989) Cortical and subcortical argyrophilic

grains characterize a disease associated with adult onset

dementia. Neuropathol Appl Neurobiol 15:13–26

19. Braak H, Braak E (1991) Neuropathological stageing of

Alzheimer-related changes. Acta Neuropathol 82:239–259

20. Braak H, de Vos RAI, Bohl J, Del Tredici K (2006) Gastric

a-synuclein immunoreactive inclusions in Meissner’s and

Auerbach’s plexuses in cases staged for Parkinson’s disease-

related brain pathology. Neurosci Lett 396:67–72

21. Braak H, Del Tredici K (2008) Nervous system pathology in

sporadic Parkinson’s disease. Neurology 70:1916–1925

22. Braak H, Del Tredici K, Rub U et al (2003) Staging of brain

pathology related to sporadic Parkinson’s disease. Neurobiol

Aging 24:197–211

23. Braak H, Rub U, Gai WP, Del Tredici K (2003) Idiopathic

Parkinson’s disease: possible routes by which vulnerable

neuronal types may be subject to neuroinvasion by an unknown

pathogen. J Neural Transm 110:517–536

24. Braak H, Sastre M, Bohl JR, de Vos RA, Del Tredici K (2007)

Parkinson’s disease: lesions in dorsal horn layer I, involvement

of parasympathetic and sympathetic pre- and postganglionic

neurons. Acta Neuropathol 113:421–429

25. Braak H, Sastre M, Del Tredici K (2007) Development of

a-synuclein immunoreactive astrocytes in the forebrain parallels

stages of intraneuronal pathology in sporadic Parkinson’s dis-

ease. Acta Neuropathol 114:231–241

26. Brownell B, Oppenheimer DR, Hughes JT (1970) The central

nervous system in motor neurone disease. J Neurol Neurosurg

Psychiatry 33:338–357

27. Cersosimo MG, Benarroch EE (2008) Neural control of the

gastrointestinal tract: implications for Parkinson disease. Mov

Disord 23:1065–1075

28. Chaumette T, Lebouvier T, Aubert P et al (2009) Neurochemical

plasticity in the enteric nervous system of a primate animal

model of experimental Parkinsonism. Neurogastroenterol Motil

21:215–222

29. Croisier E, Graeber MB (2006) Glial degeneration and reactive

gliosis in alpha-synucleinopathies: the emerging concept of

primary gliodegeneration. Acta Neuropathol 112:517–530

30. Dabby R, Djaldetti R, Shahmurov M et al (2006) Skin biopsy for

assessment of autonomic denervation in Parkinson’s disease.

J Neural Transm 113:1169–1176

31. Dabir DV, Robinson MB, Swanson E et al (2006) Impaired

glutamate transport in a mouse model of tau pathology in

astrocytes. J Neurosci 26:644–654

32. Davidson Y, Kelley T, Mackenzie IR et al (2007) Ubiquitinated

pathological lesions in frontotemporal lobar degeneration con-

tain the TAR DNA-binding protein, TDP-43. Acta Neuropathol

113:521–533

33. Del Tredici K, Hawkes CH, Ghebremedhin E, Braak H (2010)

Lewy pathology in the submandibular gland of individuals with

incidental Lewy body disease and sporadic Parkinson’s disease.

Acta Neuropathol 119:703–707

34. Del Tredici K, Rub U, De Vos RA, Bohl JR, Braak H (2002)

Where does parkinson disease pathology begin in the brain?

J Neuropathol Exp Neurol 61:413–426

35. DelleDonne A, Klos J, Fujishiro H et al (2008) Incidental Lewy

body disease and preclinical Parkinson’s disease. Arch Neurol

65:1074–1080

36. Den Hartog Jager WA (1970) Histochemistry of adrenal bodies

in Parkinson’s disease. Arch Neurol 23:528–533

37. Den Hartog Jager WA, Bethlem J (1960) The distribution of

Lewy bodies in the central and autonomic nervous systems in

idiopathic paralysis agitans. J Neurol Neurosurg Psychiatry

23:283–290

38. Dickson DW, Bergeron C, Chin SS et al (2002) Office of rare

diseases neuropathologic criteria for corticobasal degeneration.

J Neuropathol Exp Neurol 61:935–946

39. Dickson DW, Fujishiro H, DelleDonne A et al (2008) Evidence

that incidental Lewy body disease is pre-symptomatic Parkin-

son’s disease. Acta Neuropathol 115:437–444

40. Dickson DW, Fujishiro H, Orr C et al (2009) Neuropathology of

non-motor features of Parkinson disease. Parkinsonism Relat

Disord 15(Suppl 3):S1–S5

41. Djaldetti R, Lev N, Melamed E (2009) Lesions outside the CNS

in Parkinson’s disease. Mov Disord 24:793–800

42. Dobbs RJ, Dobbs SM, Weller C et al (2008) Helicobacter

hypothesis for idiopathic Parkinsonism: before and beyond.

Helicobacter 13:309–322

43. Engelender S (2008) Ubiquitination of a-synuclein and

autophagy in Parkinson’s disease. Autophagy 4:372–374

8 Acta Neuropathol (2010) 120:1–12

123

Page 9: Involvement of the peripheral nervous system in

44. Ferrer I, Santpere G, van Leeuwen FW (2008) Argyrophilic

grain disease. Brain 131:1416–1432

45. Forno LS, Norville RL (1976) Ultrastructure of Lewy bodies in

the stellate ganglion. Acta Neuropathol 34:183–197

46. Foulds PG, Davidson Y, Mishra M et al (2009) Plasma phos-

phorylated-TDP-43 protein levels correlate with brain pathology

in frontotemporal lobar degeneration. Acta Neuropathol

118:647–658

47. Foulds P, McAuley E, Gibbons L et al (2008) TDP-43 protein in

plasma may index TDP-43 brain pathology in Alzheimer’s

disease and frontotemporal lobar degeneration. Acta Neuropa-

thol 116:141–146

48. Fujishiro H, Frigerio R, Burnett M et al (2008) Cardiac sym-

pathetic denervation correlates with clinical and pathologic

stages of Parkinson’s disease. Mov Disord 23:1085–1092

49. Fujiwara H, Hasegawa M, Dohmae N et al (2002) a-Synuclein is

phosphorylated in synucleinopathy lesions. Nature Cell Biol

4:160–164

50. Fumimura Y, Ikemura M, Saito Y et al (2007) Analysis of the

adrenal gland is useful for evaluating pathology of the peripheral

autonomic nervous system in Lewy body disease. J Neuropathol

Exp Neurol 66:354–362

51. Ghebremedhin E, Del Tredici K, Langston JW, Braak H (2009)

Diminished tyrosine hydroxylase immunoreactivity in the car-

diac conduction system and myocardium in Parkinson’s disease:

an anatomical study. Acta Neuropathol 118:777–784

52. Hague K, Lento P, Morgello S, Caro S, Kaufmann H (1997) The

distribution of Lewy bodies in pure autonomic failure: autopsy

findings and review of the literature. Acta Neuropathol 94:192–196

53. Hasegawa M, Arai T, Nonaka T et al (2008) Phosphorylated

TDP-43 in frontotemporal lobar degeneration and amyotrophic

lateral sclerosis. Ann Neurol 64:60–70

54. Hattori M, Hashizume Y, Yoshida M et al (2003) Distribution of

astrocytic plaques in the corticobasal degeneration brain and

comparison with tuft-shaped astrocytes in the progressive

supranuclear palsy brain. Acta Neuropathol 106:143–149

55. Hawkes CH, Del Tredici K, Braak H (2007) Parkinson’s disease:

a dual-hit hypothesis. Neuropathol Appl Neurobiol 33:599–614

56. Hayashi Y, Kakita A, Yamada M et al (1998) Hereditary den-

tatorubral-pallidoluysian atrophy: detection of widespread

ubiquitinated neuronal and glial intranuclear inclusions in the

brain. Acta Neuropathol 96:547–552

57. Herzog E (1928) Histopathologische Veranderungen im Sym-

pathicus und ihre Bedeutung. Dtsch Z Nervenheilk 107:75–80

58. Higuchi M, Zhang B, Forman MS et al (2005) Axonal degen-

eration induced by targeted expression of mutant human tau in

oligodendrocytes of transgenic mice that model glial tauopa-

thies. J Neurosci 25:9434–9443

59. Horimoto Y, Matsumoto M, Akatsu H et al (2003) Autonomic

dysfunctions in dementia with Lewy bodies. J Neurol 250:530–

533

60. Ikeda K, Haga C, Akiyama H et al (1992) Coexistence of paired

helical filaments and glial filaments in astrocytic processes

within ghost tangles. Neurosci Lett 148:126–128

61. Ikemura M, Saito Y, Sengoku R et al (2008) Lewy body

pathology involves cutaneous nerves. J Neuropathol Exp Neurol

67:945–953

62. Iwanaga K, Wakabayashi K, Yoshimoto M et al (1999) Lewy

body-type degeneration in cardiac plexus in Parkinson’s and

incidental Lewy body diseases. Neurology 52:1269–1271

63. Iwasaki Y, Yoshida M, Hashizume Y, Hattori M, Aiba I, Sobue

G (2007) Widespread spinal cord involvement in progressive

supranuclear palsy. Neuropathology 27:331–340

64. Iwasaki Y, Yoshida M, Hattori M et al (2004) Distribution of

tuft-shaped astrocytes in the cerebral cortex in progressive

supranuclear palsy. Acta Neuropathol 108:399–405

65. Iwasaki Y, Yoshida M, Hattori M, Hashizume Y, Sobue G

(2005) Widespread spinal cord involvement in corticobasal

degeneration. Acta Neuropathol 109:632–638

66. Jackson M, Lennox G, Balsitis M, Lowe J (1995) Lewy body

dysphagia. J Neurol Neurosurg Psychiatry 58:756–758

67. Kanazawa M, Sanpei K, Toyoshima Y, Kawachi I, Honma Y,

Takahashi H (2007) An autopsy case of dementia with Lewy

bodies showing autonomic failure and dementia as the initial

symptoms. Mov Disord 22:1212–1213

68. Kanda T, Tsukagoshi H, Oda M, Miyamoto K, Tanabe H (1996)

Changes of unmyelinated nerve fibers in sural nerve in amyo-

trophic lateral sclerosis, Parkinson’s disease and multiple system

atrophy. Acta Neuropathol 91:145–154

69. Kasai T, Tokuda T, Ishigami N et al (2009) Increased TDP-43

protein in cerebrospinal fluid of patients with amyotrophic lat-

eral sclerosis. Acta Neuropathol 117:55–62

70. Kato T, Hirano A, Weinberg MN, Jacobs AK (1986) Spinal cord

lesions in progressive supranuclear palsy: some new observa-

tions. Acta Neuropathol 71:11–14

71. Kaufmann H, Hague K, Perl D (2001) Accumulation of alpha-

synuclein in autonomic nerves in pure autonomic failure. Neu-

rology 56:980–981

72. Kaufmann H, Nahm K, Purohit D, Wolfe D (2004) Autonomic

failure as the initial presentation of Parkinson disease and

dementia with Lewy bodies. Neurology 63:1093–1095

73. Kawano H, Okada R, Yano K (2003) Histological study on the

distribution of autonomic nerves in the human heart. Heart

Vessels 18:32–39

74. Kawasaki H, Murayama S, Tomonaga M, Izumiyama N,

Shimada H (1987) Neurofibrillary tangles in human upper

cervical ganglia: morphological study with immunohisto-

chemistry and electron microscopy. Acta Neuropathol 75:156–

159

75. Kikuchi H, Doh-ura K, Kira J, Iwaki T (1999) Preferential

neurodegeneration in the cervical spinal cord of progressive

supranuclear palsy. Acta Neuropathol 97:577–584

76. Klos KJ, Ahlskog JE, Josephs KA et al (2006) a-Synuclein

pathology in the spinal cords of neurologically asymptomatic

aged individuals. Neurology 66:1100–1102

77. Komori T (1999) Tau-positive glial inclusions in progressive

supranuclear palsy, corticobasal degeneration and Pick’s dis-

ease. Brain Pathol 9:663–679

78. Konagaya M, Sakai M, Matsuoka Y, Konagaya Y, Hashizume Y

(1999) Multiple system atrophy with remarkable frontal atrophy.

Acta Neuropathol 97:423–428

79. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW

(2008) Lewy body-like pathology in long-term embryonic nigral

transplants in Parkinson’s disease. Nat Med 14:504–506

80. Kosaka K (1978) Lewy bodies in cerebral cortex. Report of

three cases. Acta Neuropathol 42:127–134

81. Kosaka K, Iseki E, Odawara T, Yamamoto T (1996) Cerebral

type of Lewy body disease. Neuropathology 16:32–35

82. Kovacs GG, Botond G, Budka H (2010) Protein coding of

neurodegenerative dementias: the neuropathological basis of

biomarker diagnostics. Acta Neuropathol 119:389–408

83. Kovari E, Burkhardt K, Lobrinus JA, Bouras C (2007) Lewy

body dysphagia. Acta Neuropathol 114:295–298

84. Kuo YM, Li Z, Jiao Y, Gaborit N et al (2010) Extensive enteric

nervous system abnormalities in mice transgenic for artificial

chromosomes containing Parkinson disease-associated a-syn-

uclein gene mutations precede central nervous system changes.

Hum Mol Genet 19:1633–1650

85. Kupsky WJ, Grimes MM, Sweeting J, Bertsch R, Cote LJ (1987)

Parkinson’s disease and megacolon: concentric hyaline inclu-

sions (Lewy bodies) in enteric ganglion cells. Neurology

37:1253–1255

Acta Neuropathol (2010) 120:1–12 9

123

Page 10: Involvement of the peripheral nervous system in

86. Langston JW (2006) The Parkinson’s complex: Parkinsonism is

just the tip of the iceberg. Ann Neurol 59:591–596

87. Lebouvier T, Chaumette T, Damier P et al (2008) Pathological

lesions in colonic biopsies during Parkinson’s disease. Gut

57:1741–1743

88. Lerner A, Bagic A (2008) Olfactory pathogenesis of idiopathic

Parkinson disease revisited. Mov Disord 23:1076–1084

89. Li JY, Englund E, Holton JL et al (2008) Lewy bodies in grafted

neurons in subjects with Parkinson’s disease suggest host-to-

graft disease propagation. Nat Med 14:501–503

90. Li M, Nakagomi Y, Kobayashi Y et al (1998) Nonneural nuclear

inclusions of androgen receptor protein in spinal and bulbar

muscular atrophy. Am J Pathol 153:695–701

91. Lim SY, Fox SH, Lang AE (2009) Overview of the extranigral

aspects of Parkinson disease. Arch Neurol 66:167–172

92. Manome T, Kobayashi T, Oguchi K, Tsukagoshi H, Fujiwara M

(1980) An autopsy case of amyotrophic lateral sclerosis with

dysesthesia and superficial sensory disturbance. Clin Neurol

(Tokyo) 20:225–232

93. Matsumoto S, Hirano A, Goto S (1990) Spinal cord neurofi-

brillary tangles of Guamanian amyotrophic lateral sclerosis and

parkinsonism-dementia complex: an immunohistochemical

study. Neurology 40:975–979

94. Meco G, Rubino A, Caravona N, Valente M (2008) Sexual

dysfunction in Parkinson’s disease. Parkinsonism Relat Disord

14:451–456

95. Miki Y, Mori F, Wakabayashi K, Kuroda N, Orimo S (2009)

Incidental Lewy body disease restricted to the heart and stellate

ganglia. Mov Disord 24:2299–2301

96. Miki Y, Tomiyama M, Ueno T et al (2010) Clinical availability

of skin biopsy in the diagnosis of Parkinson’s disease. Neurosci

Lett 469:357–359

97. Miller N, Noble E, Jones D, Burn D (2006) Hard to swallow:

dysphagia in Parkinson’s disease. Age Ageing 35:614–618

98. Minguez-Castellanos A, Chamorro CE, Escamilla-Sevilla F et al

(2007) Do a-synuclein aggregates in autonomic plexuses pre-

date Lewy body disorders? Neurology 68:2012–2018

99. Miyamoto T, Miyamoto M, Inoue Y, Usui Y, Suzuki K, Hirata

K (2006) Reduced cardiac 123I-MIBG scintigraphy in idiopathic

REM sleep behavior disorder. Neurology 67:2236–2238

100. Mori F, Inenaga C, Yoshimoto M et al (2002) a-Synuclein

immunoreactivity in normal and neoplastic Schwann cells. Acta

Neuropathol 103:145–151

101. Mori F, Nishie M, Kakita A, Yoshimoto M, Takahashi H, Wa-

kabayashi K (2006) Relationship among a-synuclein

accumulation, dopamine synthesis, and neurodegeneration in

Parkinson disease substantia nigra. J Neuropathol Exp Neurol

65:808–815

102. Mori F, Tanji K, Zhang H, Kakita A, Takahashi H, Wakabayashi

K (2008) a-Synuclein pathology in the neostriatum in Parkin-

son’s disease. Acta Neuropathol 115:453–459

103. Mori F, Tanji K, Zhang HX et al (2008) Maturation process of

TDP-43-positive neuronal cytoplasmic inclusions in amyotro-

phic lateral sclerosis with and without dementia. Acta

Neuropathol 116:193–203

104. Nakano I, Iwatsubo T, Otsuka N et al (1992) Paired helical

filaments in astrocytes: Electron microscopy and immunohisto-

chemistry in a case of atypical Alzheimer’s disease. Acta

Neuropathol 83:228–232

105. Nakazato Y, Yamazaki H, Hirato J, Ishida Y, Yamaguchi H

(1990) Oligodendroglial microtubular tangles in multiple system

atrophy. J Neuropathol Exp Neurol 49:521–530

106. Neumann M, Sampathu DM, Kwong LK et al (2006) Ubiqui-

tinated TDP-43 in frontotemporal lobar degeneration and

amyotrophic lateral sclerosis. Science 314:130–133

107. Nishie M, Mori F, Fujiwara H et al (2004) Accumulation of

phosphorylated a-synuclein in the brain and peripheral ganglia

of patients with multiple system atrophy. Acta Neuropathol

107:292–298

108. Nishie M, Mori F, Yoshimoto M, Takahashi H, Wakabayashi K

(2004) A quantitative investigation of neuronal cytoplasmic and

intranuclear inclusions in the pontine and inferior olivary nuclei

in multiple system atrophy. Neuropathol Appl Neurobiol

30:546–554

109. Nishihira Y, Tan CF, Hoshi Y et al (2009) Sporadic amyotrophic

lateral sclerosis of long duration is associated with relatively

mild TDP-43 pathology. Acta Neuropathol 117:45–53

110. Nishihira Y, Tan CF, Onodera O et al (2008) Sporadic amyo-

trophic lateral sclerosis: two pathological patterns shown by

analysis of distribution of TDP-43-immunoreactive neuronal and

glial cytoplasmic inclusions. Acta Neuropathol 116:169–182

111. Nishihira Y, Tan CF, Toyoshima Y et al (2009) Sporadic

amyotrophic lateral sclerosis: widespread multisystem degen-

eration with TDP-43 pathology in a patient after long-term

survival on a respirator. Neuropathology 29:689–696

112. Nishimura M, Namba Y, Ikeda K, Akiguchi I, Oda M (1993)

Neurofibrillary tangles in the neurons of spinal dorsal root

ganglia of patients with progressive supranuclear palsy. Acta

Neuropathol 85:453–457

113. Nolano M, Provitera V, Estraneo A et al (2008) Sensory deficit

in Parkinson’s disease: evidence of a cutaneous denervation.

Brain 131:1903–1911

114. Ohama E, Ikuta F (1976) Parkinson’s disease. Distribution of

Lewy bodies and monoamine neurons system. Acta Neuropathol

34:311–319

115. Oinas M, Paetau A, Myllykangas L, Notkola IL, Kalimo H,

Polvikoski T (2010) a-Synuclein pathology in the spinal cord

autonomic nuclei associates with a-synuclein pathology in the

brain: a population-based Vantaa 85? study. Acta Neuropathol

119:715–722

116. Olanow CW, Prusiner SB (2009) Is Parkinson’s disease a prion

disorder? Proc Natl Acad Sci USA 106:12571–12572

117. Orimo S, Amino T, Itoh Y et al (2005) Cardiac sympathetic

denervation precedes neuronal loss in the sympathetic ganglia in

Lewy body disease. Acta Neuropathol 109:583–588

118. Orimo S, Amino T, Uchihara T et al (2007) Decreased cardiac

uptake of MIBG is a potential biomarker for the presence of

Lewy bodies. J Neurol 254(Suppl 4):21–28

119. Orimo S, Amino T, Yokochi M et al (2005) Preserved cardiac

sympathetic nerve accounts for normal cardiac uptake of MIBG

in PARK2. Mov Disord 20:1350–1353

120. Orimo S, Kanazawa T, Nakamura A et al (2007) Degeneration

of cardiac sympathetic nerve can occur in multiple system

atrophy. Acta Neuropathol 113:81–86

121. Orimo S, Oka T, Miura H et al (2002) Sympathetic cardiac

denervation in Parkinson’s disease and pure autonomic failure

but not in multiple system atrophy. J Neurol Neurosurg Psy-

chiatry 73:776–777

122. Orimo S, Takahashi A, Uchihara T et al (2007) Degeneration of

cardiac sympathetic nerve begins in the early disease process of

Parkinson’s disease. Brain Pathol 17:24–30

123. Orimo S, Uchihara T, Nakamura A et al (2008) Axonal a-syn-

uclein aggregates herald centripetal degeneration of cardiac

sympathetic nerve in Parkinson’s disease. Brain 131:642–650

124. Orimo S, Uchihara T, Nakamura A et al (2008) Cardiac sym-

pathetic denervation in Parkinson’s disease linked to SNCAduplication. Acta Neuropathol 116:575–577

125. Oyanagi K, Wakabayashi K, Ohama E et al (1990) Lewy bodies

in the lower sacral parasympathetic neurons of a patient with

Parkinson’s disease. Acta Neuropathol 80:558–559

10 Acta Neuropathol (2010) 120:1–12

123

Page 11: Involvement of the peripheral nervous system in

126. Ozawa T, Paviour D, Quinn NP et al (2004) The spectrum

of pathological involvement of the striatonigral and olivo-

pontocerebellar systems in multiple system atrophy:

clinicopathological correlations. Brain 127:2657–2671

127. Papp MI, Kahn JE, Lantos PL (1989) Glial cytoplasmic inclu-

sions in the CNS of patients with multiple system atrophy

(striatonigral degeneration, olivopontocerebellar atrophy and

Shy-Drager syndrome). J Neurol Sci 94:79–100

128. Parent A (1996) Carpenter’s human neuroanatomy, 9th edn.

Williams & Wilkins, Baltimore

129. Pfeiffer RF (2003) Gastrointestinal dysfunction in Parkinson’s

disease. Lancet Neurol 2:107–116

130. Phillips RJ, Walter GC, Ringer BE, Higgs KM, Powley TL

(2009) Alpha-synuclein immunopositive aggregates in the

myenteric plexus of the aging Fischer 344 rat. Exp Neurol

220:109–119

131. Piao YS, Hayashi S, Hasegawa M et al (2001) Co-localization of

a-synuclein and phosphorylated tau in neuronal and glial cyto-

plasmic inclusions in a patient with multiple system atrophy of

long duration. Acta Neuropathol 101:285–293

132. Piao YS, Wakabayashi K, Kakita A et al (2003) Neuropathology

with clinical correlations of sporadic amyotrophic lateral scle-

rosis: 102 autopsy cases examined between 1962 and 2000.

Brain Pathol 13:10–22

133. Qualman SJ, Haupt HM, Yang P, Hamilton SR (1984) Esoph-

ageal Lewy bodies associated with ganglion cell loss in

achalasia Similarity to Parkinson’s disease. Gastroenterology

87:848–856

134. Ross GW, Abbott RD, Petrovitch H et al (2006) Association of

olfactory dysfunction with incidental Lewy bodies. Mov Disord

21:2062–2067

135. Ross CA, Poirier MA (2004) Protein aggregation and neurode-

generative disease. Nat Med 10(Suppl):S10–S17

136. Saito Y, Kawashima A, Ruberu NN et al (2003) Accumulation

of phosphorylated a-synuclein in aging human brain. J Neuro-

pathol Exp Neurol 62:644–654

137. Saito Y, Murayama S (2000) Expression of tau immunoreac-

tivity in the spinal motor neurons of Alzheimer’s disease.

Neurology 55:1727–1729

138. Saito Y, Rubbru NN, Sawabe M et al (2004) Staging of argyr-

ophilic grains: an age-associated tauopathy. J Neuropathol Exp

Neurol 63:911–918

139. Sathasivam K, Hobbs C, Turmaine M et al (1999) Formation of

polyglutamine inclusions in non-CNS tissue. Hum Mol Genet

8:813–822

140. Schmidt RE, Beaudet LN, Plurad SB, Dorsey DA (1997) Axonal

cytoskeletal pathology in aged and diabetic human sympathetic

autonomic ganglia. Brain Res 769:375–383

141. Sengoku R, Saito Y, Ikemura M et al (2008) Incidence and

extent of Lewy body-related a-synucleinopathy in aging human

olfactory bulb. J Neuropathol Exp Neurol 67:1072–1083

142. Shankle WR, Landing BH, Ang SM, Chui H, Villarreal-

Engelhardt G, Zarow C (1993) Studies of the enteric nervous

system in Alzheimer disease and other dementias of the

elderly: enteric neurons in Alzheimer disease. Modern Pathol

6:10–14

143. Shibuya K, Nagatomo H, Iwabuchi K, Inoue M, Yagishita S,

Itoh Y (2000) Asymmetrical temporal lobe atrophy with massive

neuronal inclusions in multiple system atrophy. J Neurol Sci

179:50–58

144. Shishido T, Ikemura M, Obi T et al (2010) a-Synuclein accu-

mulation in skin nerve fibers revealed by skin biopsy in pure

autonomic failure. Neurology 74:608–610

145. Singaram C, Ashraf W, Gaumnitz EA et al (1995) Dopaminergic

defect of enteric nervous system in Parkinson’s disease patients

with chronic constipation. Lancet 346:861–864

146. Sone M, Yoshida M, Hashizume Y, Hishikawa N, Sobue G

(2005) a-Synuclein-immunoreactive structure formation is

enhanced in sympathetic ganglia of patients with multiple sys-

tem atrophy. Acta Neuropathol 110:19–26

147. Steele JC, Richardson JC, Olszewski J (1964) Progressive

supranuclear palsy. A heterogeneous degeneration involving the

brain stem, basal ganglia and cerebellum with vertical gaze and

pseudobulbar palsy, nuchal dystonia and dementia. Arch Neurol

10:333–359

148. Su M, Yoshida Y, Hirata Y, Watahiki Y, Nagata K (2001)

Primary involvement of the motor area in association with the

nigrostriatal pathway in multiple system atrophy: neuropatho-

logical and morphometric evaluations. Acta Neuropathol

101:57–64

149. Sugie M, Goto J, Kawamura M, Ota H (2005) Increased nor-

epinephrine-associated adrenomedullary inclusions in

Parkinson’s disease. Pathol Int 55:130–136

150. Suzuki M, Mikami H, Watanabe T et al (2010) Increased

expression of TDP-43 in the skin of amyotrophic lateral sclerosis.

Acta Neurol Scand (epub ahead of print). doi:10.1111/j.1600-

0404.2010.01321.x

151. Takahashi H, Ohama E, Ikuta F (1991) Are Bunina bodies of

endoplasmic reticulum origin? An ultrastructural study of sub-

thalamic eosinophilic inclusions in a case of atypical motor

neuron disease. Acta Pathol Jpn 41:889–894

152. Takahashi H, Oyanagi K, Takeda S, Hinokuma K, Ikuta F

(1989) Occurrence of 15-nm-wide straight tubules in neocortical

neurons in progressive supranuclear palsy. Acta Neuropathol

79:233–239

153. Takeda S, Yamazaki K, Miyakawa T, Arai H (1993) Parkinson’s

disease with involvement of the parasympathetic ganglia. Acta

Neuropathol 86:397–398

154. Takeda S, Yamazaki K, Miyakawa T, Arai H (1994) Morpho-

logical investigations of the peripheral autonomic nervous

system in Parkinson’s disease, progressive supranuclear palsy

and senile dementia of Alzheimer type. Brain Pathol 4:531

155. Taniwaki T, Minohara M, Hara H, Ohyagi Y, Yamada T, Kira J

(2000) Striatonigral degeneration with motor neuron disease. J

Neurol 247:395–396

156. Tashiro K, Moriwaka F, Matsuura T, Matsumoto A, Hamada T

(1988) Sensory findings in amyotrophic lateral sclerosis. In:

Tsubaki T, Yase Y (eds) Amytrophic lateral sclerosis. Elsevier,

Amsterdam, pp 183–188

157. Tellez-Nagel I, Wisniewski HM (1973) Ultrastructure of neu-

rofibrillary tangles in Steele-Richardson-Olszewski syndrome.

Arch Neurol 29:324–327

158. Terry RD (2000) Do neuronal inclusions kill the cell? J Neural

Transm (Suppl) 59:91–93

159. Tomonaga M, Saito M, Yoshimura M, Shimada H, Tohgi H

(1978) Ultrastructure of the Bunina bodies in anterior horn cells

of amyotrophic lateral sclerosis. Acta Neuropathol 42:81–86

160. Tsika E, Moysidou M, Guo J et al (2010) Distinct region-spe-

cific a-synuclein oligomers in A53T transgenic mice:

Implications for neurodegeneration. J Neurosci 30:3409–3418

161. Tsuchiya K, Ozawa E, Haga C et al (2000) Constant involve-

ment of the Betz cells and pyramidal tract in multiple system

atrophy: a clinicopathological study of seven autopsy cases.

Acta Neuropathol 99:628–636

162. van Ingelghem E, van Zandijcke M, Lammens M (1994) Pure

autonomic failure: a new case with clinical, biochemical, and

necropsy data. J Neurol Neurosurg Psychiatry 57:745–747

163. Wakabayashi K, Furuta A, Takahashi H, Ikuta F (1989)

Occurrence of neurofibrillary tangles in the celiac ganglia. Acta

Neuropathol 78:448

164. Wakabayashi K, Hayashi S, Kakita A et al (1998) Accumulation

of a-synuclein/NACP is a cytopathological feature common to

Acta Neuropathol (2010) 120:1–12 11

123

Page 12: Involvement of the peripheral nervous system in

Lewy body disease and multiple system atrophy. Acta Neuro-

pathol 96:445–452

165. Wakabayashi K, Hayashi S, Morita T, Shibasaki Y, Watanabe

Y, Takahashi H (1999) Neurofibrillary tangles in the peripheral

sympathetic ganglia of non-Alzheimer elderly individuals. Clin

Neuropathol 18:171–175

166. Wakabayashi K, Hayashi S, Yoshimoto M, Kudo H, Takahashi

H (2000) NACP/a-synuclein-positive filamentous inclusions in

astrocytes and oligodendrocytes of Parkinson’s disease brains.

Acta Neuropathol 99:14–20

167. Wakabayashi K, Horikawa Y, Oyake M, Suzuki S, Morita T,

Takahashi H (1998) Sporadic motor neuron disease with severe

sensory neuronopathy. Acta Neuropathol 95:426–430

168. Wakabayashi K, Takahashi H (1997) The intermediolateral

nucleus and Clarke’s column in Parkinson’s disease. Acta

Neuropathol 94:287–289

169. Wakabayashi K, Takahashi H (1997) Neuropathology of auto-

nomic nervous system in Parkinson’s disease. Eur Neurol

38(Suppl 2):2–7

170. Wakabayashi K, Takahashi H (2006) Cellular pathology in

multiple system atrophy. Neuropathology 26:338–345

171. Wakabayashi K, Takahashi H, Obata K, Ikuta F (1992) Immu-

nocytochemical localization of synaptic vesicle-specific protein

in Lewy body-containing neurons in Parkinson’s disease. Neu-

rosci Lett 138:237–240

172. Wakabayashi K, Takahashi H, Ohama E, Ikuta F (1989) Tyro-

sine hydroxylase-immunoreactive intrinsic neurons in the

Auerbach’s and Meissner’s plexuses of humans. Neurosci Lett

96:259–263

173. Wakabayashi K, Takahashi H, Ohama E, Ikuta F (1990) Par-

kinson’s disease: an immunohistochemical study of Lewy body-

containing neurons in the enteric nervous system. Acta Neuro-

pathol 79:581–583

174. Wakabayashi K, Takahashi H, Ohama E, Takeda S, Ikuta F

(1993) Lewy bodies in the visceral autonomic nervous system in

Parkinson’s disease. Adv Neurol 60:609–612

175. Wakabayashi K, Takahashi H, Takeda S, Ohama E, Ikuta F (1988)

Parkinson’s disease: the presence of Lewy bodies in Auerbach’s

and Meissner’s plexuses. Acta Neuropathol 76:217–221

176. Wakabayashi K, Tanji K, Mori F, Takahashi H (2007) The

Lewy body in Parkinson’s disease: molecules implicated in the

formation and degradation of a-synuclein aggregates. Neuro-

pathology 27:494–506

177. Wenning GK, Stefanova N, Jellinger KA, Poewe W, Sch-

lossmacher MG (2008) Multiple system atrophy: a primary

oligodendrogliopathy. Ann Neurol 64:239–246

178. Williams AJ, Paulson HL (2008) Polyglutamine neurodegener-

ation: protein misfolding revisited. Trends Neurosci 31:521–528

179. Yamada M, Hayashi S, Tsuji S, Takahashi H (2001) Involve-

ment of the cerebral cortex and autonomic ganglia in Machado-

Joseph disease. Acta Neuropathol 101:140–144

180. Yamada M, Sato T, Tsuji S, Takahashi H (2008) CAG repeat

disorder models and human neuropathology: similarities and

differences. Acta Neuropathol 115:71–86

181. Yamada M, Wood JD, Shimohata T et al (2001) Widespread

occurrence of intranuclear atrophin-1 accumulation in the cen-

tral nervous system neurons of patients with dentatorubral-

pallidoluysian atrophy. Ann Neurol 49:14–23

182. Yamanaka Y, Asahina M, Hiraga A, Sakakibara R, Oka H,

Hattori T (2007) Over 10 years of isolated autonomic failure

preceding dementia and Parkinsonism in 2 patients with Lewy

body disease. Mov Disord 22:595–597

12 Acta Neuropathol (2010) 120:1–12

123