You are on page 1of 12

REVIEWS

The clinical approach to autonomic failure


in neurological disorders
Eduardo E. Benarroch
Abstract | Central or peripheral neurological disorders can manifest with autonomic failure or autonomic
hyperactivity, which may affect the sympathetic, parasympathetic and/or enteric nervous systems. Disorders
causing autonomic failure can be classified according to the presence or absence of associated neurological
manifestations, such as peripheral neuropathy or parkinsonism, and their temporal profile (acute or subacute,
chronic progressive, static, or episodic). A systematic approach allows focused evaluation to detect treatable,
potentially disabling or life-threatening conditions. Subacute isolated autonomic failure affecting sympathetic,
parasympathetic and enteric nervous system function, in various combinations, occurs in autoimmune
autonomic ganglionopathy, which might be the first manifestation of an underlying neoplasm. Autonomic
failure can be an important feature of several types of peripheral neuropathy, including sensorimotor peripheral
neuropathies, sensory ganglionopathy, and distal painful peripheral neuropathies. Progressive autonomic
failure occurs in neurodegenerative synucleinopathies such as multiple system atrophy and Lewy body
disorders. Autonomic failure may also occur in hereditary leukoencephalopathies or prion disorders. This
Review outlines the clinical approach to patients with generalized autonomic failure, focusing predominantly
on classification and diagnosis, but also touching briefly on treatment and management.

Benarroch, E. E. Nat. Rev. Neurol. advance online publication 27 May 2014; doi:10.1038/nrneurol.2014.88

Introduction Clinical approach to autonomic failure


Autonomic disorders manifest with autonomic failure Disorders associated with autonomic failure can be clas­
or hyperactivity, which may be generalized or focal. sified according to the type and severity of autonomic
Autonomic failure can affect the sympathetic, para­sympa­ manifestations, associated neurological symptoms, and
thetic or enteric nervous systems, either in isolation or in temporal profile (Box 1). The temporal profile of onset
various combinations, and can result from lesions at any and progression has important implications for diag­
level of the CNS or PNS. Sympathetic failure manifests nosis, and for guidance of further laboratory evalua­
primarily with orthostatic hypotension and anhidrosis tion (Figure 1). Isolated autonomic failure of acute or
(absence of sweating), cranial parasympathetic failure subacute onset suggests an immune cause such as auto­
with intolerance to light, dry eyes (xerophthalmia) and immune autonomic ganglionopathy (AAG), including
dry mouth (xerostomia), sacral parasympathetic failure paraneoplastic autonomic neuropathy, or a toxic cause
with urinary retention and erectile dysfunction, and such as effects of medications. Pure autonomic failure
enteric nervous system (ENS) failure with gastroparesis (PAF) refers to the slow development of general­ized
and constipation. In some cases, postural tachycardia auto­nomic failure in the absence of motor or sensory
syndrome is a manifestation of an underlying autonomic symp­toms. Manifestations associated with chronic and
neuropathy. Many symptoms attributed to ‘dysautonomia’ pro­gressive generalized autonomic failure, such as ataxia
in otherwise healthy young patients, such as gastroparesis or parkinsonism, suggest a degenerative cause, typi­
or urinary retention, are rarely associated with objective cally a synuclein­opathy such as multiple system atrophy
evidence of autonomic failure. (MSA), or a Lewy body disorder such as Parkinson
This Review will focus on the clinical approach to disease (PD) or dementia with Lewy bodies (DLB). Auto­
patients with generalized autonomic failure. The degen­ nomic failure can also be a salient feature of various types
erative synucleinopathies, autonomic ganglionopathies of peripheral neuropathy, including distal sensori­motor
and autonomic neuropathies have been reviewed in peripheral neuropathy, painful or ataxic forms of sensory
detail elsewhere.1–5 Rather than providing an exhaustive ganglionopathy, distal painful small fibre ­neuropathy
discussion of the wide spectrum of disorders associated (SFN), and acute motor polyradiculopathy.
with autonomic failure, this article will focus on specific
Department of examples to emphasize clinical cues and outline recent Manifestations and pathophysiology
Neurology, Mayo Clinic,
200 First Street SW, concepts of the underlying pathobiology. Orthostatic hypotension
Rochester, MN 55905, Orthostatic hypotension is defined as a sustained reduc­
USA.
benarroch.eduardo@ Competing interests tion in systolic blood pressure of at least 20 mmHg or
mayo.edu The author declares no competing interests. diastolic blood pressure of 10 mmHg within 3 min of

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION  |  1


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points Box 1 | Neurological causes of autonomic failure


■■ Autonomic failure can occur in isolation, in association with peripheral A. Isolated autonomic failure
neuropathy, or as a manifestation of a neurodegenerative disorder 1. Acute or subacute
■■ Disorders associated with autonomic failure can be classified according to (a) Autoimmune autonomic ganglionopathy
the type and severity of autonomic manifestations, associated neurological (b) Paraneoplastic autonomic neuropathy
symptoms, and temporal profile 2. Progressive
■■ Acute or subacute autonomic failure suggests an autoimmune autonomic (a) Pure autonomic failure
ganglionopathy, which may be paraneoplastic
B. Progressive autonomic failure associated with
■■ In patients with parkinsonism and autonomic failure, early onset and
parkinsonism, ataxia or dementia
progression of orthostatic hypotension or urogenital dysfunction, urinary
1. Multiple system atrophy
incontinence, generalized anhidrosis, and/or laryngeal stridor are highly
2. Lewy body disorders
suggestive of multiple system atrophy
(a) Parkinson disease
■■ Autonomic failure can occur with any type of diabetic or amyloid neuropathy
(b) Dementia with Lewy bodies
■■ Important causes of painful neuropathy associated with autonomic failure
3. Others
include Sjögren syndrome, HIV infections, Fabry disease, and sodium
(a) Familial leukoencephalopathies
channelopathies
(b) Prion disorders
C. Autonomic failure associated with peripheral
neuropathy
standing or head-up tilt.6 Some patients with neuro­ 1. Chronic sensorimotor neuropathies
genic orthostatic hypotension have postprandial hypo­ (a) Diabetes
tension. 7 Manifestations of orthostatic hypotension (b) Amyloidosis
include lightheadedness, blurred vision and neck pain (c) Other metabolic disorders (vitamin B12 deficiency,
(coat hanger distribution) among others; the symptoms uraemia)
(d) Toxic neuropathies
are worse in the morning, after meals, or on exposure
2. Sensory ganglionopathies
to heat.8,9 (a) Sjögren syndrome
Neurogenic orthostatic hypotension is the mani­ (b) Paraneoplastic
festation of impaired sympathetically mediated vaso­ 3. Distal painful neuropathies
constriction of skeletal muscle and mesenteric vessels in (a) Diabetes
response to baroreceptor unloading due to orthostatic (b) Amyloidosis
stress. Neurogenic orthostatic hypotension may occur (c) Idiopathic (sodium channelopathies)
(d) Infectious (HIV)
as a consequence of disorders affecting the barosensi­
(e) Hereditary
tive sympathoexcitatory neurons in the rostral ventro­ (i) Hereditary sensory and autonomic neuropathy
lateral medulla (for example, MSA); spinal preganglionic (ii) Fabry disease
sympathetic neurons (for example, MSA or PD); auto­ (iii) Sodium channelopathies
nomic ganglia (for example, AAG or PAF); unmyelin­ 4. Acute or subacute motor polyradiculopathy or
ated axons (SFN); noradrenaline availability (for neuropathy
example, dopamine β‑hydroxylase deficiency); or vas­ (a) Guillain–Barré syndrome
cular α1-adrenergic receptors (typically as a side effect (b) Porphyria
5. Acute autonomic and sensory neuropathy
of drugs).10
6. Ross syndrome (segmental anhidrosis, Adie pupils
and areflexia).
Anhidrosis
Sweating is an important thermoregulatory activity
mediated by the sympathetic nervous system through supraspinal reflex coordinated by the pontine mictur­
cholinergic activation of muscarinic M 3 receptors in ition centre, which activates the sacral pre­gangli­onic
the eccrine sweat glands. Depending on its distribu­ para­sympa­thetic output to the bladder detrusor while
tion and severity, anhidrosis might be asymptomatic ­simultaneously inhibiting the Onuf nucleus.13
or might manifest with compensatory hyperhidrosis Neurogenic bladder can manifest with detrusor over­
(in unaffected areas) or heat intolerance. Anhidrosis activity or underactivity.14 Detrusor overactivity pro­
in autonomic failure may reflect impairment of central duces urinary urgency with or without incontinence,
thermoregulatory pathways, spinal preganglionic urinary frequency, and nocturia. Reduced detrusor
sudomotor units, cholinergic sympathetic ganglion activity leads to incomplete bladder emptying, increased
neurons, peripheral unmyelinated axons, or M3 ­receptors post-void residual, low peak urinary flow rate and, even­
at the sudomotor unit.11 tually, urinary retention and overflow incontinence.
Impaired micturition can result from lesions affecting
Neurogenic bladder and sexual dysfunction the bladder afferents, sacral parasympathetic neurons or
Normal bladder function includes a urine storage their axons, or cholinergic muscarinic neurotransmis­
phase and a micturition phase, which are controlled sion. Neurogenic bladder is commonly associated with
at all levels of the neuraxis.12 Urine storage depends on erectile and ejaculatory dysfunction in men and poor
spinal reflexes mediated by the lumbosacral sympathetic vaginal lubrication in women. Erectile dysfunction
noradrenergic and sacral cholinergic motor neurons of reflects impaired sacral parasympathetic output and
the Onuf nucleus. Normal micturition depends on a nitric oxide release in the erectile tissue.

2  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Autonomic failure

Associated
manifestations None (‘pure’ autonomic failure) Parkinsonism, ataxia Peripheral neuropathy

Acute or subacute Chronic, progressive Subacute Distal Distal painful


sensory sensorimotor
Onset/ or any other
subtype type

Autoimmune ‘Pure’ Lewy body Multiple Sjögren Diabetes Vasculitis


autonomic autonomic disorders system syndrome Amyloidosis HIV
Main or ganglionopathy failure ■ PD atrophy
typical causes Paraneoplastic Idiopathic
Paraneoplastic ■ DLB Fabry disease
autonomic Hereditary
ganglionopathy Autoantibodies
(SSA/SSB, paraneoplastic)
Salivary gland biopsy

Autonomic studies Autonomic studies, neuroimaging (MRI), Autonomic studies, electromyography/nerve


Specific conduction studies, FBG, SPEP, UPEP,
evaluation* Autoantibodies polysomnogram, urodynamic studies,
(gnAChR, abdominal fat aspirate, ANCA, HIV,
paraneoplastic) neuropsychometrics α-galactosidase, punch skin biopsy

Figure 1 | Evaluating the main causes of autonomic failure. All patients with autonomic failure should undergo
determination of serum glucose (or in some cases haemoglobin A1c), thyroid stimulating hormone and vitamin B 12 levels.
Autonomic laboratory testing is helpful to detect and assess severity of autonomic failure. Patients with acute isolated
autonomic failure should also be tested for gnAChR and paraneoplastic antibodies, and SSA and SSB autoantibodies in
cases with prominent sensory symptoms. Patients with chronic progressive autonomic failure, including multiple system
atrophy, PD or DLB, should also undergo MRI (if possible), polysomnography, urodynamic studies and, in some cases,
neuropsychometric testing. Evaluation of patients with autonomic failure associated with peripheral neuropathy includes
electromyography and nerve conduction studies, SPEP and UPEP with immunofixation, abdominal fat aspirate
and/or sural nerve biopsy for detection of amyloidosis and, in some cases, determination of ANCA, HIV serology or
α‑galactosidase. Punch skin biopsy can also be helpful to detect small fibre neuropathy. *Include thermoregulatory sweat
test, sudomotor axon reflex tests, heart rate response to deep breathing and Valsalva manoeuvre, beat-to-beat blood
pressure profile during the Valsalva manoeuvre, and blood pressure and heart rate responses to head-up tilt.
Abbreviations: ANCA, antineutrophil cytoplasmic antibodies; DLB, dementia with Lewy bodies; FBG, fasting blood glucose;
gnAChR, ganglionic nicotinic acetylcholine receptor; PD, Parkinson disease; SPEP, serum protein electrophoresis; UPEP,
urine protein electrophoresis.

Gastrointestinal dysmotility after standing; and examination of the skin to identify


Control of gastrointestinal motility depends on intrin­ areas of localized or generalized absence of or excessive
sic reflexes mediated by the ENS, and their modulation sweating, and changes in skin temperature or colour in
by vagal and paravertebral sympathetic inputs. Upper the hands and feet.
gastro­intestinal motility is primarily controlled by the
vagal reflexes involving the nucleus of the solitary tract Laboratory tests
and dorsal motor nucleus of the vagus in the medulla;15 General laboratory tests that should be performed in
peristalsis in the lower gastrointestinal tract depends patients with autonomic failure include determination
primarily on local ENS reflexes;16 and paravertebral of serum glucose (or in some cases haemoglobin A1c),
­sympathetic reflexes inhibit gastrointestinal motility. thyroid-stimulating hormone and vitamin B12 levels.
Symptoms of delayed oesophageal transit include Serum and urine protein electrophoresis with immuno­
dysphagia and regurgitation. Delayed gastric emptying fixation, including light-chain quantitation, are indi­
produces early satiety, anorexia, nausea, bloating, belch­ cated to detect amyloid light-chain (AL) amyloidosis.17
ing, postprandial vomiting, and pain. Lower gastro­ SSA and SSB antibody testing for Sjögren syndrome may
intestinal dysmotility manifests with constipation and, be helpful in some cases. Determination of ganglionic
­occasionally, diarrhoea. nicotinic acetylcholine receptor (gnAChR) anti­bodies,18
as well as paraneoplastic antibodies (particularly anti-
Evaluation Hu, P/Q and N‑type voltage-gated calcium channel
Clinical and voltage-gated potassium complex anti­b odies), is
In addition to a careful history and general physical and indicated in all patients with subacute onset of symp­
neurological examination, clinical evaluation of patients toms.18,19 Determination of forearm venous catecho­
with suspected autonomic failure includes assessment of lamines, including noradrenaline, dopamine and
pupil size, symmetry and reactivity with both bright and adrenaline, in the supine position and after 5–10 min
dim light; measurement of blood pressure and heart rate of standing may be helpful in some cases; the results
after 2 min in the supine position and at 1 min and 2 min require careful interpretation, however, as they can be

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION  |  3


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

affected by concomitant use of drugs or impaired pre­ These conditions include MSA, 1,39–41 and Lewy body
synaptic noradrenaline reuptake.20 Low or undetectable ­disorders such as PD, DLB or PAF.42,43
noradrenaline and adrenaline levels associated with
increased dopamine levels indicate a deficit of dopamine Multiple system atrophy
β‑hydroxylase as the cause of orthostatic hypotension.21 MSA was initially defined as a sporadic neurodegenera­
tive disease characterized by any combination of auto­
Autonomic function tests nomic failure, parkinsonism, cerebellar ataxia and/or
Autonomic laboratory evaluation is indicated to deter­ pyramidal signs.44 The prevalence of MSA has been esti­
mine the extent and severity of autonomic failure; to mated at 4–8 cases per 100,000 people, and its incidence
detect autonomic failure in patients with parkinsonism varies from 0.6 cases per 100,000 person-years in the
or ataxia; to assess small fibre function in patients with general population to 3 cases per 100,000 person-years
peripheral neuropathy or ganglionopathy; to evalu­ in people above 50 years of age.1,40
ate patients with symptoms of orthostatic intolerance; The neuropathological hallmark of MSA is the accumu­
and to obtain objective evidence of disease progression lation of α‑synuclein-immunoreactive glial cytoplasmic
or response to medications.22,23 The most commonly inclusions in oligodendrocytes, and neuronal loss in the
used tests assess sudomotor, cardiovagal and adren­ striatum, substantia nigra pars compacta, pontine nuclei,
ergic vasomotor functions. Sudomotor function tests inferior olivary nuclei, cerebellum, and premotor auto­
include the thermoregulatory sweat test (TST), which nomic nuclei.45 According to current criteria, probable
assesses the integrity of central and peripheral sudo­ MSA is defined as a sporadic, progressive, adult-onset
motor pathways,22 and sudomotor axon reflex tests such (over 30 years of age) disease characterized by autonomic
as the quanti­tative sudomotor axon reflex test (QSART)23 failure, with urinary incontinence and erectile dysfunc­
and the quanti­tative direct and indirect test of sudo­ tion (in males) or orthostatic hypotension (blood pres­
motor function (QDIRT),24 which assess the peripheral sure decrease ≥30 mmHg systolic or ≥15 mmHg diastolic
sympa­thetic cholinergic innervation of the sweat glands. within 3 min of standing), as well as parkinsonism poorly
Tests that assess vagal control of the sinus node (cardio­ responsive to levodopa (MSA‑P), or cerebellar syndrome
vagal function) include the heart rate response to deep (MSA‑C).39 A large European study including 437 patients
breathing,25 and the heart rate response to the Valsalva with probable (72%) or possible (28%) MSA showed that
manoeuvre, or Valsalva ratio.26 Tests that indirectly assess symptomatic autonomic failure was present in almost all
sympathetic vasomotor function include the beat-to-beat cases; the most frequent manifestations were urinary dys­
blood pressure profile during the Valsalva manoeuvre27 function (83%) and orthostatic hypotension (75%).41 In a
and the blood pressure response to head-up tilt.28 The study on 29 autopsy-confirmed cases, a pattern consist­
physiological basis, methods, normal and abnormal ing of severe and progressive adrenergic and sudomotor
responses, and pitfalls of these and other autonomic failure was highly predictive of MSA.46
­function tests have been the subject of several reviews.29–31 Although MSA is still generally regarded as a sporadic
disease, several studies have identified familial cases.1
Other tests A recent study identified mutations of the coenzyme Q2
Electromyography is indicated for patients with periph­ (COQ2) gene in Japanese families with MSA, and COQ2
eral neuropathy or ganglionopathy, and MRI may be variants were also associated with an increased risk of
helpful in evaluation of central autonomic disorders. sporadic MSA.47
Gastrointestinal motility and urodynamic studies might Median survival from symptom onset in MSA is
be indicated in some cases. Polysomnography to detect 8–10 years, but the spectrum ranges from 4–15 years.
sleep-related respiratory dysfunction is indicated in all Natural history studies indicate that presence of the
patients with suspected MSA. parkinsonian variant, early onset of autonomic failure,
incomplete bladder emptying, and shorter duration
Tissue biopsy of symptoms at baseline are all factors that predict
Skin biopsy with assessment of intraepidermal nerve shorter survival.48–50
fibre density (IENFD) can be useful in the evaluation In MSA, orthostatic hypotension occurs at earlier
of SFN;32,33 immunocytochemical markers allow quanti­ stages and is more severe than in PD. 51 Orthostatic
fication of the density of innervation of sweat gland34 hypotension in MSA reflects involvement of pre­gangli­
and pilomotor 35 nerves. Abdominal fat aspirate or sural onic sympathetic neurons52 and sympathoexcitatory
nerve biopsy are indicated to evaluate for amyloidosis.36,37 neurons of the rostral ventrolateral medulla.53 Urogenital
Immunostaining and laser microdissection, along with manifestations may herald the onset of MSA.51 Bladder
mass spectrometry-based proteomic analysis of amyloid dysfunction in MSA is characterized by urinary urgency,
deposition, allow identification of the specific subtype of followed by incontinence and incomplete bladder empty­
amyloid protein.38 ing. These manifestations result from a combination of
detrusor hyperreflexia and urethral sphincter weakness
Neurodegenerative synucleinopathies followed by detrusor contraction failure,54 and they
Autonomic failure is an important manifestation of reflect involvement of the pontine micturition centre,55
neuro­degenerative disorders characterized by the pres­ sacral preganglionic neurons 56 and Onuf nucleus. 57
ence of intracellular inclusions containing α‑synuclein. Erectile dysfunction is almost always present in male

4  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

patients with MSA.58 Upper gastrointestinal symptoms, syndrome reflecting involvement of the autonomic gan­
constipation and faecal incontinence occur frequently; glion cells and their postganglionic axons. This disorder
anal sphincter electromyography shows large motor can have a variety of different pathobiological substrates.
unit potentials indicating denervation due to loss of Patients with PAF have been shown to have accumula­
Onuf nucleus motor neurons.59 Anhidrosis occurs in the tion of α‑synuclein-containing Lewy bodies and neurites
majority of patients with MSA, and the percentage of in sympathetic ganglia peripheral tissues, including skin
the body affected by anhidrosis may help to differentiate sympathetic nerves.74,75 However, some cases of PAF are
MSA from PD.60,61 associated with low-titre gnAChR antibodies, suggesting
An important manifestation of disrupted brainstem that this condition can reflect a chronic form of AAG in
homeostatic mechanisms in MSA is sleep-related dis­ some instances.77
ordered breathing, including sleep apnoea and laryn­
geal stridor, which occur in up to 70% of patients with Parkinson disease
MSA.62–64 Whereas the development of central hypo­ Autonomic dysfunction—particularly gastrointestinal
ventilation and laryngeal stridor is closely related to the dysmotility—is a prominent nonmotor manifestation
severity of autonomic failure, respiratory manifestations of PD.43 Gastrointestinal dysmotility in PD can affect
can occur early in the course of MSA.65 Laryngeal dys­ any level of the gastrointestinal tract.78 Gastrointestinal
tonia with inspiratory adduction of the vocal cords might symptoms can occur at an early stage of the disease;79 for
underlie laryngeal stridor in at least some individuals with example, constipation may precede the development of
MSA.66 Impaired automatic ventilation reflects involve­ motor symptoms by several years.80,81 Excessive drooling
ment of the brainstem respiratory network, including in PD must reflect oropharyngeal dysphagia, as salivary
the pre-Bötzinger complex 67 and the medullary raphe,68 secretion is reduced even at an early stage of disease;82,83
which may predispose patients to sudden death.69 this finding is consistent with the presence of α‑synuclein
pathology in the salivary gland.84 Oesophageal dysmotility
Lewy body disorders and delayed gastric emptying are frequent manifestations
Accumulation of Lewy neurites and Lewy bodies in in PD,85,86 and are likely to reflect early involvement of
the ENS, autonomic ganglia, peripheral autonomic the dorsal motor nucleus of the vagus.70 The prevalence
nerve terminals, and intermediolateral cell column of constipation ranges from 20–89%,79 and is primarily
can be detected at autopsy of cases of incidental Lewy attributable to slow colonic transit, reflecting the early
body disease (ILBD), PAF, early-stage PD, and DLB.70–73 involvement of the ENS.87 However, defecatory dysfunc­
α‑Synuclein inclusions can be also detected in cutan­eous tion due to paradoxical contraction of the ­puborectalis
nerves of patients with PAF 74 or PD.75 These findings muscle is also an important contributory factor.85,88
indicate that Lewy body disorders constitute a contin­ Orthostatic hypotension is estimated to occur in
uum of progressive involvement of areas of the periph­ 16–58% of patients with PD.89 In general, orthostatic
eral autonomic nervous system, and subsequently the hypotension in PD is asymptomatic and tends to occur
CNS, by α‑synuclein neuropathology, which may range at later stages of disease than in MSA, but in a subgroup
clinically from asymptomatic cases (ILBD) to pure auto­ of patients it may be early and prominent, manifesting
nomic failure (PAF), and might eventually manifest with even before the initiation of dopaminergic therapy.89
­parkinsonism (PD), dementia (DLB) or both. Urinary symptoms occur in 38–71% of patients with
PD.77,90,91 The most prominent symptom is nocturia, fol­
Pure autonomic failure lowed by urgency and frequency, and the most common
PAF was first described by Bradbury and Eggleston in urodynamic finding is detrusor overactivity.92 Nocturia is
1925, and was initially termed idiopathic orthostatic the only urinary symptom that improves after deep brain
hypotension. PAF is a rare, sporadic, adult-onset dis­ stimulation of the subthalamic nucleus.
order characterized by symptomatic orthostatic hypo­
tension and variable gastrointestinal, bladder and sexual Differential diagnosis
dysfunction, in the absence of somatic motor deficits. The differential diagnosis between MSA‑P and PD with
Onset is typically between 50 and 70 years of age.42 autonomic failure can be difficult. A prospective study
The symptoms of PAF are insidious in onset, and are showed that autonomic indices, particularly the percent­
less progressive and disabling than those in other neuro­ age of the body affected by anhidrosis, were significantly
degenerative autonomic disorders. Some patients may and persistently more abnormal in MSA than in PD with
have bladder symptoms, erectile or ejaculatory dysfunc­ autonomic failure.61 Urodynamic findings such as large
tion, or impaired sweating, before developing ortho­ post-micturitional residual volume (>100 ml), detrusor
static hypotension. Constipation is common, but other external sphincter dyssynergia, and open bladder neck
symptoms of gastrointestinal dysmotility are rare.76 The at the start of filling are highly suggestive of MSA. 54
diagnosis of PAF is always tentative; after a few years, Anal sphincter denervation due to loss of Onuf nucleus
many patients with presumed PAF may develop cerebel­ motor neurons may help to distinguish MSA from PD
lar, extrapyramidal or cognitive deficits indicating MSA, within the first few years of onset of motor symptoms,
PD or DLB. Thus, the diagnosis of PAF requires at least a but this finding is not specific for MSA, and can occur
5 year history of isolated autonomic dysfunction without in progressive supranuclear palsy or in PD patients with
other neurological manifestations. PAF is a clinical chronic constipation.93

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION  |  5


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Structural brain imaging, including diffusion- autonomic failure, including gastrointestinal enteropathy.
weighted and diffusion tensor MRI, and functional Early recognition of these disorders is important because
imaging, including PET and single-photon emis­ they are disabling and might be the initial manifestation
sion CT (SPECT), might aid the differential diagnosis of an underlying neoplasm. In addition, patients with
between MSA and PD.1 The presence of atrophy in the autoimmune autonomic disorders may show substantial
putamen, middle cerebellar peduncle and pons on MRI improvement with i­ mmunomodulatory therapy.4,109
serve as supporting features for the diagnosis of pos­
sible MSA.39 Signal changes on T2-weighted images, Autoimmune autonomic ganglionopathy
including the slit-like void signal (hypointensity of AAG (formerly known as acute pandysautonomia or
the posterior putamen surrounded by hyperintense idiopathic subacute autonomic neuropathy 18,110) is
lateral putaminal rim) and the hot cross bun sign (due characterized by severe autonomic failure that develops
to myelin loss in the basis pontis) are more charac­ over the course of days or weeks in a previously healthy
teristic of MSA but are not specific for this condition. person. The onset of symptoms may follow a viral infec­
Diffusion-weighted imaging shows elevated apparent tion, minor surgical procedure, or vaccination. The
diffusion coefficient in the putamen, pons and middle most common manifestation is generalized sympathetic,
cerebellar peduncle in MSA.94 In MSA‑P, but not in PD, parasympathetic and ENS failure, but the spectrum is
brain perfusion SPECT shows striatal hypoperfusion,95 broad; symptoms might be confined to pure choliner­
and 18F-fluorodeoxyglucose PET shows striatal hypo­ gic neuropathy manifested by sicca syndrome, isolated
metabolism.96 By contrast, dopamine transporter SPECT adrenergic neuropathy, or isolated gastrointestinal dys­
using 123I-FP-CIT demonstrates reduced striatal binding motility. Patients may recover spontaneously, but only
in both disorders.97 one-third experience substantial functional ­improvement
Myocardial scintigraphy using 123I-metaiodo­benzyl­ without treatment.
guanidine (MIBG) 98 or 6‑18F-fluorodopamine PET 99 Approximately 50% of cases of subacute AAG are
assesses postganglionic sympathetic innervation of the associ­ated with gnAChR antibodies18 that block trans­
heart; these modalities are potential tools to differenti­ mission at the autonomic ganglia. Antibody titres
ate between MSA and Lewy body disorders. However, correlate with the severity of autonomic failure.19,111
although initial studies suggested that loss of cardiac gnAChR antibodies can also be associated with chronic
sympathetic innervation in PD or PAF could help in or restricted forms of autonomic dysfunction,77 and with
distinguishing Lewy body disorders from MSA,99,100 neurological or paraneoplastic disorders unrelated to
decreased cardiac MIBG uptake has also been found in the autonomic nervous system.19 In one series, 30% of
up to 30% of patients with MSA,101 possibly reflecting gnAChR-seropositive patients had other paraneoplastic
coexistent Lewy body pathology. antibodies in the setting of occult cancer, most com­
monly adenocarcinoma.19 In some cases, AAG coexists
Dementia with Lewy bodies with other autoimmune disorders, such as myasthenia
Severe autonomic dysfunction, as well as repeated falls gravis, in patients with occult cancer.112,113
and syncope, are features supporting the diagnosis of Several observational studies report that patients
DLB. 102 Orthostatic hypotension, urinary frequency with gnAChR-seropositive AAG benefit from intra­
and urge incontinence can be disabling symptoms in venous immunoglobulin, plasma exchange, prednisone,
DLB,103,104 and symptomatic orthostatic hypotension mycophenolate, azathioprine and rituximab, either
may occur in 30–50% of cases. On the basis of auto­ individually or in combination. 19,114–116 A substantial
nomic laboratory evaluation, the severity of autonomic percentage of patients with AAG are seronegative for
failure in DLB is intermediate between that seen in MSA gnAChR and other autoantibodies;117 some of these
and PD.105 patients may also show symptomatic improvement with
­immunomodulatory therapy.116
Other neurodegenerative disorders
Other central neurodegenerative disorders can manifest Paraneoplastic autonomic neuropathy
with autonomic failure that reflects peripheral autonomic A type of subacute autonomic neuropathy that is clinically
involvement. These conditions, which may clinically indistinguishable from idiopathic AAG might occur as
mimic MSA or Lewy body disorders, include fragile X a manifestation of an occult neoplasm, most commonly
tremor–ataxia syndrome, in which bladder symptoms small-cell lung carcinoma associated with anti-Hu
might be prominent;106 adult-onset autosomal dominant antibodies or, less frequently, thymoma or other neo­
leukodystrophy, which is associated with pure sympa­ plasms.118,119 Like AAG, paraneoplastic autonomic neuro­
thetic failure;107 and a novel prion disorder character­ pathy may manifest with generalized autonomic failure,
ized by diarrhoea and length-dependent, predominantly or with isolated enteric ganglionopathy leading to the
sensory and autonomic axonal neuropathy, followed by intestinal pseudo-obstruction syndrome.120,121
cognitive decline.108
Acute autonomic and sensory neuropathy
Autoimmune autonomic disorders Acute autonomic and sensory neuropathy is a rare dis­
Autoimmune autonomic disorders are characterized order that differs from AAG in that profound autonomic
by acute or subacute onset of generalized or restricted failure is associated with various degrees of sensory

6  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

impairment, including loss of pain and temperature Diabetic autonomic neuropathy can affect cardio­
sensation, followed by sensory ataxia due to impair­ vascular, gastrointestinal, urogenital and sudomotor
ment of proprioception.109,122,123 The mean age of onset functions to various degrees. In most of the typical cases,
is in the late second decade, but can vary from child­ a combination of cardiovascular tests and sudomotor
hood to the elderly. A triggering event, such as respira­ tests allows its detection in otherwise asympto­matic
tory or gastrointestinal tract infections, is reported in patients. An impaired heart rate response to deep
two-thirds of patients. Gastrointestinal dysmotility is breathing­—an indicator of cardiovagal failure—has the
the most prominent autonomic symptom; the sensory greatest specificity (~80%).131,132 Characteristic mani­
loss affects the proximal regions of the limbs, face, scalp festations of diabetic cardiovascular autonomic neuro­
and trunk, and tends to be asymmetrical and segmen­ pathy include resting tachycardia, exercise intolerance,
tal. Pain in the involved region is a common and serious orthostatic hypotension, intraoperative cardiovascular
symptom. Other features of this disorder are reduction of instability, and silent myocardial infarction–silent ischae­
sensory nerve action potentials and increased T2 signal mia syndrome. Cardiovascular autonomic neuropathy
in the posterior columns of the cervical spinal cord. 123 has been associated with increased morbidity and mor­
Autopsy studies have shown severe neuronal cell loss in tality,133 so screening for this condition should be per­
the thoracic sympathetic and dorsal root ganglia, and formed at the time of diagnosis of type 2 diabetes and
in Auerbach’s plexus.123 5 years after the diagnosis of type 1 diabetes, particularly
in high-risk patients.129
Sjögren syndrome Upper gastrointestinal dysmotility, which is present
Primary Sjögren syndrome manifests with a wide variety in 40–50% of longstanding cases of diabetes, may
of peripheral neuropathies or ganglionopathies.124 Dorsal affect postprandial blood glucose levels and glycaemic
root ganglionopathy may manifest with neuropathic pain control. Constipation is the most frequent gastrointesti­
or ataxic neuropathy; autonomic neuropathy might be nal symptom in diabetes, and was reported by 60% of
prominent in some cases, but is the least frequent form patients in one study.134 Profuse, watery and predomi­
of neuropathy overall.124 nantly nocturnal diarrhoea135 and faecal incontinence136
Autoantibodies against muscarinic M3 receptors have may be disabling in some patients. Bladder symptoms
been identified in a subset of patients with Sjögren syn­ occur in up to 50% of patients; 43–87% of type 1 and 25%
drome.125 These antibodies are particularly prevalent in of type 2 diabetic patients have abnormal urodynamic
juvenile cases126 and have cholinergic blocking actions findings.137,138 Erectile dysfunction has a reported preva­
in vitro.127 Some patients with Sjögren syndrome and lence of 35–90% in patients with diabetes.139,140 Length-
chronic progressive autonomic failure have gnAChR dependent loss of thermoregulatory sweating is common,
antibodies, which may predict a positive response to and is occasionally associated with compensatory hyper­
immunomodulatory therapy.128 hidrosis.22 Gustatory sweating can result from aberrant
innervation of sweat glands by parasympathetic fibres.141
Peripheral neuropathies Some patients with diabetes develop an acute painful
Peripheral neuropathies associated with autonomic neuropathy associated with autonomic failure within a
failure present with various phenotypes, including typical short time of commencing tight glucose control.142 The
distal symmetric sensorimotor neuropathy; dorsal root symptoms may be associated with abnormal autonomic
ganglionopathy manifesting with neuropathic pain or test results and reduced IENFD on punch skin biopsy,
sensory ataxia; demyelinating, predominantly motor and can all improve over time.142
polyradiculopathy; or distal painful neuropathy. The
spectrum of severity of autonomic failure is highly vari­ Amyloid neuropathy
able in these disorders, ranging from being a major cause Amyloidoses manifesting with autonomic neuro­
of disability, as can occur in neuropathies associated pathy include the AL type (which may be associated
with diabetes or amyloidosis, to restricted distal vaso­ with myeloma or macroglobulinaemia), and familial
motor and sudomotor impairment in the lower limbs, amyloid polyneuropathy (FAP), which is most com­
as observed in painful SFN. monly linked to mutations in the transthyretin (TTR)
gene. The types of amyloid neuropathy associated with
Diabetic autonomic neuropathy autonomic failure are highly heterogeneous. 143,144 In a
Diabetic neuropathies are heterogeneous: they include retrospective study on 65 cases of AL amyloidosis or
typical diabetic sensorimotor polyneuropathy, painful FAP, generalized autonomic failure was associated with
diabetic neuropathy, and diabetic autonomic neuro­ painful (62%) or nonpainful (17%) sensorimotor poly­
pathy.129 Estimates of the prevalence of diabetic auto­ neuropathy or distal SFN (5%); autonomic failure also
nomic neuropathy vary depending of the source of occurred without neuropathy (11%), but polyneuro­
information (community, clinic or tertiary referral centre) pathy without generalized autonomic failure was infre­
and the type of tests performed to assess autonomic func­ quent (6%).143 In AL amyloidosis, peripheral neuropathy
tion.129 Using strict criteria based on autonomic testing may be the presenting feature or an incidental finding
abnormalities, the prevalence in one study was 16.8% for of the disease, and can occur in up to 20% of cases. 145
patients with type 1 and 34.3% for patients with type 2 The median survival of patients with AL amyloidosis
diabetes mellitus.130 who have peripheral neuropathy has improved with the

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION  |  7


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | General management of autonomic failure


Symptom Approach Comments
Orthostatic Patient education: recognize atypical symptoms of The most important aspect of management
hypotension orthostatic hypotension; get up slowly from bed; urinate
in the sitting position; avoid hot, humid environments;
exercise regularly; avoid straining; keep a blood
pressure diary; discontinue aggravating drugs
Sleep with the head elevated 4 in (reverse Avoids supine hypertension and nocturnal polyuria
Trendelenburg position)
Postural countermanouevres (for example, leg crossing, Increases orthostatic tolerance
squatting)
Support garments (waist-high support stockings, Reduces venous pooling
abdominal binder)
Volume expansion (daily intake of 8–10 g sodium, Initial step before pharmacological management
fluid intake of at least 2.0–2.5 l)
Rapid ingestion of 500 ml tap water Rapidly increases blood pressure independently
of volume expansion
Dietary changes; avoid alcohol; eat small, frequent Prevents splanchnic vasodilation
meals (carbohydrate is best ingested with the last
meal of the day)
Florinef (0.1–0.4 mg daily) Adverse effects include supine hypertension, ankle
oedema, hypokalaemia, hypomagnesaemia, headache,
and reduced effects of warfarin
Midodrine (10–40 mg daily) Adverse effects include scalp tingling or itching,
goose bumps, supine hypertension, and urinary urgency
or retention
Pyridostigmine (60–240 mg daily) Adverse effects include nausea, abdominal cramps,
diarrhoea, increased salivation, urinary urgency and
bradycardia
Droxidopa (100–600 mg TID) Main potential adverse effect is supine hypertension
Erythropoietin (25–75 U/kg SQ) Iron supplementation is usually required; supine
hypertension may occur
Desmopressin nasal spray (5–40 μg) Risk of water intoxication and hyponatraemia
Octreotide (25–200 μg SQ) Adverse effects include nausea and abdominal cramps,
hypertension, risk of gallstones and postprandial
hyperglycaemia
Neurogenic Anticholinergics (for example, oxybutynin, tolterodine); Adverse effects include constipation, dry mouth, risk
bladder clean intermittent self-catheterization of glaucoma, and worsening of cognitive function
Drooling Anticholinergics; botulinum toxin Anticholinergics may worsen confusion; botulinum toxin
may worsen dysphagia
Gastroparesis Metoclopramide (5–20 mg QID); domperidone Gastroparesis may impair absorption of levodopa;
(10–30 mg QID); erythromycin (50–250 mg QID); metoclopramide is contraindicated in parkinsonian
pyridostigmine (30–60 mg TID); bethanechol (25 mg disorders; domperidone may cause QT prolongation,
QID); nutritional support (low-fat, low-fibre oral and is not available in the USA
supplementation; infusion of formula via a jejunal
feeding tube)
Constipation Fibre supplementation (15 g per day); bulk agents Donepezil and other cholinesterase inhibitors may
(for example, psyllium [1 tsp up to three times daily], cause diarrhoea
methylcellulose); osmotic laxatives (for example, milk
of magnesia [two tablets up to four times daily],
polyethylene glycol [17 g in 25 ml once or twice daily],
lactulose [15–30 ml TID]); docusate (stool softener,
100 mg BID); lubiprostone (24 μg BID); pyridostigmine
(up to 180–540 mg); bisacodyl (10 mg up to three times
a week); antibiotics to prevent bacterial overgrowth;
surgery for intractable colonic inertia
Erectile Oral phosphodiesterase type 5 inhibitors (for example, Adverse effects include headache, flushing, nasal
dysfunction sildenafil 25–100 mg) congestion, blue vision, and non-arteritic ischaemic
optic neuropathy; these drugs may worsen orthostatic
hypotension
Intracavernous drugs (for example, alprostadil, Adverse effects include penile pain, oedema and
papaverine) haematoma, palpable nodules or plaques, and priapism
Abbreviations: BID, twice a day; QID, four times a day; SQ, subcutaneous; TID, three times a day.

8  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

combination of melphalan, prednisone and autologous The principles of management include discontinua­
stem cell transplantation.146 tion of potentially causative drugs; immunomodula­
The clinical and pathological features of FAP have tory therapy for autoimmune disorders; management
been reviewed in detail elsewhere.147,148 Over 100 muta­ of diabetes, amyloidosis or other potentially treatable
tions in the TTR gene have been identified, the most cause of autonomic neuropathy; patient education; non-­
common (~50% of cases of FAP worldwide) of which pharmacological approaches; and pathophysiologically
causes a Val30Met substitution. Different TTR variants based drug therapy. Patient education is a fundamen­
contribute to variable age at diagnosis, degree of cardiac tal aspect of management. For example, patients with
involvement, and survival.149 The first-line specific treat­ orthostatic hypotension should be instructed to recog­
ment of choice for Val30Met TTR-FAP is liver transplan­ nize atypical symptoms and avoid precipitating factors.
tation.150 In addition, tafamidis, a stabilizer of tetrameric Appropriate fluid and sodium intake, dietary adjust­
transthyretin, has shown short-term effectiveness in ments and regular exercise are beneficial in most cases
slowing the progression of peripheral neuropathy at very of autonomic failure.
early stages of Val30Met TTR-FAP.151
Conclusions
Painful small fibre neuropathies The wide spectrum of clinical manifestations and dis­
Selective involvement of postganglionic as well as noci­ orders associated with autonomic failure requires a
cep­tive fibres in painful SFN typically manifests with systematic clinical and laboratory approach to establish
distal anhidrosis (or sometimes hyperhidrosis) and the diagnosis, particularly in cases where autonomic
vasomotor dysfunction (generally erythema, but some­ failure is disabling or life-threatening. A careful history
times vasoconstriction and coldness) in the lower limbs. and examination is the mainstay of diagnosis, and lab­
In some cases, SFN can result in more-generalized oratory and other ancillary tests must be prioritized to
autonomic failure that may be symptomatic or detect­ search for potentially treatable causes. Patient education,
able with autonomic testing. Important examples of avoidance of precipitating factors, non-pharmacological
painful SFN are those associated with HIV infections152 approaches, and pathophysiologically based drug therapy
and Fabry disease,153 in addition to the types related to form the basis of current treatment of these disorders.
dia­betes and amyloidosis, as highlighted above. Distal Further elucidation of the pathobiological mechanisms
sympathetic sudomotor and vasomotor failure in con­ that lead to immune, metabolic or degenerative damage
junction with neuropathic pain also occur in sodium or dysfunction of central and peripheral autonomic
channelopathies associated with gain-of-function neurons will provide more-specific therapeutic targets.
SCNA9 mutations, both in familial syndromes154 and in
a substantial proportion of idiopathic SFN cases.155 The
Review criteria
SCNA9 gene encodes the Nav1.7 voltage-gated sodium
channel, which is expressed in nociceptive dorsal root A PubMed search from 1st January 1975 to 15th December
ganglion and sympathetic neurons. These gain-of-­ 2013 was performed using the following terms (individually
and in various combinations): “autonomic failure”,
function mutations cause increased excitability of the
“autonomic neuropathy”, “orthostatic hypotension”,
nociceptor and hypoexcitability of sympathetic ganglion
“multiple system atrophy”, “Parkinson disease”, “Lewy
neurons,156,157 which might explain the coexistence of body disease”, “dementia with Lewy bodies”, “autoimmune
neuropathic pain and postganglionic sympathetic failure. autonomic ganglionopathy”, “paraneoplastic”, “diabetic
neuropathy”, “amyloid neuropathy”, “Sjögren syndrome”,
Management issues “leukoencephalopathy”, “fragile X” and “prion”. The
A detailed discussion of the management of autonomic search was limited to full articles published in English.
failure is beyond the scope of this Review, but the general Other articles were identified from bibliographies of the
retrieved articles.
approach is summarized in this section and in Table 1.

1. Stefanova, N., Bucke, P., Duerr, S. & mediated syncope and the postural tachycardia 11. Cheshire, W. P. & Freeman, R. Disorders
Wenning, G. K. Multiple system atrophy: an syndrome. Auton. Neurosci. 161, 46–48 (2011). of sweating. Semin. Neurol. 23, 399–406
update. Lancet Neurol. 8, 1172–1178 (2009). 7. Mathias, C. J. et al. Differential blood pressure (2003).
2. Cersosimo, M. G. & Benarroch, E. E. Autonomic and hormonal effects after glucose and xylose 12. Fowler, C. J., Griffiths, D. & de Groat, W. C. The
involvement in Parkinson’s disease: pathology, ingestion in chronic autonomic failure. Clin. Sci. neural control of micturition. Nat. Rev. Neurosci.
pathophysiology, clinical features and possible (Lond.) 77, 85–92 (1989). 9, 453–466 (2008).
peripheral biomarkers. J. Neurol. Sci. 313, 8. Mathias, C. J., Mallipeddi, R. & Bleasdale- 13. Holstege, G. Micturition and the soul. J. Comp.
57–63 (2012). Barr, K. Symptoms associated with orthostatic Neurol. 493, 15–20 (2005).
3. Vernino, S. Autoimmune and paraneoplastic hypotension in pure autonomic failure and 14. Panicker, J. N. & Fowler, C. J. The bare
channelopathies. Neurotherapeutics 4, 305–314 multiple system atrophy. J. Neurol. 246, essentials: uro-neurology. Pract. Neurol. 10,
(2007). 893–898 (1999). 178–185 (2010).
4. Muppidi, S. & Vernino, S. Autoimmune 9. Freeman, R. Clinical practice. Neurogenic 15. Travagli, R. A., Hermann, G. E., Browning, K. N.
autonomic failure. Handb. Clin. Neurol. 117, orthostatic hypotension. N. Engl. J. Med. 358, & Rogers, R. C. Brainstem circuits regulating
321–327 (2013). 615–624 (2008). gastric function. Annu. Rev. Physiol. 68, 279–305
5. Freeman, R. Autonomic peripheral neuropathy. 10. Benarroch, E. E. The arterial baroreflex: (2006).
Lancet 365, 1259–1270 (2005). functional organization and involvement in 16. Furness, J. B. The enteric nervous system and
6. Freeman, R. et al. Consensus statement on the neurologic disease. Neurology 71, 1733–1738 neurogastroenterology. Nat. Rev. Gastroenterol.
definition of orthostatic hypotension, neurally (2008). Hepatol. 9, 286–294 (2012).

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION  |  9


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

17. Kyle, R. A. & Bayrd, E. D. Amyloidosis: review of 38. Klein, C. J. et al. Mass spectrometric-based 57. Chalmers, D. & Swash, M. Selective
236 cases. Medicine (Baltimore) 54, 271–299 proteomic analysis of amyloid neuropathy type in vulnerability of urinary Onuf motoneurons in
(1975). nerve tissue. Arch. Neurol. 68, 195–199 (2011). Shy–Drager syndrome. J. Neurol. 234, 259–260
18. Vernino, S. et al. Autoantibodies to ganglionic 39. Gilman, S. et al. Second consensus statement (1987).
acetylcholine receptors in autoimmune on the diagnosis of multiple system atrophy. 58. Kirchhof, K., Apostolidis, A. N., Mathias, C. J. &
autonomic neuropathies. N. Engl. J. Med. 343, Neurology 71, 670–676 (2008). Fowler, C. J. Erectile and urinary dysfunction may
847–855 (2000). 40. Wenning, G. K., Ben Shlomo, Y., Magalhaes, M., be the presenting features in patients with
19. McKeon, A., Lennon, V. A., Lachance, D. H., Daniel, S. E. & Quinn, N. P. Clinical features and multiple system atrophy: a retrospective study.
Fealey, R. D. & Pittock, S. J. Ganglionic natural history of multiple system atrophy. An Int. J. Impot. Res. 15, 293–298 (2003).
acetylcholine receptor autoantibody: oncological, analysis of 100 cases. Brain 117, 835–845 59. Sakakibara, R. et al. Colonic transit time,
neurological, and serological accompaniments. (1994). sphincter EMG, and rectoanal videomanometry
Arch. Neurol. 66, 735–741 (2009). 41. Kollensperger, M. et al. Presentation, diagnosis, in multiple system atrophy. Mov. Disord. 19,
20. Goldstein, D. S. Catecholamines 101. Clin. and management of multiple system atrophy in 924–929 (2004).
Auton. Res. 20, 331–352 (2010). Europe: final analysis of the European multiple 60. Sandroni, P., Ahlskog, J. E., Fealey, R. D.
21. Robertson, D. et al. Isolated failure of autonomic system atrophy registry. Mov. Disord. 25, & Low, P. A. Autonomic involvement in
noradrenergic neurotransmission. Evidence for 2604–2612 (2010). extrapyramidal and cerebellar disorders.
impaired β‑hydroxylation of dopamine. N. Engl. 42. Kaufmann, H. & Biaggioni, I. Autonomic failure in Clin. Auton. Res. 1, 147–155 (1991).
J. Med. 314, 1494–1497 (1986). neurodegenerative disorders. Semin. Neurol. 23, 61. Lipp, A. et al. Prospective differentiation of
22. Fealey, R. D., Low, P. A. & Thomas, J. E. 351–363 (2003). multiple system atrophy from Parkinson disease,
Thermoregulatory sweating abnormalities in 43. Chaudhuri, K. R. & Schapira, A. H. Non-motor with and without autonomic failure. Arch. Neurol.
diabetes mellitus. Mayo Clin. Proc. 64, 617–628 symptoms of Parkinson’s disease: dopaminergic 66, 742–750 (2009).
(1989). pathophysiology and treatment. Lancet Neurol. 62. Iranzo, A. Sleep and breathing in multiple system
23. Low, P. A. Autonomic nervous system function. 8, 464–474 (2009). atrophy. Curr. Treat. Options Neurol. 9, 347–353
J. Clin. Neurophysiol. 10, 14–27 (1993). 44. Graham, J. G. & Oppenheimer, D. R. Orthostatic (2007).
24. Gibbons, C. H., Illigens, B. M., Centi, J. & hypotension and nicotine sensitivity in a case of 63. Ghorayeb, I., Bioulac, B. & Tison, F. Sleep
Freeman, R. QDIRT: quantitative direct and multiple system atrophy. J. Neurol. Neurosurg. disorders in multiple system atrophy. J. Neural
indirect test of sudomotor function. Neurology Psychiatry 32, 28–34 (1969). Transm. 112, 1669–1675 (2005).
70, 2299–2304 (2008). 45. Papp, M. I., Kahn, J. E. & Lantos, P. L. Glial 64. Silber, M. H. & Levine, S. Stridor and death
25. Bennett, T. et al. Assessment of vagal control of cytoplasmic inclusions in the CNS of patients in multiple system atrophy. Mov. Disord. 15,
the heart in diabetes. Measures of R–R interval with multiple system atrophy (striatonigral 699–704 (2000).
variation under different conditions. Br. Heart J. degeneration, olivopontocerebellar atrophy 65. Glass, G. A., Josephs, K. A. & Ahlskog, J. E.
39, 25–28 (1977). and Shy–Drager syndrome). J. Neurol. Sci. 94, Respiratory insufficiency as the primary
26. Booth, R. W. & Ryan, J. M. The clinical use of the 79–100 (1989). presenting symptom of multiple-system atrophy.
Valsalva maneuver. Heart Bull. 10, 111–113 46. Iodice, V. et al. Autopsy confirmed multiple Arch. Neurol. 63, 978–981 (2006).
(1961). system atrophy cases: Mayo experience and 66. Vetrugno, R. et al. Sleep-related stridor due to
27. Sandroni, P., Benarroch, E. E. & Low, P. A. role of autonomic function tests. J. Neurol. dystonic vocal cord motion and neurogenic
Pharmacological dissection of components of Neurosurg. Psychiatry 83, 453–459 (2012). tachypnea/tachycardia in multiple system
the Valsalva maneuver in adrenergic failure. 47. Multiple-System Atrophy Research Collaboration. atrophy. Mov. Disord. 22, 673–678 (2007).
J. Appl. Physiol. 71, 1563–1567 (1991). Mutations in COQ2 in familial and sporadic 67. Schwarzacher, S. W., Rub, U. & Deller, T.
28. Cooke, J. et al. Sitting and standing blood multiple-system atrophy. N. Engl. J. Med. 369, Neuroanatomical characteristics of the human
pressure measurements are not accurate for the 233–244 (2013). pre-Bötzinger complex and its involvement in
diagnosis of orthostatic hypotension. QJM 102, 48. Tada, M. et al. Early development of autonomic neurodegenerative brainstem diseases. Brain
335–339 (2009). dysfunction may predict poor prognosis in 134, 24–35 (2011).
29. Low, P. A. Testing the autonomic nervous patients with multiple system atrophy. Arch. 68. Benarroch, E. E., Schmeichel, A. M., Low, P. A. &
system. Semin. Neurol. 23, 407–421 (2003). Neurol. 64, 256–260 (2007). Parisi, J. E. Depletion of putative chemosensitive
30. Freeman, R. Assessment of cardiovascular 49. O’Sullivan, S. S. et al. Clinical outcomes of respiratory neurons in the ventral medullary
autonomic function. Clin. Neurophysiol. 117, progressive supranuclear palsy and multiple surface in multiple system atrophy. Brain 130,
716–730 (2006). system atrophy. Brain 131, 1362–1372 (2008). 469–475 (2007).
31. Hilz, M. J. & Dutsch, M. Quantitative studies of 50. Wenning, G. K. et al. The natural history of 69. Tada, M. et al. Depletion of medullary
autonomic function. Muscle Nerve 33, 6–20 multiple system atrophy: a prospective European serotonergic neurons in patients with multiple
(2006). cohort study. Lancet Neurol. 12, 264–274 (2013). system atrophy who succumbed to sudden
32. Lauria, G. et al. European Federation of 51. Wenning, G. K. et al. Time course of symptomatic death. Brain 132, 1810–1819 (2009).
Neurological Societies/Peripheral Nerve Society orthostatic hypotension and urinary incontinence 70. Braak, H. et al. Stanley Fahn Lecture 2005:
Guideline on the use of skin biopsy in the in patients with postmortem confirmed the staging procedure for the inclusion body
diagnosis of small fiber neuropathy. Report of a parkinsonian syndromes: a clinicopathological pathology associated with sporadic Parkinson’s
joint task force of the European Federation of study. J. Neurol. Neurosurg. Psychiatry 67, disease reconsidered. Mov. Disord. 21,
Neurological Societies and the Peripheral Nerve 620–623 (1999). 2042–2051 (2006).
Society. Eur. J. Neurol. 17, 903–912, e44–e49 52. Oppenheimer, D. R. Lateral horn cells in 71. Dickson, D. W. et al. Evidence that incidental
(2010). progressive autonomic failure. J. Neurol. Sci. 46, Lewy body disease is pre-symptomatic
33. Donadio, V. et al. Peripheral autonomic 393–404 (1980). Parkinson’s disease. Acta Neuropathol. 115,
neuropathy: diagnostic contribution of skin 53. Benarroch, E. E., Smithson, I. L., Low, P. A. & 437–444 (2008).
biopsy. J. Neuropathol. Exp. Neurol. 71, Parisi, J. E. Depletion of catecholaminergic 72. Jellinger, K. A. A critical reappraisal of current
1000–1008 (2012). neurons of the rostral ventrolateral medulla in staging of Lewy-related pathology in human
34. Gibbons, C. H., Illigens, B. M., Wang, N. & multiple systems atrophy with autonomic failure. brain. Acta Neuropathol. 116, 1–16 (2008).
Freeman, R. Quantification of sweat gland Ann. Neurol. 43, 156–163 (1998). 73. Beach, T. G. et al. Multi-organ distribution of
innervation: a clinical-pathologic correlation. 54. Winge, K. & Fowler, C. J. Bladder dysfunction in phosphorylated alpha-synuclein histopathology
Neurology 72, 1479–1486 (2009). Parkinsonism: mechanisms, prevalence, in subjects with Lewy body disorders. Acta
35. Nolano, M. et al. Quantification of pilomotor symptoms, and management. Mov. Disord. 21, Neuropathol. 119, 689–702 (2010).
nerves: a new tool to evaluate autonomic 737–745 (2006). 74. Shishido, T. et al. α‑Synuclein accumulation in
involvement in diabetes. Neurology 75, 55. Benarroch, E. E. & Schmeichel, A. M. Depletion skin nerve fibers revealed by skin biopsy in pure
1089–1097 (2010). of corticotrophin-releasing factor neurons in the autonomic failure. Neurology 74, 608–610
36. Libbey, C. A., Skinner, M. & Cohen, A. S. Use of pontine micturition area in multiple system (2010).
abdominal fat tissue aspirate in the diagnosis atrophy. Ann. Neurol. 50, 640–645 (2001). 75. Wang, N., Gibbons, C. H., Lafo, J. & Freeman, R.
of systemic amyloidosis. Arch. Intern. Med. 143, 56. Kennedy, P. G. & Duchen, L. W. A quantitative α‑Synuclein in cutaneous autonomic nerves.
1549–1552 (1983). study of intermediolateral column cells in motor Neurology 81, 1604–1610 (2013).
37. Kyle, R. A. & Greipp, P. R. Amyloidosis (AL). neuron disease and the Shy–Drager syndrome. 76. Polinsky, R. J. Clinical autonomic
Clinical and laboratory features in 229 cases. J. Neurol. Neurosurg. Psychiatry 48, 1103–1106 neuropharmacology. Neurol. Clin. 8, 77–92
Mayo Clin. Proc. 58, 665–683 (1983). (1985). (1990).

10  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

77. Klein, C. M. et al. The spectrum of autoimmune 97. Varrone, A., Marek, K. L., Jennings, D., ganglionopathy: treatment by plasma exchanges
autonomic neuropathies. Ann. Neurol. 53, Innis, R. B. & Seibyl, J. P. [123I]β-CIT SPECT and rituximab. Clin. Auton. Res. 19, 259–262
752–758 (2003). imaging demonstrates reduced density of (2009).
78. Pfeiffer, R. F. Gastrointestinal dysfunction in striatal dopamine transporters in Parkinson’s 116. Iodice, V. et al. Efficacy of immunotherapy
Parkinson’s disease. Parkinsonism Relat. Disord. disease and multiple system atrophy. Mov. in seropositive and seronegative putative
17, 10–15 (2011). Disord. 16, 1023–1032 (2001). autoimmune autonomic ganglionopathy.
79. Siddiqui, M. F., Rast, S., Lynn, M. J., Auchus, A. P. 98. King, A. E., Mintz, J. & Royall, D. R. Meta-analysis Neurology 72, 2002–2008 (2009).
& Pfeiffer, R. F. Autonomic dysfunction in of 123I-MIBG cardiac scintigraphy for the 117. Sandroni, P. et al. Idiopathic autonomic
Parkinson’s disease: a comprehensive symptom diagnosis of Lewy body-related disorders. neuropathy: comparison of cases seropositive
survey. Parkinsonism Relat. Disord. 8, 277–284 Mov. Disord. 26, 1218–1224 (2011). and seronegative for ganglionic acetylcholine
(2002). 99. Goldstein, D. S., Holmes, C., Cannon, R. O. 3rd, receptor antibody. Arch. Neurol. 61, 44–48
80. Abbott, R. D. et al. Frequency of bowel Eisenhofer, G. & Kopin, I. J. Sympathetic (2004).
movements and the future risk of Parkinson’s cardioneuropathy in dysautonomias. N. Engl. 118. Lucchinetti, C. F., Kimmel, D. W. & Lennon, V. A.
disease. Neurology 57, 456–462 (2001). J. Med. 336, 696–702 (1997). Paraneoplastic and oncologic profiles of patients
81. Savica, R. et al. Medical records documentation 100. Treglia, G. et al. Diagnostic performance seropositive for type 1 antineuronal nuclear
of constipation preceding Parkinson disease: of iodine‑123‑metaiodobenzylguanidine autoantibodies. Neurology 50, 652–657 (1998).
a case–control study. Neurology 73, 1752–1758 scintigraphy in differential diagnosis between 119. Winkler, A. S., Dean, A., Hu, M., Gregson, N. &
(2009). Parkinson’s disease and multiple-system atrophy: Chaudhuri, K. R. Phenotypic and neuropathologic
82. Proulx, M., de Courval, F. P., Wiseman, M. A. & a systematic review and a meta-analysis. Clin. heterogeneity of anti-Hu antibody-related
Panisset, M. Salivary production in Parkinson’s Neurol. Neurosurg. 113, 823–829 (2011). paraneoplastic syndrome presenting with
disease. Mov. Disord. 20, 204–207 (2005). 101. Nagayama, H. et al. Abnormal cardiac progressive dysautonomia: report of two cases.
83. Cersosimo, M. G. et al. Hyposialorrhea as an [123I]‑meta‑iodobenzylguanidine uptake in Clin. Auton. Res. 11, 115–118 (2001).
early manifestation of Parkinson disease. multiple system atrophy. Mov. Disord. 25, 120. Chinn, J. S. & Schuffler, M. D. Paraneoplastic
Auton. Neurosci. 150, 150–151 (2009). 1744–1747 (2010). visceral neuropathy as a cause of severe
84. Del Tredici, K., Hawkes, C. H., Ghebremedhin, E. 102. McKeith, I. G. Consensus guidelines for the gastrointestinal motor dysfunction.
& Braak, H. Lewy pathology in the submandibular clinical and pathologic diagnosis of dementia Gastroenterology 95, 1279–1286 (1988).
gland of individuals with incidental Lewy body with Lewy bodies (DLB): report of the 121. De Giorgio, R. et al. Inflammatory neuropathies
disease and sporadic Parkinson’s disease. Consortium on DLB International Workshop. of the enteric nervous system. Gastroenterology
Acta Neuropathol. 119, 703–713 (2010). J. Alzheimers. Dis. 9, 417–423 (2006). 126, 1872–1883 (2004).
85. Edwards, L. L., Quigley, E. M., Harned, R. K., 103. Stubendorff, K., Aarsland, D., Minthon, L. & 122. Colan, R. V., Snead, O. C. 3rd, Oh, S. J. &
Hofman, R. & Pfeiffer, R. F. Characterization Londos, E. The impact of autonomic dysfunction Kashlan, M. B. Acute autonomic and sensory
of swallowing and defecation in Parkinson’s on survival in patients with dementia with Lewy neuropathy. Ann. Neurol. 8, 441–444 (1980).
disease. Am. J. Gastroenterol. 89, 15–25 bodies and Parkinson’s disease with dementia. 123. Koike, H. et al. Clinicopathological features of
(1994). PLoS ONE 7, e45451 (2012). acute autonomic and sensory neuropathy. Brain
86. Castell, J. A. et al. Manometric abnormalities 104. Kanazawa, M. et al. An autopsy case of 133, 2881–2896 (2010).
of the oesophagus in patients with Parkinson’s dementia with Lewy bodies showing autonomic 124. Pavlakis, P. P. et al. Peripheral neuropathies in
disease. Neurogastroenterol. Motil. 13, 361–364 failure and dementia as the initial symptoms. Sjögren syndrome: a new reappraisal. J. Neurol.
(2001). Mov. Disord. 22, 1212–1213 (2007). Neurosurg. Psychiatry 82, 798–802 (2011).
87. Wakabayashi, K., Takahashi, H., Takeda, S., 105. Thaisetthawatkul, P. et al. Autonomic dysfunction 125. Dawson, L., Tobin, A., Smith, P. & Gordon, T.
Ohama, E. & Ikuta, F. Parkinson’s disease: in dementia with Lewy bodies. Neurology 62, Antimuscarinic antibodies in Sjögren’s
the presence of Lewy bodies in Auerbach’s 1804–1809 (2004). syndrome: where are we, and where are we
and Meissner’s plexuses. Acta Neuropathol. 76, 106. Leehey, M. A. Fragile X‑associated tremor/ataxia going? Arthritis Rheum. 52, 2984–2995 (2005).
217–221 (1988). syndrome: clinical phenotype, diagnosis, and 126. Nakamura, Y. et al. High prevalence of
88. Sakakibara, R., Uchiyama, T., Yamanishi, T., treatment. J. Investig. Med. 57, 830–836 (2009). autoantibodies to muscarinic‑3 acetylcholine
Shirai, K. & Hattori, T. Bladder and bowel 107. Guaraldi, P. et al. Isolated noradrenergic failure in receptor in patients with juvenile-onset Sjögren
dysfunction in Parkinson’s disease. J. Neural adult-onset autosomal dominant leukodystrophy. syndrome. Ann. Rheum. Dis. 67, 136–137
Transm. 115, 443–460 (2008). Auton. Neurosci. 159, 123–126 (2011). (2008).
89. Sharabi, Y. & Goldstein, D. S. Mechanisms 108. Mead, S. et al. A novel prion disease associated 127. Park, K., Park, S. & Jackson, M. W. The inhibitory
of orthostatic hypotension and supine with diarrhea and autonomic neuropathy. N. Engl. effects of antimuscarinic autoantibodies in the
hypertension in Parkinson disease. J. Neurol. J. Med. 369, 1904–1914 (2013). sera of primary Sjogren syndrome patients on
Sci. 310, 123–128 (2011). 109. Koike, H., Watanabe, H. & Sobue, G. The the gastrointestinal motility. Mol. Immunol. 56,
90. Winge, K., Skau, A. M., Stimpel, H., spectrum of immune-mediated autonomic 583–587 (2013).
Nielsen, K. K. & Werdelin, L. Prevalence of neuropathies: insights from the 128. Kondo, T. et al. Autoimmune autonomic
bladder dysfunction in Parkinsons disease. clinicopathological features. J. Neurol. ganglionopathy with Sjögren’s syndrome:
Neurourol. Urodyn. 25, 116–122 (2006). Neurosurg. Psychiatry 84, 98–106 (2013). significance of ganglionic acetylcholine receptor
91. Blackett, H., Walker, R. & Wood, B. Urinary 110. Suarez, G. A., Fealey, R. D., Camilleri, M. & antibody and therapeutic approach. Auton.
dysfunction in Parkinson’s disease: a review. Low, P. A. Idiopathic autonomic neuropathy: Neurosci. 146, 33–35 (2009).
Parkinsonism Relat. Disord. 15, 81–87 (2009). clinical, neurophysiologic, and follow-up studies 129. Tesfaye, S. et al. Diabetic neuropathies: update
92. Winge, K. & Nielsen, K. K. Bladder dysfunction on 27 patients. Neurology 44, 1675–1682 on definitions, diagnostic criteria, estimation of
in advanced Parkinson’s disease. Neurourol. (1994). severity, and treatments. Diabetes Care 33,
Urodyn. 31, 1279–1283 (2012). 111. Gibbons, C. H. & Freeman, R. Antibody titers 2285–2293 (2010).
93. Sakakibara, R., Uchiyama, T., Yamanishi, T. & predict clinical features of autoimmune 130. Ziegler, D., Gries, F. A., Spuler, M. & Lessmann, F.
Kishi, M. Sphincter EMG as a diagnostic tool in autonomic ganglionopathy. Auton. Neurosci. 146, The epidemiology of diabetic neuropathy.
autonomic disorders. Clin. Auton. Res. 19, 20–31 8–12 (2009). Diabetic Cardiovascular Autonomic Neuropathy
(2009). 112. Vernino, S., Cheshire, W. P. & Lennon, V. A. Multicenter Study Group. J. Diabetes
94. Seppi, K. et al. Progression of putaminal Myasthenia gravis with autoimmune autonomic Complications 6, 49–57 (1992).
degeneration in multiple system atrophy: a serial neuropathy. Auton. Neurosci. 88, 187–192 131. Low, P. A. et al. Autonomic symptoms and
diffusion MR study. Neuroimage 31, 240–245 (2001). diabetic neuropathy: a population-based study.
(2006). 113. Peltier, A. C. et al. Coexistent autoimmune Diabetes Care 27, 2942–2947 (2004).
95. Feigin, A. et al. Tc‑99m ethylene cysteinate autonomic ganglionopathy and myasthenia 132. England, J. D. et al. Practice Parameter:
dimer SPECT in the differential diagnosis of gravis associated with non‑small‑cell lung evaluation of distal symmetric polyneuropathy:
parkinsonism. Mov. Disord. 17, 1265–1270 cancer. Muscle Nerve 41, 416–419 (2010). role of autonomic testing, nerve biopsy, and skin
(2002). 114. Gibbons, C. H., Vernino, S. A. & Freeman, R. biopsy (an evidence-based review). Report of the
96. Kwon, K. Y., Choi, C. G., Kim, J. S., Lee, M. C. Combined immunomodulatory therapy in American Academy of Neurology, American
& Chung, S. J. Comparison of brain MRI and autoimmune autonomic ganglionopathy. Association of Neuromuscular and
18
F-FDG PET in the differential diagnosis of Arch. Neurol. 65, 213–217 (2008). Electrodiagnostic Medicine, and American
multiple system atrophy from Parkinson’s 115. Imrich, R., Vernino, S., Eldadah, B. A., Holmes, C. Academy of Physical Medicine and Rehabilitation.
disease. Mov. Disord. 22, 2352–2358 (2007). & Goldstein, D. S. Autoimmune autonomic Neurology 72, 177–184 (2009).

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION  |  11


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

133. Vinik, A. I. & Ziegler, D. Diabetic cardiovascular 142. Gibbons, C. H. & Freeman, R. Treatment- 150. Yamashita, T. et al. Long-term survival after liver
autonomic neuropathy. Circulation 115, induced diabetic neuropathy: a reversible transplantation in patients with familial amyloid
387–397 (2007). painful autonomic neuropathy. Ann. Neurol. 67, polyneuropathy. Neurology 78, 637–643 (2012).
134. Maleki, D. et al. Gastrointestinal tract symptoms 534–541 (2010). 151. Coelho, T. et al. Tafamidis for transthyretin
among persons with diabetes mellitus in the 143. Wang, A. K., Fealey, R. D., Gehrking, T. L. & familial amyloid polyneuropathy: a randomized,
community. Arch. Intern. Med. 160, 2808–2816 Low, P. A. Patterns of neuropathy and autonomic controlled trial. Neurology 79, 785–792 (2012).
(2000). failure in patients with amyloidosis. Mayo Clin. 152. Robinson-Papp, J., Sharma, S., Simpson, D. M.
135. Lysy, J., Israeli, E. & Goldin, E. The prevalence Proc. 83, 1226–1230 (2008). & Morgello, S. Autonomic dysfunction is
of chronic diarrhea among diabetic patients. 144. Cappellari, M. et al. Variable presentations of common in HIV and associated with distal
Am. J. Gastroenterol. 94, 2165–2170 (1999). TTR-related familial amyloid polyneuropathy in symmetric polyneuropathy. J. Neurovirol. 19,
136. Schiller, L. R. et al. Pathogenesis of fecal seventeen patients. J. Peripher. Nerv. Syst. 16, 172–180 (2013).
incontinence in diabetes mellitus: evidence 119–129 (2011). 153. Dutsch, M. et al. Small fiber dysfunction
for internal‑anal‑sphincter dysfunction. N. Engl. 145. Rajkumar, S. V., Gertz, M. A. & Kyle, R. A. predominates in Fabry neuropathy. J. Clin.
J. Med. 307, 1666–1671 (1982). Prognosis of patients with primary systemic Neurophysiol. 19, 575–586 (2002).
137. Bradley, W. E. Diagnosis of urinary bladder amyloidosis who present with dominant 154. Dib-Hajj, S. D., Cummins, T. R., Black, J. A. &
dysfunction in diabetes mellitus. Ann. Intern. neuropathy. Am. J. Med. 104, 232–237 (1998). Waxman, S. G. From genes to pain: Nav1.7 and
Med. 92, 323–326 (1980). 146. Gertz, M. A. et al. Stem cell transplantation human pain disorders. Trends Neurosci. 30,
138. Kaplan, S. A., Te, A. E. & Blaivas, J. G. for the management of primary systemic 555–563 (2007).
Urodynamic findings in patients with diabetic amyloidosis. Am. J. Med. 113, 549–555 155. Faber, C. G. et al. Gain of function Nav1.7
cystopathy. J. Urol. 153, 342–344 (1995). (2002). mutations in idiopathic small fiber neuropathy.
139. Bacon, C. G. et al. Association of type and 147. Planté-Bordeneuve, V. & Said, G. Familial amyloid Ann. Neurol. 71, 26–39 (2012).
duration of diabetes with erectile dysfunction polyneuropathy. Lancet Neurol. 10, 1086–1097 156. Rush, A. M. et al. A single sodium channel
in a large cohort of men. Diabetes Care 25, (2011). mutation produces hyper- or hypoexcitability in
1458–1463 (2002). 148. Said, G. & Planté-Bordeneuve, V. TTR-familial different types of neurons. Proc. Natl Acad. Sci.
140. Malavige, L. S. & Levy, J. C. Erectile dysfunction amyloid polyneuropathy—neurological aspects. USA 103, 8245–8250 (2006).
in diabetes mellitus. J. Sex. Med. 6, 1232–1247 Amyloid 19 (Suppl. 1), 25–27 (2012). 157. Han, C. et al. Functional profiles of SCN9A
(2009). 149. Arruda-Olson, A. M. et al. Genotype, variants in dorsal root ganglion neurons and
141. Watkins, P. J. Facial sweating after food: a new echocardiography, and survival in familial superior cervical ganglion neurons correlate with
sign of diabetic autonomic neuropathy. Br. Med. J. transthyretin amyloidosis. Amyloid 20, 263–268 autonomic symptoms in small fibre neuropathy.
1, 583–587 (1973). (2013). Brain 135, 2613–2628 (2012).

12  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved

You might also like