SlideShare a Scribd company logo
1 of 12
Download to read offline
NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 1
Department of
Neurology, Mayo Clinic,
200 First Street SW,
Rochester, MN 55905,
USA.
benarroch.eduardo@
mayo.edu
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
Autonomic disorders manifest with autonomic failure
or hyperactivity, which may be generalized or focal.
Autonomic failure can affect the sympathetic, para­sympa­
thetic or enteric nervous systems, either in isolation or in
various combinations, and can result from lesions at any
level of the CNS or PNS. Sympathetic failure manifests
primarily with orthostatic hypotension and anhidrosis
(absence of sweating), cranial parasympathetic failure
with intolerance to light, dry eyes (xerophthalmia) and
dry mouth (xerostomia), sacral parasympathetic failure
with urinary retention and erectile dysfunction, and
enteric nervous system (ENS) failure with gastroparesis
and constipation. In some cases, postural tachycardia
syndrome is a manifestation of an underlying autonomic
neuropathy. Many symptoms attributed to ‘dysautonomia’
in otherwise healthy young patients, such as gastroparesis
or urinary retention, are rarely associated with objective
evidence of autonomic failure.
This Review will focus on the clinical approach to
patients with generalized autonomic failure. The degen­
erative synucleinopathies, autonomic ganglionopathies
and autonomic neuropathies have been reviewed in
detail elsewhere.1–5
Rather than providing an exhaustive
discussion of the wide spectrum of disorders associated
with autonomic failure, this article will focus on specific
examples to emphasize clinical cues and outline recent
concepts of the underlying pathobiology.
Clinical approach to autonomic failure
Disorders associated with autonomic failure can be clas­
sified according to the type and severity of autonomic
manifestations, associated neurological symptoms, and
temporal profile (Box 1). The temporal profile of onset
and progression has important implications for diag­
nosis, and for guidance of further laboratory evalua­
tion (Figure 1). Isolated autonomic failure of acute or
subacute onset suggests an immune cause such as auto­
immune autonomic ganglionopathy (AAG), including
paraneoplastic autonomic neuropathy, or a toxic cause
such as effects of medications. Pure autonomic failure
(PAF) refers to the slow development of general­
ized
auto­
nomic failure in the absence of motor or sensory
symp­
toms. Manifestations associated with chronic and
pro­
gressive generalized autonomic failure, such as ataxia
or parkinsonism, suggest a degenerative cause, typi­
cally a synuclein­
opathy such as multiple system atrophy
(MSA), or a Lewy body disorder such as Parkinson
disease (PD) or dementia with Lewy bodies (DLB). Auto­
nomic failure can also be a salient feature of various types
of peripheral neuropathy, including distal sensori­
motor
peripheral neuropathy, painful or ataxic forms of sensory
ganglionopathy, distal painful small fibre ­
neuropathy
(SFN), and acute motor polyradiculopathy.
Manifestations and pathophysiology
Orthostatic hypotension
Orthostatic hypotension is defined as a sustained reduc­
tion in systolic blood pressure of at least 20 mmHg or
diastolic blood pressure of 10 mmHg within 3 min of
Competing interests
The author declares no competing interests.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol
standing or head-up tilt.6
Some patients with neuro­
genic orthostatic hypotension have postprandial hypo­
tension.7
Manifestations of orthostatic hypotension
include lightheadedness, blurred vision and neck pain
(coat hanger distribution) among others; the symptoms
are worse in the morning, after meals, or on exposure
to heat.8,9
Neurogenic orthostatic hypotension is the mani­
festation of impaired sympathetically mediated vaso­
constriction of skeletal muscle and mesenteric vessels in
response to baroreceptor unloading due to orthostatic
stress. Neurogenic orthostatic hypotension may occur
as a consequence of disorders affecting the barosensi­
tive sympathoexcitatory neurons in the rostral ventro­
lateral medulla (for example, MSA); spinal preganglionic
sympathetic neurons (for example, MSA or PD); auto­
nomic ganglia (for example, AAG or PAF); unmyelin­
ated axons (SFN); noradrenaline availability (for
example, dopamine β‑hydroxylase deficiency); or vas­
cular α1-adrenergic receptors (typically as a side effect
of drugs).10
Anhidrosis
Sweating is an important thermoregulatory activity
mediated by the sympathetic nervous system through
cholinergic activation of muscarinic M3
receptors in
the eccrine sweat glands. Depending on its distribu­
tion and severity, anhidrosis might be asymptomatic
or might manifest with compensatory hyperhidrosis
(in unaffected areas) or heat intolerance. Anhidrosis
in autonomic failure may reflect impairment of central
thermoregulatory pathways, spinal preganglionic
sudomotor units, cholinergic sympathetic ganglion
neurons, peripheral unmyelinated axons, or M3
­receptors
at the sudomotor unit.11
Neurogenic bladder and sexual dysfunction
Normal bladder function includes a urine storage
phase and a micturition phase, which are controlled
at all levels of the neuraxis.12
Urine storage depends on
spinal reflexes mediated by the lumbosacral sympathetic
noradrenergic and sacral cholinergic motor neurons of
the Onuf nucleus. Normal micturition depends on a
Key points
■
■ Autonomic failure can occur in isolation, in association with peripheral
neuropathy, or as a manifestation of a neurodegenerative disorder
■
■ Disorders associated with autonomic failure can be classified according to
the type and severity of autonomic manifestations, associated neurological
symptoms, and temporal profile
■
■ Acute or subacute autonomic failure suggests an autoimmune autonomic
ganglionopathy, which may be paraneoplastic
■
■ In patients with parkinsonism and autonomic failure, early onset and
progression of orthostatic hypotension or urogenital dysfunction, urinary
incontinence, generalized anhidrosis, and/or laryngeal stridor are highly
suggestive of multiple system atrophy
■
■ Autonomic failure can occur with any type of diabetic or amyloid neuropathy
■
■ Important causes of painful neuropathy associated with autonomic failure
include Sjögren syndrome, HIV infections, Fabry disease, and sodium
channelopathies
supraspinal reflex coordinated by the pontine mictur­
ition centre, which activates the sacral pre­
gangli­
onic
para­
sympa­
thetic output to the bladder detrusor while
­
simultaneously inhibiting the Onuf nucleus.13
Neurogenic bladder can manifest with detrusor over­
activity or underactivity.14
Detrusor overactivity pro­
duces urinary urgency with or without incontinence,
urinary frequency, and nocturia. Reduced detrusor
activity leads to incomplete bladder emptying, increased
post-void residual, low peak urinary flow rate and, even­
tually, urinary retention and overflow incontinence.
Impaired micturition can result from lesions affecting
the bladder afferents, sacral parasympathetic neurons or
their axons, or cholinergic muscarinic neurotransmis­
sion. Neurogenic bladder is commonly associated with
erectile and ejaculatory dysfunction in men and poor
vaginal lubrication in women. Erectile dysfunction
reflects impaired sacral parasympathetic output and
nitric oxide release in the erectile tissue.
Box 1 | Neurological causes of autonomic failure
A. Isolated autonomic failure
1. Acute or subacute
(a) Autoimmune autonomic ganglionopathy
(b) Paraneoplastic autonomic neuropathy
2. Progressive
(a) Pure autonomic failure
B. Progressive autonomic failure associated with
parkinsonism, ataxia or dementia
1. Multiple system atrophy
2. Lewy body disorders
(a) Parkinson disease
(b) Dementia with Lewy bodies
3. Others
(a) Familial leukoencephalopathies
(b) Prion disorders
C. Autonomic failure associated with peripheral
neuropathy
1. Chronic sensorimotor neuropathies
(a) Diabetes
(b) Amyloidosis
(c) Other metabolic disorders (vitamin B12
deficiency,
uraemia)
(d) Toxic neuropathies
2. Sensory ganglionopathies
(a) Sjögren syndrome
(b) Paraneoplastic
3. Distal painful neuropathies
(a) Diabetes
(b) Amyloidosis
(c) Idiopathic (sodium channelopathies)
(d) Infectious (HIV)
(e) Hereditary
(i) Hereditary sensory and autonomic neuropathy
(ii) Fabry disease
(iii) Sodium channelopathies
4. Acute or subacute motor polyradiculopathy or
neuropathy
(a) Guillain–Barré syndrome
(b) Porphyria
5. Acute autonomic and sensory neuropathy
6. Ross syndrome (segmental anhidrosis, Adie pupils
and areflexia).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 3
Gastrointestinal dysmotility
Control of gastrointestinal motility depends on intrin­
sic reflexes mediated by the ENS, and their modulation
by vagal and paravertebral sympathetic inputs. Upper
gastro­
intestinal motility is primarily controlled by the
vagal reflexes involving the nucleus of the solitary tract
and dorsal motor nucleus of the vagus in the medulla;15
peristalsis in the lower gastrointestinal tract depends
primarily on local ENS reflexes;16
and paravertebral
­
sympathetic reflexes inhibit gastrointestinal motility.
Symptoms of delayed oesophageal transit include
dysphagia and regurgitation. Delayed gastric emptying
produces early satiety, anorexia, nausea, bloating, belch­
ing, postprandial vomiting, and pain. Lower gastro­
intestinal dysmotility manifests with constipation and,
­occasionally, diarrhoea.
Evaluation
Clinical
In addition to a careful history and general physical and
neurological examination, clinical evaluation of patients
with suspected autonomic failure includes assessment of
pupil size, symmetry and reactivity with both bright and
dim light; measurement of blood pressure and heart rate
after 2min in the supine position and at 1min and 2min
after standing; and examination of the skin to identify
areas of localized or generalized absence of or excessive
sweating, and changes in skin temperature or colour in
the hands and feet.
Laboratory tests
General laboratory tests that should be performed in
patients with autonomic failure include determination
of serum glucose (or in some cases haemoglobin A1c),
thyroid-stimulating hormone and vitamin B12
levels.
Serum and urine protein electrophoresis with immuno­
fixation, including light-chain quantitation, are indi­
cated to detect amyloid light-chain (AL) amyloidosis.17
SSA and SSB antibody testing for Sjögren syndrome may
be helpful in some cases. Determination of ganglionic
nicotinic acetylcholine receptor (gnAChR) anti­bodies,18
as well as paraneoplastic antibodies (particularly anti-
Hu, P/Q and N‑type voltage-gated calcium channel
and voltage-gated potassium complex anti­
bodies), is
indicated in all patients with subacute onset of symp­
toms.18,19
Determination of forearm venous catecho­
lamines, including noradrenaline, dopamine and
adrenaline, in the supine position and after 5–10 min
of standing may be helpful in some cases; the results
require careful interpretation, however, as they can be
Parkinsonism, ataxia
Autonomic studies, neuroimaging (MRI),
polysomnogram, urodynamic studies,
neuropsychometrics
Autonomic studies, electromyography/nerve
conduction studies, FBG, SPEP
, UPEP
,
abdominal fat aspirate, ANCA, HIV,
α-galactosidase, punch skin biopsy
Associated
manifestations
Onset/
subtype
Main or
typical causes
Specific
evaluation*
Autonomic studies
Autoantibodies
(gnAChR,
paraneoplastic)
Sjögren
syndrome
Paraneoplastic
Diabetes
Amyloidosis
‘Pure’
autonomic
failure
Multiple
system
atrophy
Lewy body
disorders
■ PD
■ DLB
Autoimmune
autonomic
ganglionopathy
Paraneoplastic
autonomic
ganglionopathy
Vasculitis
HIV
Idiopathic
Fabry disease
Hereditary
Peripheral neuropathy
None (‘pure’ autonomic failure)
Chronic, progressive Subacute
sensory
Distal painful
Distal
sensorimotor
or any other
type
Acute or subacute
Autoantibodies
(SSA/SSB, paraneoplastic)
Salivary gland biopsy
Autonomic failure
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 B12
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.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol
affected by concomitant use of drugs or impaired pre­
synaptic noradrenaline reuptake.20
Low or undetectable
noradrenaline and adrenaline levels associated with
increased dopamine levels indicate a deficit of dopamine
β‑hydroxylase as the cause of orthostatic hypotension.21
Autonomic function tests
Autonomic laboratory evaluation is indicated to deter­
mine the extent and severity of autonomic failure; to
detect autonomic failure in patients with parkinsonism
or ataxia; to assess small fibre function in patients with
peripheral neuropathy or ganglionopathy; to evalu­
ate patients with symptoms of orthostatic intolerance;
and to obtain objective evidence of disease progression
or response to medications.22,23
The most commonly
used tests assess sudomotor, cardiovagal and adren­
ergic vasomotor functions. Sudomotor function tests
include the thermoregulatory sweat test (TST), which
assesses the integrity of central and peripheral sudo­
motor pathways,22
and sudomotor axon reflex tests such
as the quanti­tative sudomotor axon reflex test (QSART)23
and the quanti­
tative direct and indirect test of sudo­
motor function (QDIRT),24
which assess the peripheral
sympa­thetic cholinergic innervation of the sweat glands.
Tests that assess vagal control of the sinus node (cardio­
vagal function) include the heart rate response to deep
breathing,25
and the heart rate response to the Valsalva
manoeuvre, or Valsalva ratio.26
Tests that indirectly assess
sympathetic vasomotor function include the beat-to-beat
blood pressure profile during the Valsalva manoeuvre27
and the blood pressure response to head-up tilt.28
The
physiological basis, methods, normal and abnormal
responses, and pitfalls of these and other autonomic
­function tests have been the subject of several reviews.29–31
Other tests
Electromyography is indicated for patients with periph­
eral neuropathy or ganglionopathy, and MRI may be
helpful in evaluation of central autonomic disorders.
Gastrointestinal motility and urodynamic studies might
be indicated in some cases. Polysomnography to detect
sleep-related respiratory dysfunction is indicated in all
patients with suspected MSA.
Tissue biopsy
Skin biopsy with assessment of intraepidermal nerve
fibre density (IENFD) can be useful in the evaluation
of SFN;32,33
immunocytochemical markers allow quanti­
fication of the density of innervation of sweat gland34
and pilomotor35
nerves. Abdominal fat aspirate or sural
nerve biopsy are indicated to evaluate for amyloidosis.36,37
Immunostaining and laser microdissection, along with
mass spectrometry-based proteomic analysis of amyloid
deposition, allow identification of the specific subtype of
amyloid protein.38
Neurodegenerative synucleinopathies
Autonomic failure is an important manifestation of
neuro­
degenerative disorders characterized by the pres­
ence of intracellular inclusions containing α‑synuclein.
These conditions include MSA,1,39–41
and Lewy body
­
disorders such as PD, DLB or PAF.42,43
Multiple system atrophy
MSA was initially defined as a sporadic neurodegenera­
tive disease characterized by any combination of auto­
nomic failure, parkinsonism, cerebellar ataxia and/or
pyramidal signs.44
The prevalence of MSA has been esti­
mated at 4–8 cases per 100,000 people, and its incidence
varies from 0.6 cases per 100,000 person-years in the
general population to 3 cases per 100,000 person-years
in people above 50 years of age.1,40
The neuropathological hallmark of MSA is the accumu­
lation of α‑synuclein-immunoreactive glial cytoplasmic
inclusions in oligodendrocytes, and neuronal loss in the
striatum, substantia nigra pars compacta, pontine nuclei,
inferior olivary nuclei, cerebellum, and premotor auto­
nomic nuclei.45
According to current criteria, probable
MSA is defined as a sporadic, progressive, adult-onset
(over 30 years of age) disease characterized by autonomic
failure, with urinary incontinence and erectile dysfunc­
tion (in males) or orthostatic hypotension (blood pres­
sure decrease ≥30mmHg systolic or ≥15mmHg diastolic
within 3min of standing), as well as parkinsonism poorly
responsive to levodopa (MSA‑P), or cerebellar syndrome
(MSA‑C).39
A large European study including 437 patients
with probable (72%) or possible (28%) MSA showed that
symptomatic autonomic failure was present in almost all
cases; the most frequent manifestations were urinary dys­
function (83%) and orthostatic hypotension (75%).41
In a
study on 29 autopsy-confirmed cases, a pattern consist­
ing of severe and progressive adrenergic and sudomotor
failure was highly predictive of MSA.46
Although MSA is still generally regarded as a sporadic
disease, several studies have identified familial cases.1
A recent study identified mutations of the coenzyme Q2
(COQ2) gene in Japanese families with MSA, and COQ2
variants were also associated with an increased risk of
sporadic MSA.47
Median survival from symptom onset in MSA is
8–10 years, but the spectrum ranges from 4–15 years.
Natural history studies indicate that presence of the
parkinsonian variant, early onset of autonomic failure,
incomplete bladder emptying, and shorter duration
of symptoms at baseline are all factors that predict
shorter survival.48–50
In MSA, orthostatic hypotension occurs at earlier
stages and is more severe than in PD.51
Orthostatic
hypotension in MSA reflects involvement of pre­
gangli­
onic sympathetic neurons52
and sympathoexcitatory
neurons of the rostral ventrolateral medulla.53
Urogenital
manifestations may herald the onset of MSA.51
Bladder
dysfunction in MSA is characterized by urinary urgency,
followed by incontinence and incomplete bladder empty­
ing. These manifestations result from a combination of
detrusor hyperreflexia and urethral sphincter weakness
followed by detrusor contraction failure,54
and they
reflect involvement of the pontine micturition centre,55
sacral preganglionic neurons56
and Onuf nucleus.57
Erectile dysfunction is almost always present in male
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 5
patients with MSA.58
Upper gastrointestinal symptoms,
constipation and faecal incontinence occur frequently;
anal sphincter electromyography shows large motor
unit potentials indicating denervation due to loss of
Onuf nucleus motor neurons.59
Anhidrosis occurs in the
majority of patients with MSA, and the percentage of
the body affected by anhidrosis may help to differentiate
MSA from PD.60,61
An important manifestation of disrupted brainstem
homeostatic mechanisms in MSA is sleep-related dis­
ordered breathing, including sleep apnoea and laryn­
geal stridor, which occur in up to 70% of patients with
MSA.62–64
Whereas the development of central hypo­
ventilation and laryngeal stridor is closely related to the
severity of autonomic failure, respiratory manifestations
can occur early in the course of MSA.65
Laryngeal dys­
tonia with inspiratory adduction of the vocal cords might
underlie laryngeal stridor in at least some individuals with
MSA.66
Impaired automatic ventilation reflects involve­
ment of the brainstem respiratory network, including
the pre-Bötzinger complex67
and the medullary raphe,68
which may predispose patients to sudden death.69
Lewy body disorders
Accumulation of Lewy neurites and Lewy bodies in
the ENS, autonomic ganglia, peripheral autonomic
nerve terminals, and intermediolateral cell column
can be detected at autopsy of cases of incidental Lewy
body disease (ILBD), PAF, early-stage PD, and DLB.70–73
α‑Synuclein inclusions can be also detected in cutan­eous
nerves of patients with PAF74
or PD.75
These findings
indicate that Lewy body disorders constitute a contin­
uum of progressive involvement of areas of the periph­
eral autonomic nervous system, and subsequently the
CNS, by α‑synuclein neuropathology, which may range
clinically from asymptomatic cases (ILBD) to pure auto­
nomic failure (PAF), and might eventually manifest with
­
parkinsonism (PD), dementia (DLB) or both.
Pure autonomic failure
PAF was first described by Bradbury and Eggleston in
1925, and was initially termed idiopathic orthostatic
hypotension. PAF is a rare, sporadic, adult-onset dis­
order characterized by symptomatic orthostatic hypo­
tension and variable gastrointestinal, bladder and sexual
dysfunction, in the absence of somatic motor deficits.
Onset is typically between 50 and 70 years of age.42
The symptoms of PAF are insidious in onset, and are
less progressive and disabling than those in other neuro­
degenerative autonomic disorders. Some patients may
have bladder symptoms, erectile or ejaculatory dysfunc­
tion, or impaired sweating, before developing ortho­
static hypotension. Constipation is common, but other
symptoms of gastrointestinal dysmotility are rare.76
The
diagnosis of PAF is always tentative; after a few years,
many patients with presumed PAF may develop cerebel­
lar, extrapyramidal or cognitive deficits indicating MSA,
PD or DLB. Thus, the diagnosis of PAF requires at least a
5 year history of isolated autonomic dysfunction without
other neurological manifestations. PAF is a clinical
syndrome reflecting involvement of the autonomic gan­
glion cells and their postganglionic axons. This disorder
can have a variety of different pathobiological substrates.
Patients with PAF have been shown to have accumula­
tion of α‑synuclein-containing Lewy bodies and neurites
in sympathetic ganglia peripheral tissues, including skin
sympathetic nerves.74,75
However, some cases of PAF are
associated with low-titre gnAChR antibodies, suggesting
that this condition can reflect a chronic form of AAG in
some instances.77
Parkinson disease
Autonomic dysfunction—particularly gastrointestinal
dysmotility—is a prominent nonmotor manifestation
of PD.43
Gastrointestinal dysmotility in PD can affect
any level of the gastrointestinal tract.78
Gastrointestinal
symptoms can occur at an early stage of the disease;79
for
example, constipation may precede the development of
motor symptoms by several years.80,81
Excessive drooling
in PD must reflect oropharyngeal dysphagia, as salivary
secretion is reduced even at an early stage of disease;82,83
this finding is consistent with the presence of α‑synuclein
pathology in the salivary gland.84
Oesophageal dysmotility
and delayed gastric emptying are frequent manifestations
in PD,85,86
and are likely to reflect early involvement of
the dorsal motor nucleus of the vagus.70
The prevalence
of constipation ranges from 20–89%,79
and is primarily
attributable to slow colonic transit, reflecting the early
involvement of the ENS.87
However, defecatory dysfunc­
tion due to paradoxical contraction of the ­
puborectalis
muscle is also an important contributory factor.85,88
Orthostatic hypotension is estimated to occur in
16–58% of patients with PD.89
In general, orthostatic
hypotension in PD is asymptomatic and tends to occur
at later stages of disease than in MSA, but in a subgroup
of patients it may be early and prominent, manifesting
even before the initiation of dopaminergic therapy.89
Urinary symptoms occur in 38–71% of patients with
PD.77,90,91
The most prominent symptom is nocturia, fol­
lowed by urgency and frequency, and the most common
urodynamic finding is detrusor overactivity.92
Nocturia is
the only urinary symptom that improves after deep brain
stimulation of the subthalamic nucleus.
Differential diagnosis
The differential diagnosis between MSA‑P and PD with
autonomic failure can be difficult. A prospective study
showed that autonomic indices, particularly the percent­
age of the body affected by anhidrosis, were significantly
and persistently more abnormal in MSA than in PD with
autonomic failure.61
Urodynamic findings such as large
post-micturitional residual volume (100ml), detrusor
external sphincter dyssynergia, and open bladder neck
at the start of filling are highly suggestive of MSA.54
Anal sphincter denervation due to loss of Onuf nucleus
motor neurons may help to distinguish MSA from PD
within the first few years of onset of motor symptoms,
but this finding is not specific for MSA, and can occur
in progressive supranuclear palsy or in PD patients with
chronic constipation.93
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol
Structural brain imaging, including diffusion-
weighted and diffusion tensor MRI, and functional
imaging, including PET and single-photon emis­
sion CT (SPECT), might aid the differential diagnosis
between MSA and PD.1
The presence of atrophy in the
putamen, middle cerebellar peduncle and pons on MRI
serve as supporting features for the diagnosis of pos­
sible MSA.39
Signal changes on T2-weighted images,
including the slit-like void signal (hypointensity of
the posterior putamen surrounded by hyperintense
lateral putaminal rim) and the hot cross bun sign (due
to myelin loss in the basis pontis) are more charac­
teristic of MSA but are not specific for this condition.
Diffusion-weighted imaging shows elevated apparent
diffusion coefficient in the putamen, pons and middle
cerebellar peduncle in MSA.94
In MSA‑P, but not in PD,
brain perfusion SPECT shows striatal hypoperfusion,95
and 18
F-fluorodeoxyglucose PET shows striatal hypo­
metabolism.96
By contrast, dopamine transporter SPECT
using 123
I-FP-CIT demonstrates reduced striatal binding
in both disorders.97
Myocardial scintigraphy using 123
I-metaiodo­benzyl­
guanidine (MIBG)98
or 6‑18
F-fluorodopamine PET99
assesses postganglionic sympathetic innervation of the
heart; these modalities are potential tools to differenti­
ate between MSA and Lewy body disorders. However,
although initial studies suggested that loss of cardiac
sympathetic innervation in PD or PAF could help in
distinguishing Lewy body disorders from MSA,99,100
decreased cardiac MIBG uptake has also been found in
up to 30% of patients with MSA,101
possibly reflecting
coexistent Lewy body pathology.
Dementia with Lewy bodies
Severe autonomic dysfunction, as well as repeated falls
and syncope, are features supporting the diagnosis of
DLB.102
Orthostatic hypotension, urinary frequency
and urge incontinence can be disabling symptoms in
DLB,103,104
and symptomatic orthostatic hypotension
may occur in 30–50% of cases. On the basis of auto­
nomic laboratory evaluation, the severity of autonomic
failure in DLB is intermediate between that seen in MSA
and PD.105
Other neurodegenerative disorders
Other central neurodegenerative disorders can manifest
with autonomic failure that reflects peripheral autonomic
involvement. These conditions, which may clinically
mimic MSA or Lewy body disorders, include fragile X
tremor–ataxia syndrome, in which bladder symptoms
might be prominent;106
adult-onset autosomal dominant
leukodystrophy, which is associated with pure sympa­
thetic failure;107
and a novel prion disorder character­
ized by diarrhoea and length-dependent, predominantly
sensory and autonomic axonal neuropathy, followed by
cognitive decline.108
Autoimmune autonomic disorders
Autoimmune autonomic disorders are characterized
by acute or subacute onset of generalized or restricted
autonomic failure, including gastrointestinal enteropathy.
Early recognition of these disorders is important because
they are disabling and might be the initial manifestation
of an underlying neoplasm. In addition, patients with
autoimmune autonomic disorders may show substantial
improvement with ­
immunomodulatory therapy.4,109
Autoimmune autonomic ganglionopathy
AAG (formerly known as acute pandysautonomia or
idiopathic subacute autonomic neuropathy18,110
) is
characterized by severe autonomic failure that develops
over the course of days or weeks in a previously healthy
person. The onset of symptoms may follow a viral infec­
tion, minor surgical procedure, or vaccination. The
most common manifestation is generalized sympathetic,
parasympathetic and ENS failure, but the spectrum is
broad; symptoms might be confined to pure choliner­
gic neuropathy manifested by sicca syndrome, isolated
adrenergic neuropathy, or isolated gastrointestinal dys­
motility. Patients may recover spontaneously, but only
one-third experience substantial functional ­improvement
without treatment.
Approximately 50% of cases of subacute AAG are
associ­
ated with gnAChR antibodies18
that block trans­
mission at the autonomic ganglia. Antibody titres
correlate with the severity of autonomic failure.19,111
gnAChR antibodies can also be associated with chronic
or restricted forms of autonomic dysfunction,77
and with
neurological or paraneoplastic disorders unrelated to
the autonomic nervous system.19
In one series, 30% of
gnAChR-seropositive patients had other paraneoplastic
antibodies in the setting of occult cancer, most com­
monly adenocarcinoma.19
In some cases, AAG coexists
with other autoimmune disorders, such as myasthenia
gravis, in patients with occult cancer.112,113
Several observational studies report that patients
with gnAChR-seropositive AAG benefit from intra­
venous immunoglobulin, plasma exchange, prednisone,
mycophenolate, azathioprine and rituximab, either
individually or in combination.19,114–116
A substantial
percentage of patients with AAG are seronegative for
gnAChR and other autoantibodies;117
some of these
patients may also show symptomatic improvement with
­immunomodulatory therapy.116
Paraneoplastic autonomic neuropathy
A type of subacute autonomic neuropathy that is clinically
indistinguishable from idiopathic AAG might occur as
a manifestation of an occult neoplasm, most commonly
small-cell lung carcinoma associated with anti-Hu
antibodies or, less frequently, thymoma or other neo­
plasms.118,119
Like AAG, paraneoplastic autonomic neuro­
pathy may manifest with generalized autonomic failure,
or with isolated enteric ganglionopathy leading to the
intestinal pseudo-obstruction syndrome.120,121
Acute autonomic and sensory neuropathy
Acute autonomic and sensory neuropathy is a rare dis­
order that differs from AAG in that profound autonomic
failure is associated with various degrees of sensory
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 7
impairment, including loss of pain and temperature
sensation, followed by sensory ataxia due to impair­
ment of proprioception.109,122,123
The mean age of onset
is in the late second decade, but can vary from child­
hood to the elderly. A triggering event, such as respira­
tory or gastrointestinal tract infections, is reported in
two-thirds of patients. Gastrointestinal dysmotility is
the most prominent autonomic symptom; the sensory
loss affects the proximal regions of the limbs, face, scalp
and trunk, and tends to be asymmetrical and segmen­
tal. Pain in the involved region is a common and serious
symptom. Other features of this disorder are reduction of
sensory nerve action potentials and increased T2 signal
in the posterior columns of the cervical spinal cord.123
Autopsy studies have shown severe neuronal cell loss in
the thoracic sympathetic and dorsal root ganglia, and
in Auerbach’s plexus.123
Sjögren syndrome
Primary Sjögren syndrome manifests with a wide variety
of peripheral neuropathies or ganglionopathies.124
Dorsal
root ganglionopathy may manifest with neuropathic pain
or ataxic neuropathy; autonomic neuropathy might be
prominent in some cases, but is the least frequent form
of neuropathy overall.124
Autoantibodies against muscarinic M3
receptors have
been identified in a subset of patients with Sjögren syn­
drome.125
These antibodies are particularly prevalent in
juvenile cases126
and have cholinergic blocking actions
in vitro.127
Some patients with Sjögren syndrome and
chronic progressive autonomic failure have gnAChR
antibodies, which may predict a positive response to
immunomodulatory therapy.128
Peripheral neuropathies
Peripheral neuropathies associated with autonomic
failure present with various phenotypes, including typical
distal symmetric sensorimotor neuropathy; dorsal root
ganglionopathy manifesting with neuropathic pain or
sensory ataxia; demyelinating, predominantly motor
polyradiculopathy; or distal painful neuropathy. The
spectrum of severity of autonomic failure is highly vari­
able in these disorders, ranging from being a major cause
of disability, as can occur in neuropathies associated
with diabetes or amyloidosis, to restricted distal vaso­
motor and sudomotor impairment in the lower limbs,
as observed in painful SFN.
Diabetic autonomic neuropathy
Diabetic neuropathies are heterogeneous: they include
typical diabetic sensorimotor polyneuropathy, painful
diabetic neuropathy, and diabetic autonomic neuro­
pathy.129
Estimates of the prevalence of diabetic auto­
nomic neuropathy vary depending of the source of
information (community, clinic or tertiary referral centre)
and the type of tests performed to assess autonomic func­
tion.129
Using strict criteria based on autonomic testing
abnormalities, the prevalence in one study was 16.8% for
patients with type 1 and 34.3% for patients with type 2
diabetes mellitus.130
Diabetic autonomic neuropathy can affect cardio­
vascular, gastrointestinal, urogenital and sudomotor
functions to various degrees. In most of the typical cases,
a combination of cardiovascular tests and sudomotor
tests allows its detection in otherwise asympto­
matic
patients. An impaired heart rate response to deep
breathing­
—an indicator of cardiovagal failure—has the
greatest specificity (~80%).131,132
Characteristic mani­
festations of diabetic cardiovascular autonomic neuro­
pathy include resting tachycardia, exercise intolerance,
orthostatic hypotension, intraoperative cardiovascular
instability, and silent myocardial infarction–silent ischae­
mia syndrome. Cardiovascular autonomic neuropathy
has been associated with increased morbidity and mor­
tality,133
so screening for this condition should be per­
formed at the time of diagnosis of type 2 diabetes and
5 years after the diagnosis of type 1 diabetes, particularly
in high-risk patients.129
Upper gastrointestinal dysmotility, which is present
in 40–50% of longstanding cases of diabetes, may
affect postprandial blood glucose levels and glycaemic
control. Constipation is the most frequent gastrointesti­
nal symptom in diabetes, and was reported by 60% of
patients in one study.134
Profuse, watery and predomi­
nantly nocturnal diarrhoea135
and faecal incontinence136
may be disabling in some patients. Bladder symptoms
occur in up to 50% of patients; 43–87% of type 1 and 25%
of type 2 diabetic patients have abnormal urodynamic
findings.137,138
Erectile dysfunction has a reported preva­
lence of 35–90% in patients with diabetes.139,140
Length-
dependent loss of thermoregulatory sweating is common,
and is occasionally associated with compensatory hyper­
hidrosis.22
Gustatory sweating can result from aberrant
innervation of sweat glands by parasympathetic fibres.141
Some patients with diabetes develop an acute painful
neuropathy associated with autonomic failure within a
short time of commencing tight glucose control.142
The
symptoms may be associated with abnormal autonomic
test results and reduced IENFD on punch skin biopsy,
and can all improve over time.142
Amyloid neuropathy
Amyloidoses manifesting with autonomic neuro­
pathy include the AL type (which may be associated
with myeloma or macroglobulinaemia), and familial
amyloid polyneuropathy (FAP), which is most com­
monly linked to mutations in the transthyretin (TTR)
gene. The types of amyloid neuropathy associated with
autonomic failure are highly heterogeneous.143,144
In a
retrospective study on 65 cases of AL amyloidosis or
FAP, generalized autonomic failure was associated with
painful (62%) or nonpainful (17%) sensorimotor poly­
neuropathy or distal SFN (5%); autonomic failure also
occurred without neuropathy (11%), but polyneuro­
pathy without generalized autonomic failure was infre­
quent (6%).143
In AL amyloidosis, peripheral neuropathy
may be the presenting feature or an incidental finding
of the disease, and can occur in up to 20% of cases.145
The median survival of patients with AL amyloidosis
who have peripheral neuropathy has improved with the
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol
Table 1 | General management of autonomic failure
Symptom Approach Comments
Orthostatic
hypotension
Patient education: recognize atypical symptoms of
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
The most important aspect of management
Sleep with the head elevated 4in (reverse
Trendelenburg position)
Avoids supine hypertension and nocturnal polyuria
Postural countermanouevres (for example, leg crossing,
squatting)
Increases orthostatic tolerance
Support garments (waist-high support stockings,
abdominal binder)
Reduces venous pooling
Volume expansion (daily intake of 8–10g sodium,
fluid intake of at least 2.0–2.5l)
Initial step before pharmacological management
Rapid ingestion of 500ml tap water Rapidly increases blood pressure independently
of volume expansion
Dietary changes; avoid alcohol; eat small, frequent
meals (carbohydrate is best ingested with the last
meal of the day)
Prevents splanchnic vasodilation
Florinef (0.1–0.4mg daily) Adverse effects include supine hypertension, ankle
oedema, hypokalaemia, hypomagnesaemia, headache,
and reduced effects of warfarin
Midodrine (10–40mg daily) Adverse effects include scalp tingling or itching,
goose bumps, supine hypertension, and urinary urgency
or retention
Pyridostigmine (60–240mg daily) Adverse effects include nausea, abdominal cramps,
diarrhoea, increased salivation, urinary urgency and
bradycardia
Droxidopa (100–600mg TID) Main potential adverse effect is supine hypertension
Erythropoietin (25–75U/kg SQ) Iron supplementation is usually required; supine
hypertension may occur
Desmopressin nasal spray (5–40μg) Risk of water intoxication and hyponatraemia
Octreotide (25–200μg SQ) Adverse effects include nausea and abdominal cramps,
hypertension, risk of gallstones and postprandial
hyperglycaemia
Neurogenic
bladder
Anticholinergics (for example, oxybutynin, tolterodine);
clean intermittent self-catheterization
Adverse effects include constipation, dry mouth, risk
of glaucoma, and worsening of cognitive function
Drooling Anticholinergics; botulinum toxin Anticholinergics may worsen confusion; botulinum toxin
may worsen dysphagia
Gastroparesis Metoclopramide (5–20mg QID); domperidone
(10–30mg QID); erythromycin (50–250mg QID);
pyridostigmine (30–60mg TID); bethanechol (25mg
QID); nutritional support (low-fat, low-fibre oral
supplementation; infusion of formula via a jejunal
feeding tube)
Gastroparesis may impair absorption of levodopa;
metoclopramide is contraindicated in parkinsonian
disorders; domperidone may cause QT prolongation,
and is not available in the USA
Constipation Fibre supplementation (15g per day); bulk agents
(for example, psyllium [1tsp up to three times daily],
methylcellulose); osmotic laxatives (for example, milk
of magnesia [two tablets up to four times daily],
polyethylene glycol [17g in 25ml once or twice daily],
lactulose [15–30ml TID]); docusate (stool softener,
100mg BID); lubiprostone (24μg BID); pyridostigmine
(up to 180–540mg); bisacodyl (10mg up to three times
a week); antibiotics to prevent bacterial overgrowth;
surgery for intractable colonic inertia
Donepezil and other cholinesterase inhibitors may
cause diarrhoea
Erectile
dysfunction
Oral phosphodiesterase type 5 inhibitors (for example,
sildenafil 25–100mg)
Adverse effects include headache, flushing, nasal
congestion, blue vision, and non-arteritic ischaemic
optic neuropathy; these drugs may worsen orthostatic
hypotension
Intracavernous drugs (for example, alprostadil,
papaverine)
Adverse effects include penile pain, oedema and
haematoma, palpable nodules or plaques, and priapism
Abbreviations: BID, twice a day; QID, four times a day; SQ, subcutaneous; TID, three times a day.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 9
combination of melphalan, prednisone and autologous
stem cell transplantation.146
The clinical and pathological features of FAP have
been reviewed in detail elsewhere.147,148
Over 100 muta­
tions in the TTR gene have been identified, the most
common (~50% of cases of FAP worldwide) of which
causes a Val30Met substitution. Different TTR variants
contribute to variable age at diagnosis, degree of cardiac
involvement, and survival.149
The first-line specific treat­
ment of choice for Val30Met TTR-FAP is liver transplan­
tation.150
In addition, tafamidis, a stabilizer of tetrameric
transthyretin, has shown short-term effectiveness in
slowing the progression of peripheral neuropathy at very
early stages of Val30Met TTR-FAP.151
Painful small fibre neuropathies
Selective involvement of postganglionic as well as noci­
cep­
tive fibres in painful SFN typically manifests with
distal anhidrosis (or sometimes hyperhidrosis) and
vasomotor dysfunction (generally erythema, but some­
times vasoconstriction and coldness) in the lower limbs.
In some cases, SFN can result in more-generalized
autonomic failure that may be symptomatic or detect­
able with autonomic testing. Important examples of
painful SFN are those associated with HIV infections152
and Fabry disease,153
in addition to the types related to
dia­
betes and amyloidosis, as highlighted above. Distal
sympathetic sudomotor and vasomotor failure in con­
junction with neuropathic pain also occur in sodium
channelopathies associated with gain-of-function
SCNA9 mutations, both in familial syndromes154
and in
a substantial proportion of idiopathic SFN cases.155
The
SCNA9 gene encodes the Nav
1.7 voltage-gated sodium
channel, which is expressed in nociceptive dorsal root
ganglion and sympathetic neurons. These gain-of-­
function mutations cause increased excitability of the
nociceptor and hypoexcitability of sympathetic ganglion
neurons,156,157
which might explain the coexistence of
neuropathic pain and postganglionic sympathetic failure.
Management issues
A detailed discussion of the management of autonomic
failure is beyond the scope of this Review, but the general
approach is summarized in this section and in Table 1.
The principles of management include discontinua­
tion of potentially causative drugs; immunomodula­
tory therapy for autoimmune disorders; management
of diabetes, amyloidosis or other potentially treatable
cause of autonomic neuropathy; patient education; non-­
pharmacological approaches; and pathophysiologically
based drug therapy. Patient education is a fundamen­
tal aspect of management. For example, patients with
orthostatic hypotension should be instructed to recog­
nize atypical symptoms and avoid precipitating factors.
Appropriate fluid and sodium intake, dietary adjust­
ments and regular exercise are beneficial in most cases
of autonomic failure.
Conclusions
The wide spectrum of clinical manifestations and dis­
orders associated with autonomic failure requires a
systematic clinical and laboratory approach to establish
the diagnosis, particularly in cases where autonomic
failure is disabling or life-threatening. A careful history
and examination is the mainstay of diagnosis, and lab­
oratory and other ancillary tests must be prioritized to
search for potentially treatable causes. Patient education,
avoidance of precipitating factors, non-pharmacological
approaches, and pathophysiologically based drug therapy
form the basis of current treatment of these disorders.
Further elucidation of the pathobiological mechanisms
that lead to immune, metabolic or degenerative damage
or dysfunction of central and peripheral autonomic
neurons will provide more-specific therapeutic targets.
Review criteria
A PubMed search from 1st
January 1975 to 15th
December
2013 was performed using the following terms (individually
and in various combinations): “autonomic failure”,
“autonomic neuropathy”, “orthostatic hypotension”,
“multiple system atrophy”, “Parkinson disease”, “Lewy
body disease”, “dementia with Lewy bodies”, “autoimmune
autonomic ganglionopathy”, “paraneoplastic”, “diabetic
neuropathy”, “amyloid neuropathy”, “Sjögren syndrome”,
“leukoencephalopathy”, “fragile X” and “prion”. The
search was limited to full articles published in English.
Other articles were identified from bibliographies of the
retrieved articles.
1. Stefanova, N., Bucke, P
., Duerr, S. 
Wenning, G. K. Multiple system atrophy: an
update. Lancet Neurol. 8, 1172–1178 (2009).
2. Cersosimo, M. G.  Benarroch, E. E. Autonomic
involvement in Parkinson’s disease: pathology,
pathophysiology, clinical features and possible
peripheral biomarkers. J. Neurol. Sci. 313,
57–63 (2012).
3. Vernino, S. Autoimmune and paraneoplastic
channelopathies. Neurotherapeutics 4, 305–314
(2007).
4. Muppidi, S.  Vernino, S. Autoimmune
autonomic failure. Handb. Clin. Neurol. 117,
321–327 (2013).
5. Freeman, R. Autonomic peripheral neuropathy.
Lancet 365, 1259–1270 (2005).
6. Freeman, R. et al. Consensus statement on the
definition of orthostatic hypotension, neurally
mediated syncope and the postural tachycardia
syndrome. Auton. Neurosci. 161, 46–48 (2011).
7. Mathias, C. J. et al. Differential blood pressure
and hormonal effects after glucose and xylose
ingestion in chronic autonomic failure. Clin. Sci.
(Lond.) 77, 85–92 (1989).
8. Mathias, C. J., Mallipeddi, R.  Bleasdale-
Barr, K. Symptoms associated with orthostatic
hypotension in pure autonomic failure and
multiple system atrophy. J. Neurol. 246,
893–898 (1999).
9. Freeman, R. Clinical practice. Neurogenic
orthostatic hypotension. N. Engl. J. Med. 358,
615–624 (2008).
10. Benarroch, E. E. The arterial baroreflex:
functional organization and involvement in
neurologic disease. Neurology 71, 1733–1738
(2008).
11. Cheshire, W. P.  Freeman, R. Disorders
of sweating. Semin. Neurol. 23, 399–406
(2003).
12. Fowler, C. J., Griffiths, D.  de Groat, W. C. The
neural control of micturition. Nat. Rev. Neurosci.
9, 453–466 (2008).
13. Holstege, G. Micturition and the soul. J. Comp.
Neurol. 493, 15–20 (2005).
14. Panicker, J. N.  Fowler, C. J. The bare
essentials: uro-neurology. Pract. Neurol. 10,
178–185 (2010).
15. Travagli, R. A., Hermann, G. E., Browning, K. N.
 Rogers, R. C. Brainstem circuits regulating
gastric function. Annu. Rev. Physiol. 68, 279–305
(2006).
16. Furness, J. B. The enteric nervous system and
neurogastroenterology. Nat. Rev. Gastroenterol.
Hepatol. 9, 286–294 (2012).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol
17. Kyle, R. A.  Bayrd, E. D. Amyloidosis: review of
236 cases. Medicine (Baltimore) 54, 271–299
(1975).
18. Vernino, S. et al. Autoantibodies to ganglionic
acetylcholine receptors in autoimmune
autonomic neuropathies. N. Engl. J. Med. 343,
847–855 (2000).
19. McKeon, A., Lennon, V. A., Lachance, D. H.,
Fealey, R. D.  Pittock, S. J. Ganglionic
acetylcholine receptor autoantibody: oncological,
neurological, and serological accompaniments.
Arch. Neurol. 66, 735–741 (2009).
20. Goldstein, D. S. Catecholamines 101. Clin.
Auton. Res. 20, 331–352 (2010).
21. Robertson, D. et al. Isolated failure of autonomic
noradrenergic neurotransmission. Evidence for
impaired β‑hydroxylation of dopamine. N. Engl.
J. Med. 314, 1494–1497 (1986).
22. Fealey, R. D., Low, P
. A.  Thomas, J. E.
Thermoregulatory sweating abnormalities in
diabetes mellitus. Mayo Clin. Proc. 64, 617–628
(1989).
23. Low, P
. A. Autonomic nervous system function.
J. Clin. Neurophysiol. 10, 14–27 (1993).
24. Gibbons, C. H., Illigens, B. M., Centi, J. 
Freeman, R. QDIRT: quantitative direct and
indirect test of sudomotor function. Neurology
70, 2299–2304 (2008).
25. Bennett, T. et al. Assessment of vagal control of
the heart in diabetes. Measures of R–R interval
variation under different conditions. Br. Heart J.
39, 25–28 (1977).
26. Booth, R. W.  Ryan, J. M. The clinical use of the
Valsalva maneuver. Heart Bull. 10, 111–113
(1961).
27. Sandroni, P
., Benarroch, E. E.  Low, P
. A.
Pharmacological dissection of components of
the Valsalva maneuver in adrenergic failure.
J. Appl. Physiol. 71, 1563–1567 (1991).
28. Cooke, J. et al. Sitting and standing blood
pressure measurements are not accurate for the
diagnosis of orthostatic hypotension. QJM 102,
335–339 (2009).
29. Low, P
. A. Testing the autonomic nervous
system. Semin. Neurol. 23, 407–421 (2003).
30. Freeman, R. Assessment of cardiovascular
autonomic function. Clin. Neurophysiol. 117,
716–730 (2006).
31. Hilz, M. J.  Dutsch, M. Quantitative studies of
autonomic function. Muscle Nerve 33, 6–20
(2006).
32. Lauria, G. et al. European Federation of
Neurological Societies/Peripheral Nerve Society
Guideline on the use of skin biopsy in the
diagnosis of small fiber neuropathy. Report of a
joint task force of the European Federation of
Neurological Societies and the Peripheral Nerve
Society. Eur. J. Neurol. 17, 903–912, e44–e49
(2010).
33. Donadio, V. et al. Peripheral autonomic
neuropathy: diagnostic contribution of skin
biopsy. J. Neuropathol. Exp. Neurol. 71,
1000–1008 (2012).
34. Gibbons, C. H., Illigens, B. M., Wang, N. 
Freeman, R. Quantification of sweat gland
innervation: a clinical-pathologic correlation.
Neurology 72, 1479–1486 (2009).
35. Nolano, M. et al. Quantification of pilomotor
nerves: a new tool to evaluate autonomic
involvement in diabetes. Neurology 75,
1089–1097 (2010).
36. Libbey, C. A., Skinner, M.  Cohen, A. S. Use of
abdominal fat tissue aspirate in the diagnosis
of systemic amyloidosis. Arch. Intern. Med. 143,
1549–1552 (1983).
37. Kyle, R. A.  Greipp, P
. R. Amyloidosis (AL).
Clinical and laboratory features in 229 cases.
Mayo Clin. Proc. 58, 665–683 (1983).
38. Klein, C. J. et al. Mass spectrometric-based
proteomic analysis of amyloid neuropathy type in
nerve tissue. Arch. Neurol. 68, 195–199 (2011).
39. Gilman, S. et al. Second consensus statement
on the diagnosis of multiple system atrophy.
Neurology 71, 670–676 (2008).
40. Wenning, G. K., Ben Shlomo, Y., Magalhaes, M.,
Daniel, S. E.  Quinn, N. P
. Clinical features and
natural history of multiple system atrophy. An
analysis of 100 cases. Brain 117, 835–845
(1994).
41. Kollensperger, M. et al. Presentation, diagnosis,
and management of multiple system atrophy in
Europe: final analysis of the European multiple
system atrophy registry. Mov. Disord. 25,
2604–2612 (2010).
42. Kaufmann, H.  Biaggioni, I. Autonomic failure in
neurodegenerative disorders. Semin. Neurol. 23,
351–363 (2003).
43. Chaudhuri, K. R.  Schapira, A. H. Non-motor
symptoms of Parkinson’s disease: dopaminergic
pathophysiology and treatment. Lancet Neurol.
8, 464–474 (2009).
44. Graham, J. G.  Oppenheimer, D. R. Orthostatic
hypotension and nicotine sensitivity in a case of
multiple system atrophy. J. Neurol. Neurosurg.
Psychiatry 32, 28–34 (1969).
45. Papp, M. I., Kahn, J. E.  Lantos, P
. L. Glial
cytoplasmic inclusions in the CNS of patients
with multiple system atrophy (striatonigral
degeneration, olivopontocerebellar atrophy
and Shy–Drager syndrome). J. Neurol. Sci. 94,
79–100 (1989).
46. Iodice, V. et al. Autopsy confirmed multiple
system atrophy cases: Mayo experience and
role of autonomic function tests. J. Neurol.
Neurosurg. Psychiatry 83, 453–459 (2012).
47. Multiple-System Atrophy Research Collaboration.
Mutations in COQ2 in familial and sporadic
multiple-system atrophy. N. Engl. J. Med. 369,
233–244 (2013).
48. Tada, M. et al. Early development of autonomic
dysfunction may predict poor prognosis in
patients with multiple system atrophy. Arch.
Neurol. 64, 256–260 (2007).
49. O’Sullivan, S. S. et al. Clinical outcomes of
progressive supranuclear palsy and multiple
system atrophy. Brain 131, 1362–1372 (2008).
50. Wenning, G. K. et al. The natural history of
multiple system atrophy: a prospective European
cohort study. Lancet Neurol. 12, 264–274 (2013).
51. Wenning, G. K. et al. Time course of symptomatic
orthostatic hypotension and urinary incontinence
in patients with postmortem confirmed
parkinsonian syndromes: a clinicopathological
study. J. Neurol. Neurosurg. Psychiatry 67,
620–623 (1999).
52. Oppenheimer, D. R. Lateral horn cells in
progressive autonomic failure. J. Neurol. Sci. 46,
393–404 (1980).
53. Benarroch, E. E., Smithson, I. L., Low, P
. A. 
Parisi, J. E. Depletion of catecholaminergic
neurons of the rostral ventrolateral medulla in
multiple systems atrophy with autonomic failure.
Ann. Neurol. 43, 156–163 (1998).
54. Winge, K.  Fowler, C. J. Bladder dysfunction in
Parkinsonism: mechanisms, prevalence,
symptoms, and management. Mov. Disord. 21,
737–745 (2006).
55. Benarroch, E. E.  Schmeichel, A. M. Depletion
of corticotrophin-releasing factor neurons in the
pontine micturition area in multiple system
atrophy. Ann. Neurol. 50, 640–645 (2001).
56. Kennedy, P
. G.  Duchen, L. W. A quantitative
study of intermediolateral column cells in motor
neuron disease and the Shy–Drager syndrome.
J. Neurol. Neurosurg. Psychiatry 48, 1103–1106
(1985).
57. Chalmers, D.  Swash, M. Selective
vulnerability of urinary Onuf motoneurons in
Shy–Drager syndrome. J. Neurol. 234, 259–260
(1987).
58. Kirchhof, K., Apostolidis, A. N., Mathias, C. J. 
Fowler, C. J. Erectile and urinary dysfunction may
be the presenting features in patients with
multiple system atrophy: a retrospective study.
Int. J. Impot. Res. 15, 293–298 (2003).
59. Sakakibara, R. et al. Colonic transit time,
sphincter EMG, and rectoanal videomanometry
in multiple system atrophy. Mov. Disord. 19,
924–929 (2004).
60. Sandroni, P
., Ahlskog, J. E., Fealey, R. D.
 Low, P
. A. Autonomic involvement in
extrapyramidal and cerebellar disorders.
Clin. Auton. Res. 1, 147–155 (1991).
61. Lipp, A. et al. Prospective differentiation of
multiple system atrophy from Parkinson disease,
with and without autonomic failure. Arch. Neurol.
66, 742–750 (2009).
62. Iranzo, A. Sleep and breathing in multiple system
atrophy. Curr.Treat. Options Neurol. 9, 347–353
(2007).
63. Ghorayeb, I., Bioulac, B.  Tison, F. Sleep
disorders in multiple system atrophy. J. Neural
Transm. 112, 1669–1675 (2005).
64. Silber, M. H.  Levine, S. Stridor and death
in multiple system atrophy. Mov. Disord. 15,
699–704 (2000).
65. Glass, G. A., Josephs, K. A.  Ahlskog, J. E.
Respiratory insufficiency as the primary
presenting symptom of multiple-system atrophy.
Arch. Neurol. 63, 978–981 (2006).
66. Vetrugno, R. et al. Sleep-related stridor due to
dystonic vocal cord motion and neurogenic
tachypnea/tachycardia in multiple system
atrophy. Mov. Disord. 22, 673–678 (2007).
67. Schwarzacher, S. W., Rub, U.  Deller, T.
Neuroanatomical characteristics of the human
pre-Bötzinger complex and its involvement in
neurodegenerative brainstem diseases. Brain
134, 24–35 (2011).
68. Benarroch, E. E., Schmeichel, A. M., Low, P
. A. 
Parisi, J. E. Depletion of putative chemosensitive
respiratory neurons in the ventral medullary
surface in multiple system atrophy. Brain 130,
469–475 (2007).
69. Tada, M. et al. Depletion of medullary
serotonergic neurons in patients with multiple
system atrophy who succumbed to sudden
death. Brain 132, 1810–1819 (2009).
70. Braak, H. et al. Stanley Fahn Lecture 2005:
the staging procedure for the inclusion body
pathology associated with sporadic Parkinson’s
disease reconsidered. Mov. Disord. 21,
2042–2051 (2006).
71. Dickson, D. W. et al. Evidence that incidental
Lewy body disease is pre-symptomatic
Parkinson’s disease. Acta Neuropathol. 115,
437–444 (2008).
72. Jellinger, K. A. A critical reappraisal of current
staging of Lewy-related pathology in human
brain. Acta Neuropathol. 116, 1–16 (2008).
73. Beach, T. G. et al. Multi-organ distribution of
phosphorylated alpha-synuclein histopathology
in subjects with Lewy body disorders. Acta
Neuropathol. 119, 689–702 (2010).
74. Shishido, T. et al. α‑Synuclein accumulation in
skin nerve fibers revealed by skin biopsy in pure
autonomic failure. Neurology 74, 608–610
(2010).
75. Wang, N., Gibbons, C. H., Lafo, J.  Freeman, R.
α‑Synuclein in cutaneous autonomic nerves.
Neurology 81, 1604–1610 (2013).
76. Polinsky, R. J. Clinical autonomic
neuropharmacology. Neurol. Clin. 8, 77–92
(1990).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 11
77. Klein, C. M. et al. The spectrum of autoimmune
autonomic neuropathies. Ann. Neurol. 53,
752–758 (2003).
78. Pfeiffer, R. F. Gastrointestinal dysfunction in
Parkinson’s disease. Parkinsonism Relat. Disord.
17, 10–15 (2011).
79. Siddiqui, M. F., Rast, S., Lynn, M. J., Auchus, A. P
.
 Pfeiffer, R. F. Autonomic dysfunction in
Parkinson’s disease: a comprehensive symptom
survey. Parkinsonism Relat. Disord. 8, 277–284
(2002).
80. Abbott, R. D. et al. Frequency of bowel
movements and the future risk of Parkinson’s
disease. Neurology 57, 456–462 (2001).
81. Savica, R. et al. Medical records documentation
of constipation preceding Parkinson disease:
a case–control study. Neurology 73, 1752–1758
(2009).
82. Proulx, M., de Courval, F. P
., Wiseman, M. A. 
Panisset, M. Salivary production in Parkinson’s
disease. Mov. Disord. 20, 204–207 (2005).
83. Cersosimo, M. G. et al. Hyposialorrhea as an
early manifestation of Parkinson disease.
Auton. Neurosci. 150, 150–151 (2009).
84. Del Tredici, K., Hawkes, C. H., Ghebremedhin, E.
 Braak, H. Lewy pathology in the submandibular
gland of individuals with incidental Lewy body
disease and sporadic Parkinson’s disease.
Acta Neuropathol. 119, 703–713 (2010).
85. Edwards, L. L., Quigley, E. M., Harned, R. K.,
Hofman, R.  Pfeiffer, R. F. Characterization
of swallowing and defecation in Parkinson’s
disease. Am. J. Gastroenterol. 89, 15–25
(1994).
86. Castell, J. A. et al. Manometric abnormalities
of the oesophagus in patients with Parkinson’s
disease. Neurogastroenterol. Motil. 13, 361–364
(2001).
87. Wakabayashi, K., Takahashi, H., Takeda, S.,
Ohama, E.  Ikuta, F. Parkinson’s disease:
the presence of Lewy bodies in Auerbach’s
and Meissner’s plexuses. Acta Neuropathol. 76,
217–221 (1988).
88. Sakakibara, R., Uchiyama, T., Yamanishi, T.,
Shirai, K.  Hattori, T. Bladder and bowel
dysfunction in Parkinson’s disease. J. Neural
Transm. 115, 443–460 (2008).
89. Sharabi, Y.  Goldstein, D. S. Mechanisms
of orthostatic hypotension and supine
hypertension in Parkinson disease. J. Neurol.
Sci. 310, 123–128 (2011).
90. Winge, K., Skau, A. M., Stimpel, H.,
Nielsen, K. K.  Werdelin, L. Prevalence of
bladder dysfunction in Parkinsons disease.
Neurourol. Urodyn. 25, 116–122 (2006).
91. Blackett, H., Walker, R.  Wood, B. Urinary
dysfunction in Parkinson’s disease: a review.
Parkinsonism Relat. Disord. 15, 81–87 (2009).
92. Winge, K.  Nielsen, K. K. Bladder dysfunction
in advanced Parkinson’s disease. Neurourol.
Urodyn. 31, 1279–1283 (2012).
93. Sakakibara, R., Uchiyama, T., Yamanishi, T. 
Kishi, M. Sphincter EMG as a diagnostic tool in
autonomic disorders. Clin.Auton. Res. 19, 20–31
(2009).
94. Seppi, K. et al. Progression of putaminal
degeneration in multiple system atrophy: a serial
diffusion MR study. Neuroimage 31, 240–245
(2006).
95. Feigin, A. et al. Tc‑99m ethylene cysteinate
dimer SPECT in the differential diagnosis of
parkinsonism. Mov. Disord. 17, 1265–1270
(2002).
96. Kwon, K. Y., Choi, C. G., Kim, J. S., Lee, M. C.
 Chung, S. J. Comparison of brain MRI and
18
F-FDG PET in the differential diagnosis of
multiple system atrophy from Parkinson’s
disease. Mov. Disord. 22, 2352–2358 (2007).
97. Varrone, A., Marek, K. L., Jennings, D.,
Innis, R. B.  Seibyl, J. P
. [123
I]β-CIT SPECT
imaging demonstrates reduced density of
striatal dopamine transporters in Parkinson’s
disease and multiple system atrophy. Mov.
Disord. 16, 1023–1032 (2001).
98. King, A. E., Mintz, J.  Royall, D. R. Meta-analysis
of 123
I-MIBG cardiac scintigraphy for the
diagnosis of Lewy body-related disorders.
Mov. Disord. 26, 1218–1224 (2011).
99. Goldstein, D. S., Holmes, C., Cannon, R. O. 3rd
,
Eisenhofer, G.  Kopin, I. J. Sympathetic
cardioneuropathy in dysautonomias. N. Engl.
J. Med. 336, 696–702 (1997).
100. Treglia, G. et al. Diagnostic performance
of iodine‑123‑metaiodobenzylguanidine
scintigraphy in differential diagnosis between
Parkinson’s disease and multiple-system atrophy:
a systematic review and a meta-analysis. Clin.
Neurol. Neurosurg. 113, 823–829 (2011).
101.Nagayama, H. et al. Abnormal cardiac
[123
I]‑meta‑iodobenzylguanidine uptake in
multiple system atrophy. Mov. Disord. 25,
1744–1747 (2010).
102.McKeith, I. G. Consensus guidelines for the
clinical and pathologic diagnosis of dementia
with Lewy bodies (DLB): report of the
Consortium on DLB International Workshop.
J. Alzheimers. Dis. 9, 417–423 (2006).
103.Stubendorff, K., Aarsland, D., Minthon, L. 
Londos, E. The impact of autonomic dysfunction
on survival in patients with dementia with Lewy
bodies and Parkinson’s disease with dementia.
PLoS ONE 7, e45451 (2012).
104.Kanazawa, M. et al. An autopsy case of
dementia with Lewy bodies showing autonomic
failure and dementia as the initial symptoms.
Mov. Disord. 22, 1212–1213 (2007).
105.Thaisetthawatkul, P
. et al. Autonomic dysfunction
in dementia with Lewy bodies. Neurology 62,
1804–1809 (2004).
106.Leehey, M. A. Fragile X‑associated tremor/ataxia
syndrome: clinical phenotype, diagnosis, and
treatment. J. Investig. Med. 57, 830–836 (2009).
107.Guaraldi, P
. et al. Isolated noradrenergic failure in
adult-onset autosomal dominant leukodystrophy.
Auton. Neurosci. 159, 123–126 (2011).
108.Mead, S. et al. A novel prion disease associated
with diarrhea and autonomic neuropathy. N. Engl.
J. Med. 369, 1904–1914 (2013).
109.Koike, H., Watanabe, H.  Sobue, G. The
spectrum of immune-mediated autonomic
neuropathies: insights from the
clinicopathological features. J. Neurol.
Neurosurg. Psychiatry 84, 98–106 (2013).
110.Suarez, G. A., Fealey, R. D., Camilleri, M. 
Low, P
. A. Idiopathic autonomic neuropathy:
clinical, neurophysiologic, and follow-up studies
on 27 patients. Neurology 44, 1675–1682
(1994).
111.Gibbons, C. H.  Freeman, R. Antibody titers
predict clinical features of autoimmune
autonomic ganglionopathy. Auton. Neurosci. 146,
8–12 (2009).
112.Vernino, S., Cheshire, W. P
.  Lennon, V. A.
Myasthenia gravis with autoimmune autonomic
neuropathy. Auton. Neurosci. 88, 187–192
(2001).
113.Peltier, A. C. et al. Coexistent autoimmune
autonomic ganglionopathy and myasthenia
gravis associated with non‑small‑cell lung
cancer. Muscle Nerve 41, 416–419 (2010).
114.Gibbons, C. H., Vernino, S. A.  Freeman, R.
Combined immunomodulatory therapy in
autoimmune autonomic ganglionopathy.
Arch. Neurol. 65, 213–217 (2008).
115.Imrich, R., Vernino, S., Eldadah, B. A., Holmes, C.
 Goldstein, D. S. Autoimmune autonomic
ganglionopathy: treatment by plasma exchanges
and rituximab. Clin.Auton. Res. 19, 259–262
(2009).
116.Iodice, V. et al. Efficacy of immunotherapy
in seropositive and seronegative putative
autoimmune autonomic ganglionopathy.
Neurology 72, 2002–2008 (2009).
117.Sandroni, P
. et al. Idiopathic autonomic
neuropathy: comparison of cases seropositive
and seronegative for ganglionic acetylcholine
receptor antibody. Arch. Neurol. 61, 44–48
(2004).
118.Lucchinetti, C. F., Kimmel, D. W.  Lennon, V. A.
Paraneoplastic and oncologic profiles of patients
seropositive for type 1 antineuronal nuclear
autoantibodies. Neurology 50, 652–657 (1998).
119.Winkler, A. S., Dean, A., Hu, M., Gregson, N. 
Chaudhuri, K. R. Phenotypic and neuropathologic
heterogeneity of anti-Hu antibody-related
paraneoplastic syndrome presenting with
progressive dysautonomia: report of two cases.
Clin.Auton. Res. 11, 115–118 (2001).
120.Chinn, J. S.  Schuffler, M. D. Paraneoplastic
visceral neuropathy as a cause of severe
gastrointestinal motor dysfunction.
Gastroenterology 95, 1279–1286 (1988).
121.De Giorgio, R. et al. Inflammatory neuropathies
of the enteric nervous system. Gastroenterology
126, 1872–1883 (2004).
122.Colan, R. V., Snead, O. C. 3rd
, Oh, S. J. 
Kashlan, M. B. Acute autonomic and sensory
neuropathy. Ann. Neurol. 8, 441–444 (1980).
123.Koike, H. et al. Clinicopathological features of
acute autonomic and sensory neuropathy. Brain
133, 2881–2896 (2010).
124.Pavlakis, P
. P
. et al. Peripheral neuropathies in
Sjögren syndrome: a new reappraisal. J. Neurol.
Neurosurg. Psychiatry 82, 798–802 (2011).
125.Dawson, L., Tobin, A., Smith, P
.  Gordon, T.
Antimuscarinic antibodies in Sjögren’s
syndrome: where are we, and where are we
going? Arthritis Rheum. 52, 2984–2995 (2005).
126.Nakamura, Y. et al. High prevalence of
autoantibodies to muscarinic‑3 acetylcholine
receptor in patients with juvenile-onset Sjögren
syndrome. Ann. Rheum. Dis. 67, 136–137
(2008).
127.Park, K., Park, S.  Jackson, M. W. The inhibitory
effects of antimuscarinic autoantibodies in the
sera of primary Sjogren syndrome patients on
the gastrointestinal motility. Mol. Immunol. 56,
583–587 (2013).
128.Kondo, T. et al. Autoimmune autonomic
ganglionopathy with Sjögren’s syndrome:
significance of ganglionic acetylcholine receptor
antibody and therapeutic approach. Auton.
Neurosci. 146, 33–35 (2009).
129.Tesfaye, S. et al. Diabetic neuropathies: update
on definitions, diagnostic criteria, estimation of
severity, and treatments. Diabetes Care 33,
2285–2293 (2010).
130.Ziegler, D., Gries, F. A., Spuler, M.  Lessmann, F.
The epidemiology of diabetic neuropathy.
Diabetic Cardiovascular Autonomic Neuropathy
Multicenter Study Group. J. Diabetes
Complications 6, 49–57 (1992).
131.Low, P
. A. et al. Autonomic symptoms and
diabetic neuropathy: a population-based study.
Diabetes Care 27, 2942–2947 (2004).
132.England, J. D. et al. Practice Parameter:
evaluation of distal symmetric polyneuropathy:
role of autonomic testing, nerve biopsy, and skin
biopsy (an evidence-based review). Report of the
American Academy of Neurology, American
Association of Neuromuscular and
Electrodiagnostic Medicine, and American
Academy of Physical Medicine and Rehabilitation.
Neurology 72, 177–184 (2009).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol
133.Vinik, A. I.  Ziegler, D. Diabetic cardiovascular
autonomic neuropathy. Circulation 115,
387–397 (2007).
134.Maleki, D. et al. Gastrointestinal tract symptoms
among persons with diabetes mellitus in the
community. Arch. Intern. Med. 160, 2808–2816
(2000).
135.Lysy, J., Israeli, E.  Goldin, E. The prevalence
of chronic diarrhea among diabetic patients.
Am. J. Gastroenterol. 94, 2165–2170 (1999).
136.Schiller, L. R. et al. Pathogenesis of fecal
incontinence in diabetes mellitus: evidence
for internal‑anal‑sphincter dysfunction. N. Engl.
J. Med. 307, 1666–1671 (1982).
137.Bradley, W. E. Diagnosis of urinary bladder
dysfunction in diabetes mellitus. Ann. Intern.
Med. 92, 323–326 (1980).
138.Kaplan, S. A., Te, A. E.  Blaivas, J. G.
Urodynamic findings in patients with diabetic
cystopathy. J. Urol. 153, 342–344 (1995).
139.Bacon, C. G. et al. Association of type and
duration of diabetes with erectile dysfunction
in a large cohort of men. Diabetes Care 25,
1458–1463 (2002).
140.Malavige, L. S.  Levy, J. C. Erectile dysfunction
in diabetes mellitus. J. Sex. Med. 6, 1232–1247
(2009).
141. Watkins, P
. J. Facial sweating after food: a new
sign of diabetic autonomic neuropathy. Br. Med. J.
1, 583–587 (1973).
142.Gibbons, C. H.  Freeman, R. Treatment-
induced diabetic neuropathy: a reversible
painful autonomic neuropathy. Ann. Neurol. 67,
534–541 (2010).
143.Wang, A. K., Fealey, R. D., Gehrking, T. L. 
Low, P
. A. Patterns of neuropathy and autonomic
failure in patients with amyloidosis. Mayo Clin.
Proc. 83, 1226–1230 (2008).
144.Cappellari, M. et al. Variable presentations of
TTR-related familial amyloid polyneuropathy in
seventeen patients. J. Peripher. Nerv. Syst. 16,
119–129 (2011).
145.Rajkumar, S. V., Gertz, M. A.  Kyle, R. A.
Prognosis of patients with primary systemic
amyloidosis who present with dominant
neuropathy. Am. J. Med. 104, 232–237 (1998).
146.Gertz, M. A. et al. Stem cell transplantation
for the management of primary systemic
amyloidosis. Am. J. Med. 113, 549–555
(2002).
147.Planté-Bordeneuve, V.  Said, G. Familial amyloid
polyneuropathy. Lancet Neurol. 10, 1086–1097
(2011).
148.Said, G.  Planté-Bordeneuve, V. TTR-familial
amyloid polyneuropathy—neurological aspects.
Amyloid 19 (Suppl. 1), 25–27 (2012).
149.Arruda-Olson, A. M. et al. Genotype,
echocardiography, and survival in familial
transthyretin amyloidosis. Amyloid 20, 263–268
(2013).
150.Yamashita, T. et al. Long-term survival after liver
transplantation in patients with familial amyloid
polyneuropathy. Neurology 78, 637–643 (2012).
151.Coelho, T. et al. Tafamidis for transthyretin
familial amyloid polyneuropathy: a randomized,
controlled trial. Neurology 79, 785–792 (2012).
152.Robinson-Papp, J., Sharma, S., Simpson, D. M.
 Morgello, S. Autonomic dysfunction is
common in HIV and associated with distal
symmetric polyneuropathy. J. Neurovirol. 19,
172–180 (2013).
153.Dutsch, M. et al. Small fiber dysfunction
predominates in Fabry neuropathy. J. Clin.
Neurophysiol. 19, 575–586 (2002).
154.Dib-Hajj, S. D., Cummins, T. R., Black, J. A. 
Waxman, S. G. From genes to pain: Nav
1.7 and
human pain disorders. Trends Neurosci. 30,
555–563 (2007).
155.Faber, C. G. et al. Gain of function Nav
1.7
mutations in idiopathic small fiber neuropathy.
Ann. Neurol. 71, 26–39 (2012).
156.Rush, A. M. et al. A single sodium channel
mutation produces hyper- or hypoexcitability in
different types of neurons. Proc. Natl Acad. Sci.
USA 103, 8245–8250 (2006).
157.Han, C. et al. Functional profiles of SCN9A
variants in dorsal root ganglion neurons and
superior cervical ganglion neurons correlate with
autonomic symptoms in small fibre neuropathy.
Brain 135, 2613–2628 (2012).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved

More Related Content

Similar to 2014 nature.pdf

Metabolic disoders internal medicine and neuroscience
Metabolic disoders internal medicine and neuroscienceMetabolic disoders internal medicine and neuroscience
Metabolic disoders internal medicine and neuroscienceNeilVincentDeAsis
 
seizure and vertigo.pptx
seizure and vertigo.pptxseizure and vertigo.pptx
seizure and vertigo.pptxBIMALESHYADAV2
 
Cns neuropathy davidson010.
Cns neuropathy  davidson010.Cns neuropathy  davidson010.
Cns neuropathy davidson010.Shaikhani.
 
Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)
Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)
Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)College of Medicine, Sulaymaniyah
 
Autonomic dysfunction.ppt
Autonomic dysfunction.pptAutonomic dysfunction.ppt
Autonomic dysfunction.pptShama
 
Neurological manifestation of hepatic diseases
Neurological manifestation of hepatic diseasesNeurological manifestation of hepatic diseases
Neurological manifestation of hepatic diseasesNeurologyKota
 
FLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdf
FLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdfFLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdf
FLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdfkeerti Gour (PT) Shakya
 
Epilepsy.ppt
Epilepsy.pptEpilepsy.ppt
Epilepsy.pptShama
 
Epilepsy.ppt
Epilepsy.pptEpilepsy.ppt
Epilepsy.pptShama
 
Approach to neuroregression
Approach to neuroregressionApproach to neuroregression
Approach to neuroregressiondrswarupa
 
neurosyphilisanditsphysiotherapymanagement-210903194918.pdf
neurosyphilisanditsphysiotherapymanagement-210903194918.pdfneurosyphilisanditsphysiotherapymanagement-210903194918.pdf
neurosyphilisanditsphysiotherapymanagement-210903194918.pdfRahulJankar4
 
Neurosyphilis and its physiotherapy management
Neurosyphilis and its physiotherapy managementNeurosyphilis and its physiotherapy management
Neurosyphilis and its physiotherapy managementMuskan Rastogi
 
Septicemic encephalopathy
Septicemic encephalopathySepticemic encephalopathy
Septicemic encephalopathyNeurologyKota
 
Adverse effects antipsychotics dr ali
Adverse effects antipsychotics dr aliAdverse effects antipsychotics dr ali
Adverse effects antipsychotics dr aliOSMAN ALI MD
 
MANAGEMENT OF EPILEPSY MODIFIED 2.pptx
MANAGEMENT OF EPILEPSY MODIFIED 2.pptxMANAGEMENT OF EPILEPSY MODIFIED 2.pptx
MANAGEMENT OF EPILEPSY MODIFIED 2.pptxJENNIFERENEKWECHI
 
Approach to peripheral neuropathy
Approach to peripheral neuropathyApproach to peripheral neuropathy
Approach to peripheral neuropathyNeurologyKota
 
7ataxiaaaaa.pptx
7ataxiaaaaa.pptx7ataxiaaaaa.pptx
7ataxiaaaaa.pptxSamanSarKo2
 

Similar to 2014 nature.pdf (20)

Metabolic disoders internal medicine and neuroscience
Metabolic disoders internal medicine and neuroscienceMetabolic disoders internal medicine and neuroscience
Metabolic disoders internal medicine and neuroscience
 
seizure and vertigo.pptx
seizure and vertigo.pptxseizure and vertigo.pptx
seizure and vertigo.pptx
 
Cns neuropathy davidson010.
Cns neuropathy  davidson010.Cns neuropathy  davidson010.
Cns neuropathy davidson010.
 
Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)
Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)
Medicine 5th year, 1st & 2nd lectures (Dr. Mohammad Shaikhani)
 
Autonomic dysfunction.ppt
Autonomic dysfunction.pptAutonomic dysfunction.ppt
Autonomic dysfunction.ppt
 
Medicina felina convulsions
Medicina felina   convulsionsMedicina felina   convulsions
Medicina felina convulsions
 
Neurological manifestation of hepatic diseases
Neurological manifestation of hepatic diseasesNeurological manifestation of hepatic diseases
Neurological manifestation of hepatic diseases
 
FLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdf
FLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdfFLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdf
FLOPPY INFANT SYNDROM,CHENNELOPATHY, CRAMP.pdf
 
Epilepsy.ppt
Epilepsy.pptEpilepsy.ppt
Epilepsy.ppt
 
Epilepsy.ppt
Epilepsy.pptEpilepsy.ppt
Epilepsy.ppt
 
Encephalitis
EncephalitisEncephalitis
Encephalitis
 
Approach to neuroregression
Approach to neuroregressionApproach to neuroregression
Approach to neuroregression
 
neurosyphilisanditsphysiotherapymanagement-210903194918.pdf
neurosyphilisanditsphysiotherapymanagement-210903194918.pdfneurosyphilisanditsphysiotherapymanagement-210903194918.pdf
neurosyphilisanditsphysiotherapymanagement-210903194918.pdf
 
Neurosyphilis and its physiotherapy management
Neurosyphilis and its physiotherapy managementNeurosyphilis and its physiotherapy management
Neurosyphilis and its physiotherapy management
 
Septicemic encephalopathy
Septicemic encephalopathySepticemic encephalopathy
Septicemic encephalopathy
 
Adverse effects antipsychotics dr ali
Adverse effects antipsychotics dr aliAdverse effects antipsychotics dr ali
Adverse effects antipsychotics dr ali
 
MANAGEMENT OF EPILEPSY MODIFIED 2.pptx
MANAGEMENT OF EPILEPSY MODIFIED 2.pptxMANAGEMENT OF EPILEPSY MODIFIED 2.pptx
MANAGEMENT OF EPILEPSY MODIFIED 2.pptx
 
Approach to peripheral neuropathy
Approach to peripheral neuropathyApproach to peripheral neuropathy
Approach to peripheral neuropathy
 
Psychiatric aspect of organic illness
Psychiatric aspect of organic illnessPsychiatric aspect of organic illness
Psychiatric aspect of organic illness
 
7ataxiaaaaa.pptx
7ataxiaaaaa.pptx7ataxiaaaaa.pptx
7ataxiaaaaa.pptx
 

Recently uploaded

A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformChameera Dedduwage
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdfssuser54595a
 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...EduSkills OECD
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Sapana Sha
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfsanyamsingh5019
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxmanuelaromero2013
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introductionMaksud Ahmed
 
Separation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesSeparation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesFatimaKhan178732
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
URLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppURLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppCeline George
 
Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3JemimahLaneBuaron
 
Arihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfArihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfchloefrazer622
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...Marc Dusseiller Dusjagr
 
Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...
Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...
Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...RKavithamani
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxSayali Powar
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxGaneshChakor2
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdfQucHHunhnh
 

Recently uploaded (20)

A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy Reform
 
Staff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSDStaff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSD
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
 
Mattingly "AI & Prompt Design: The Basics of Prompt Design"
Mattingly "AI & Prompt Design: The Basics of Prompt Design"Mattingly "AI & Prompt Design: The Basics of Prompt Design"
Mattingly "AI & Prompt Design: The Basics of Prompt Design"
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdf
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptx
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introduction
 
Separation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesSeparation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and Actinides
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
URLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppURLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website App
 
Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3
 
Arihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfArihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdf
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
 
Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...
Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...
Privatization and Disinvestment - Meaning, Objectives, Advantages and Disadva...
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptx
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 

2014 nature.pdf

  • 1. NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 1 Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA. benarroch.eduardo@ mayo.edu 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 Autonomic disorders manifest with autonomic failure or hyperactivity, which may be generalized or focal. Autonomic failure can affect the sympathetic, para­sympa­ thetic or enteric nervous systems, either in isolation or in various combinations, and can result from lesions at any level of the CNS or PNS. Sympathetic failure manifests primarily with orthostatic hypotension and anhidrosis (absence of sweating), cranial parasympathetic failure with intolerance to light, dry eyes (xerophthalmia) and dry mouth (xerostomia), sacral parasympathetic failure with urinary retention and erectile dysfunction, and enteric nervous system (ENS) failure with gastroparesis and constipation. In some cases, postural tachycardia syndrome is a manifestation of an underlying autonomic neuropathy. Many symptoms attributed to ‘dysautonomia’ in otherwise healthy young patients, such as gastroparesis or urinary retention, are rarely associated with objective evidence of autonomic failure. This Review will focus on the clinical approach to patients with generalized autonomic failure. The degen­ erative synucleinopathies, autonomic ganglionopathies and autonomic neuropathies have been reviewed in detail elsewhere.1–5 Rather than providing an exhaustive discussion of the wide spectrum of disorders associated with autonomic failure, this article will focus on specific examples to emphasize clinical cues and outline recent concepts of the underlying pathobiology. Clinical approach to autonomic failure Disorders associated with autonomic failure can be clas­ sified according to the type and severity of autonomic manifestations, associated neurological symptoms, and temporal profile (Box 1). The temporal profile of onset and progression has important implications for diag­ nosis, and for guidance of further laboratory evalua­ tion (Figure 1). Isolated autonomic failure of acute or subacute onset suggests an immune cause such as auto­ immune autonomic ganglionopathy (AAG), including paraneoplastic autonomic neuropathy, or a toxic cause such as effects of medications. Pure autonomic failure (PAF) refers to the slow development of general­ ized auto­ nomic failure in the absence of motor or sensory symp­ toms. Manifestations associated with chronic and pro­ gressive generalized autonomic failure, such as ataxia or parkinsonism, suggest a degenerative cause, typi­ cally a synuclein­ opathy such as multiple system atrophy (MSA), or a Lewy body disorder such as Parkinson disease (PD) or dementia with Lewy bodies (DLB). Auto­ nomic failure can also be a salient feature of various types of peripheral neuropathy, including distal sensori­ motor peripheral neuropathy, painful or ataxic forms of sensory ganglionopathy, distal painful small fibre ­ neuropathy (SFN), and acute motor polyradiculopathy. Manifestations and pathophysiology Orthostatic hypotension Orthostatic hypotension is defined as a sustained reduc­ tion in systolic blood pressure of at least 20 mmHg or diastolic blood pressure of 10 mmHg within 3 min of Competing interests The author declares no competing interests. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 2. 2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol standing or head-up tilt.6 Some patients with neuro­ genic orthostatic hypotension have postprandial hypo­ tension.7 Manifestations of orthostatic hypotension include lightheadedness, blurred vision and neck pain (coat hanger distribution) among others; the symptoms are worse in the morning, after meals, or on exposure to heat.8,9 Neurogenic orthostatic hypotension is the mani­ festation of impaired sympathetically mediated vaso­ constriction of skeletal muscle and mesenteric vessels in response to baroreceptor unloading due to orthostatic stress. Neurogenic orthostatic hypotension may occur as a consequence of disorders affecting the barosensi­ tive sympathoexcitatory neurons in the rostral ventro­ lateral medulla (for example, MSA); spinal preganglionic sympathetic neurons (for example, MSA or PD); auto­ nomic ganglia (for example, AAG or PAF); unmyelin­ ated axons (SFN); noradrenaline availability (for example, dopamine β‑hydroxylase deficiency); or vas­ cular α1-adrenergic receptors (typically as a side effect of drugs).10 Anhidrosis Sweating is an important thermoregulatory activity mediated by the sympathetic nervous system through cholinergic activation of muscarinic M3 receptors in the eccrine sweat glands. Depending on its distribu­ tion and severity, anhidrosis might be asymptomatic or might manifest with compensatory hyperhidrosis (in unaffected areas) or heat intolerance. Anhidrosis in autonomic failure may reflect impairment of central thermoregulatory pathways, spinal preganglionic sudomotor units, cholinergic sympathetic ganglion neurons, peripheral unmyelinated axons, or M3 ­receptors at the sudomotor unit.11 Neurogenic bladder and sexual dysfunction Normal bladder function includes a urine storage phase and a micturition phase, which are controlled at all levels of the neuraxis.12 Urine storage depends on spinal reflexes mediated by the lumbosacral sympathetic noradrenergic and sacral cholinergic motor neurons of the Onuf nucleus. Normal micturition depends on a Key points ■ ■ Autonomic failure can occur in isolation, in association with peripheral neuropathy, or as a manifestation of a neurodegenerative disorder ■ ■ Disorders associated with autonomic failure can be classified according to the type and severity of autonomic manifestations, associated neurological symptoms, and temporal profile ■ ■ Acute or subacute autonomic failure suggests an autoimmune autonomic ganglionopathy, which may be paraneoplastic ■ ■ In patients with parkinsonism and autonomic failure, early onset and progression of orthostatic hypotension or urogenital dysfunction, urinary incontinence, generalized anhidrosis, and/or laryngeal stridor are highly suggestive of multiple system atrophy ■ ■ Autonomic failure can occur with any type of diabetic or amyloid neuropathy ■ ■ Important causes of painful neuropathy associated with autonomic failure include Sjögren syndrome, HIV infections, Fabry disease, and sodium channelopathies supraspinal reflex coordinated by the pontine mictur­ ition centre, which activates the sacral pre­ gangli­ onic para­ sympa­ thetic output to the bladder detrusor while ­ simultaneously inhibiting the Onuf nucleus.13 Neurogenic bladder can manifest with detrusor over­ activity or underactivity.14 Detrusor overactivity pro­ duces urinary urgency with or without incontinence, urinary frequency, and nocturia. Reduced detrusor activity leads to incomplete bladder emptying, increased post-void residual, low peak urinary flow rate and, even­ tually, urinary retention and overflow incontinence. Impaired micturition can result from lesions affecting the bladder afferents, sacral parasympathetic neurons or their axons, or cholinergic muscarinic neurotransmis­ sion. Neurogenic bladder is commonly associated with erectile and ejaculatory dysfunction in men and poor vaginal lubrication in women. Erectile dysfunction reflects impaired sacral parasympathetic output and nitric oxide release in the erectile tissue. Box 1 | Neurological causes of autonomic failure A. Isolated autonomic failure 1. Acute or subacute (a) Autoimmune autonomic ganglionopathy (b) Paraneoplastic autonomic neuropathy 2. Progressive (a) Pure autonomic failure B. Progressive autonomic failure associated with parkinsonism, ataxia or dementia 1. Multiple system atrophy 2. Lewy body disorders (a) Parkinson disease (b) Dementia with Lewy bodies 3. Others (a) Familial leukoencephalopathies (b) Prion disorders C. Autonomic failure associated with peripheral neuropathy 1. Chronic sensorimotor neuropathies (a) Diabetes (b) Amyloidosis (c) Other metabolic disorders (vitamin B12 deficiency, uraemia) (d) Toxic neuropathies 2. Sensory ganglionopathies (a) Sjögren syndrome (b) Paraneoplastic 3. Distal painful neuropathies (a) Diabetes (b) Amyloidosis (c) Idiopathic (sodium channelopathies) (d) Infectious (HIV) (e) Hereditary (i) Hereditary sensory and autonomic neuropathy (ii) Fabry disease (iii) Sodium channelopathies 4. Acute or subacute motor polyradiculopathy or neuropathy (a) Guillain–Barré syndrome (b) Porphyria 5. Acute autonomic and sensory neuropathy 6. Ross syndrome (segmental anhidrosis, Adie pupils and areflexia). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 3. NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 3 Gastrointestinal dysmotility Control of gastrointestinal motility depends on intrin­ sic reflexes mediated by the ENS, and their modulation by vagal and paravertebral sympathetic inputs. Upper gastro­ intestinal motility is primarily controlled by the vagal reflexes involving the nucleus of the solitary tract and dorsal motor nucleus of the vagus in the medulla;15 peristalsis in the lower gastrointestinal tract depends primarily on local ENS reflexes;16 and paravertebral ­ sympathetic reflexes inhibit gastrointestinal motility. Symptoms of delayed oesophageal transit include dysphagia and regurgitation. Delayed gastric emptying produces early satiety, anorexia, nausea, bloating, belch­ ing, postprandial vomiting, and pain. Lower gastro­ intestinal dysmotility manifests with constipation and, ­occasionally, diarrhoea. Evaluation Clinical In addition to a careful history and general physical and neurological examination, clinical evaluation of patients with suspected autonomic failure includes assessment of pupil size, symmetry and reactivity with both bright and dim light; measurement of blood pressure and heart rate after 2min in the supine position and at 1min and 2min after standing; and examination of the skin to identify areas of localized or generalized absence of or excessive sweating, and changes in skin temperature or colour in the hands and feet. Laboratory tests General laboratory tests that should be performed in patients with autonomic failure include determination of serum glucose (or in some cases haemoglobin A1c), thyroid-stimulating hormone and vitamin B12 levels. Serum and urine protein electrophoresis with immuno­ fixation, including light-chain quantitation, are indi­ cated to detect amyloid light-chain (AL) amyloidosis.17 SSA and SSB antibody testing for Sjögren syndrome may be helpful in some cases. Determination of ganglionic nicotinic acetylcholine receptor (gnAChR) anti­bodies,18 as well as paraneoplastic antibodies (particularly anti- Hu, P/Q and N‑type voltage-gated calcium channel and voltage-gated potassium complex anti­ bodies), is indicated in all patients with subacute onset of symp­ toms.18,19 Determination of forearm venous catecho­ lamines, including noradrenaline, dopamine and adrenaline, in the supine position and after 5–10 min of standing may be helpful in some cases; the results require careful interpretation, however, as they can be Parkinsonism, ataxia Autonomic studies, neuroimaging (MRI), polysomnogram, urodynamic studies, neuropsychometrics Autonomic studies, electromyography/nerve conduction studies, FBG, SPEP , UPEP , abdominal fat aspirate, ANCA, HIV, α-galactosidase, punch skin biopsy Associated manifestations Onset/ subtype Main or typical causes Specific evaluation* Autonomic studies Autoantibodies (gnAChR, paraneoplastic) Sjögren syndrome Paraneoplastic Diabetes Amyloidosis ‘Pure’ autonomic failure Multiple system atrophy Lewy body disorders ■ PD ■ DLB Autoimmune autonomic ganglionopathy Paraneoplastic autonomic ganglionopathy Vasculitis HIV Idiopathic Fabry disease Hereditary Peripheral neuropathy None (‘pure’ autonomic failure) Chronic, progressive Subacute sensory Distal painful Distal sensorimotor or any other type Acute or subacute Autoantibodies (SSA/SSB, paraneoplastic) Salivary gland biopsy Autonomic failure 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 B12 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. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 4. 4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol affected by concomitant use of drugs or impaired pre­ synaptic noradrenaline reuptake.20 Low or undetectable noradrenaline and adrenaline levels associated with increased dopamine levels indicate a deficit of dopamine β‑hydroxylase as the cause of orthostatic hypotension.21 Autonomic function tests Autonomic laboratory evaluation is indicated to deter­ mine the extent and severity of autonomic failure; to detect autonomic failure in patients with parkinsonism or ataxia; to assess small fibre function in patients with peripheral neuropathy or ganglionopathy; to evalu­ ate patients with symptoms of orthostatic intolerance; and to obtain objective evidence of disease progression or response to medications.22,23 The most commonly used tests assess sudomotor, cardiovagal and adren­ ergic vasomotor functions. Sudomotor function tests include the thermoregulatory sweat test (TST), which assesses the integrity of central and peripheral sudo­ motor pathways,22 and sudomotor axon reflex tests such as the quanti­tative sudomotor axon reflex test (QSART)23 and the quanti­ tative direct and indirect test of sudo­ motor function (QDIRT),24 which assess the peripheral sympa­thetic cholinergic innervation of the sweat glands. Tests that assess vagal control of the sinus node (cardio­ vagal function) include the heart rate response to deep breathing,25 and the heart rate response to the Valsalva manoeuvre, or Valsalva ratio.26 Tests that indirectly assess sympathetic vasomotor function include the beat-to-beat blood pressure profile during the Valsalva manoeuvre27 and the blood pressure response to head-up tilt.28 The physiological basis, methods, normal and abnormal responses, and pitfalls of these and other autonomic ­function tests have been the subject of several reviews.29–31 Other tests Electromyography is indicated for patients with periph­ eral neuropathy or ganglionopathy, and MRI may be helpful in evaluation of central autonomic disorders. Gastrointestinal motility and urodynamic studies might be indicated in some cases. Polysomnography to detect sleep-related respiratory dysfunction is indicated in all patients with suspected MSA. Tissue biopsy Skin biopsy with assessment of intraepidermal nerve fibre density (IENFD) can be useful in the evaluation of SFN;32,33 immunocytochemical markers allow quanti­ fication of the density of innervation of sweat gland34 and pilomotor35 nerves. Abdominal fat aspirate or sural nerve biopsy are indicated to evaluate for amyloidosis.36,37 Immunostaining and laser microdissection, along with mass spectrometry-based proteomic analysis of amyloid deposition, allow identification of the specific subtype of amyloid protein.38 Neurodegenerative synucleinopathies Autonomic failure is an important manifestation of neuro­ degenerative disorders characterized by the pres­ ence of intracellular inclusions containing α‑synuclein. These conditions include MSA,1,39–41 and Lewy body ­ disorders such as PD, DLB or PAF.42,43 Multiple system atrophy MSA was initially defined as a sporadic neurodegenera­ tive disease characterized by any combination of auto­ nomic failure, parkinsonism, cerebellar ataxia and/or pyramidal signs.44 The prevalence of MSA has been esti­ mated at 4–8 cases per 100,000 people, and its incidence varies from 0.6 cases per 100,000 person-years in the general population to 3 cases per 100,000 person-years in people above 50 years of age.1,40 The neuropathological hallmark of MSA is the accumu­ lation of α‑synuclein-immunoreactive glial cytoplasmic inclusions in oligodendrocytes, and neuronal loss in the striatum, substantia nigra pars compacta, pontine nuclei, inferior olivary nuclei, cerebellum, and premotor auto­ nomic nuclei.45 According to current criteria, probable MSA is defined as a sporadic, progressive, adult-onset (over 30 years of age) disease characterized by autonomic failure, with urinary incontinence and erectile dysfunc­ tion (in males) or orthostatic hypotension (blood pres­ sure decrease ≥30mmHg systolic or ≥15mmHg diastolic within 3min of standing), as well as parkinsonism poorly responsive to levodopa (MSA‑P), or cerebellar syndrome (MSA‑C).39 A large European study including 437 patients with probable (72%) or possible (28%) MSA showed that symptomatic autonomic failure was present in almost all cases; the most frequent manifestations were urinary dys­ function (83%) and orthostatic hypotension (75%).41 In a study on 29 autopsy-confirmed cases, a pattern consist­ ing of severe and progressive adrenergic and sudomotor failure was highly predictive of MSA.46 Although MSA is still generally regarded as a sporadic disease, several studies have identified familial cases.1 A recent study identified mutations of the coenzyme Q2 (COQ2) gene in Japanese families with MSA, and COQ2 variants were also associated with an increased risk of sporadic MSA.47 Median survival from symptom onset in MSA is 8–10 years, but the spectrum ranges from 4–15 years. Natural history studies indicate that presence of the parkinsonian variant, early onset of autonomic failure, incomplete bladder emptying, and shorter duration of symptoms at baseline are all factors that predict shorter survival.48–50 In MSA, orthostatic hypotension occurs at earlier stages and is more severe than in PD.51 Orthostatic hypotension in MSA reflects involvement of pre­ gangli­ onic sympathetic neurons52 and sympathoexcitatory neurons of the rostral ventrolateral medulla.53 Urogenital manifestations may herald the onset of MSA.51 Bladder dysfunction in MSA is characterized by urinary urgency, followed by incontinence and incomplete bladder empty­ ing. These manifestations result from a combination of detrusor hyperreflexia and urethral sphincter weakness followed by detrusor contraction failure,54 and they reflect involvement of the pontine micturition centre,55 sacral preganglionic neurons56 and Onuf nucleus.57 Erectile dysfunction is almost always present in male REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 5. NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 5 patients with MSA.58 Upper gastrointestinal symptoms, constipation and faecal incontinence occur frequently; anal sphincter electromyography shows large motor unit potentials indicating denervation due to loss of Onuf nucleus motor neurons.59 Anhidrosis occurs in the majority of patients with MSA, and the percentage of the body affected by anhidrosis may help to differentiate MSA from PD.60,61 An important manifestation of disrupted brainstem homeostatic mechanisms in MSA is sleep-related dis­ ordered breathing, including sleep apnoea and laryn­ geal stridor, which occur in up to 70% of patients with MSA.62–64 Whereas the development of central hypo­ ventilation and laryngeal stridor is closely related to the severity of autonomic failure, respiratory manifestations can occur early in the course of MSA.65 Laryngeal dys­ tonia with inspiratory adduction of the vocal cords might underlie laryngeal stridor in at least some individuals with MSA.66 Impaired automatic ventilation reflects involve­ ment of the brainstem respiratory network, including the pre-Bötzinger complex67 and the medullary raphe,68 which may predispose patients to sudden death.69 Lewy body disorders Accumulation of Lewy neurites and Lewy bodies in the ENS, autonomic ganglia, peripheral autonomic nerve terminals, and intermediolateral cell column can be detected at autopsy of cases of incidental Lewy body disease (ILBD), PAF, early-stage PD, and DLB.70–73 α‑Synuclein inclusions can be also detected in cutan­eous nerves of patients with PAF74 or PD.75 These findings indicate that Lewy body disorders constitute a contin­ uum of progressive involvement of areas of the periph­ eral autonomic nervous system, and subsequently the CNS, by α‑synuclein neuropathology, which may range clinically from asymptomatic cases (ILBD) to pure auto­ nomic failure (PAF), and might eventually manifest with ­ parkinsonism (PD), dementia (DLB) or both. Pure autonomic failure PAF was first described by Bradbury and Eggleston in 1925, and was initially termed idiopathic orthostatic hypotension. PAF is a rare, sporadic, adult-onset dis­ order characterized by symptomatic orthostatic hypo­ tension and variable gastrointestinal, bladder and sexual dysfunction, in the absence of somatic motor deficits. Onset is typically between 50 and 70 years of age.42 The symptoms of PAF are insidious in onset, and are less progressive and disabling than those in other neuro­ degenerative autonomic disorders. Some patients may have bladder symptoms, erectile or ejaculatory dysfunc­ tion, or impaired sweating, before developing ortho­ static hypotension. Constipation is common, but other symptoms of gastrointestinal dysmotility are rare.76 The diagnosis of PAF is always tentative; after a few years, many patients with presumed PAF may develop cerebel­ lar, extrapyramidal or cognitive deficits indicating MSA, PD or DLB. Thus, the diagnosis of PAF requires at least a 5 year history of isolated autonomic dysfunction without other neurological manifestations. PAF is a clinical syndrome reflecting involvement of the autonomic gan­ glion cells and their postganglionic axons. This disorder can have a variety of different pathobiological substrates. Patients with PAF have been shown to have accumula­ tion of α‑synuclein-containing Lewy bodies and neurites in sympathetic ganglia peripheral tissues, including skin sympathetic nerves.74,75 However, some cases of PAF are associated with low-titre gnAChR antibodies, suggesting that this condition can reflect a chronic form of AAG in some instances.77 Parkinson disease Autonomic dysfunction—particularly gastrointestinal dysmotility—is a prominent nonmotor manifestation of PD.43 Gastrointestinal dysmotility in PD can affect any level of the gastrointestinal tract.78 Gastrointestinal symptoms can occur at an early stage of the disease;79 for example, constipation may precede the development of motor symptoms by several years.80,81 Excessive drooling in PD must reflect oropharyngeal dysphagia, as salivary secretion is reduced even at an early stage of disease;82,83 this finding is consistent with the presence of α‑synuclein pathology in the salivary gland.84 Oesophageal dysmotility and delayed gastric emptying are frequent manifestations in PD,85,86 and are likely to reflect early involvement of the dorsal motor nucleus of the vagus.70 The prevalence of constipation ranges from 20–89%,79 and is primarily attributable to slow colonic transit, reflecting the early involvement of the ENS.87 However, defecatory dysfunc­ tion due to paradoxical contraction of the ­ puborectalis muscle is also an important contributory factor.85,88 Orthostatic hypotension is estimated to occur in 16–58% of patients with PD.89 In general, orthostatic hypotension in PD is asymptomatic and tends to occur at later stages of disease than in MSA, but in a subgroup of patients it may be early and prominent, manifesting even before the initiation of dopaminergic therapy.89 Urinary symptoms occur in 38–71% of patients with PD.77,90,91 The most prominent symptom is nocturia, fol­ lowed by urgency and frequency, and the most common urodynamic finding is detrusor overactivity.92 Nocturia is the only urinary symptom that improves after deep brain stimulation of the subthalamic nucleus. Differential diagnosis The differential diagnosis between MSA‑P and PD with autonomic failure can be difficult. A prospective study showed that autonomic indices, particularly the percent­ age of the body affected by anhidrosis, were significantly and persistently more abnormal in MSA than in PD with autonomic failure.61 Urodynamic findings such as large post-micturitional residual volume (100ml), detrusor external sphincter dyssynergia, and open bladder neck at the start of filling are highly suggestive of MSA.54 Anal sphincter denervation due to loss of Onuf nucleus motor neurons may help to distinguish MSA from PD within the first few years of onset of motor symptoms, but this finding is not specific for MSA, and can occur in progressive supranuclear palsy or in PD patients with chronic constipation.93 REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 6. 6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol Structural brain imaging, including diffusion- weighted and diffusion tensor MRI, and functional imaging, including PET and single-photon emis­ sion CT (SPECT), might aid the differential diagnosis between MSA and PD.1 The presence of atrophy in the putamen, middle cerebellar peduncle and pons on MRI serve as supporting features for the diagnosis of pos­ sible MSA.39 Signal changes on T2-weighted images, including the slit-like void signal (hypointensity of the posterior putamen surrounded by hyperintense lateral putaminal rim) and the hot cross bun sign (due to myelin loss in the basis pontis) are more charac­ teristic of MSA but are not specific for this condition. Diffusion-weighted imaging shows elevated apparent diffusion coefficient in the putamen, pons and middle cerebellar peduncle in MSA.94 In MSA‑P, but not in PD, brain perfusion SPECT shows striatal hypoperfusion,95 and 18 F-fluorodeoxyglucose PET shows striatal hypo­ metabolism.96 By contrast, dopamine transporter SPECT using 123 I-FP-CIT demonstrates reduced striatal binding in both disorders.97 Myocardial scintigraphy using 123 I-metaiodo­benzyl­ guanidine (MIBG)98 or 6‑18 F-fluorodopamine PET99 assesses postganglionic sympathetic innervation of the heart; these modalities are potential tools to differenti­ ate between MSA and Lewy body disorders. However, although initial studies suggested that loss of cardiac sympathetic innervation in PD or PAF could help in distinguishing Lewy body disorders from MSA,99,100 decreased cardiac MIBG uptake has also been found in up to 30% of patients with MSA,101 possibly reflecting coexistent Lewy body pathology. Dementia with Lewy bodies Severe autonomic dysfunction, as well as repeated falls and syncope, are features supporting the diagnosis of DLB.102 Orthostatic hypotension, urinary frequency and urge incontinence can be disabling symptoms in DLB,103,104 and symptomatic orthostatic hypotension may occur in 30–50% of cases. On the basis of auto­ nomic laboratory evaluation, the severity of autonomic failure in DLB is intermediate between that seen in MSA and PD.105 Other neurodegenerative disorders Other central neurodegenerative disorders can manifest with autonomic failure that reflects peripheral autonomic involvement. These conditions, which may clinically mimic MSA or Lewy body disorders, include fragile X tremor–ataxia syndrome, in which bladder symptoms might be prominent;106 adult-onset autosomal dominant leukodystrophy, which is associated with pure sympa­ thetic failure;107 and a novel prion disorder character­ ized by diarrhoea and length-dependent, predominantly sensory and autonomic axonal neuropathy, followed by cognitive decline.108 Autoimmune autonomic disorders Autoimmune autonomic disorders are characterized by acute or subacute onset of generalized or restricted autonomic failure, including gastrointestinal enteropathy. Early recognition of these disorders is important because they are disabling and might be the initial manifestation of an underlying neoplasm. In addition, patients with autoimmune autonomic disorders may show substantial improvement with ­ immunomodulatory therapy.4,109 Autoimmune autonomic ganglionopathy AAG (formerly known as acute pandysautonomia or idiopathic subacute autonomic neuropathy18,110 ) is characterized by severe autonomic failure that develops over the course of days or weeks in a previously healthy person. The onset of symptoms may follow a viral infec­ tion, minor surgical procedure, or vaccination. The most common manifestation is generalized sympathetic, parasympathetic and ENS failure, but the spectrum is broad; symptoms might be confined to pure choliner­ gic neuropathy manifested by sicca syndrome, isolated adrenergic neuropathy, or isolated gastrointestinal dys­ motility. Patients may recover spontaneously, but only one-third experience substantial functional ­improvement without treatment. Approximately 50% of cases of subacute AAG are associ­ ated with gnAChR antibodies18 that block trans­ mission at the autonomic ganglia. Antibody titres correlate with the severity of autonomic failure.19,111 gnAChR antibodies can also be associated with chronic or restricted forms of autonomic dysfunction,77 and with neurological or paraneoplastic disorders unrelated to the autonomic nervous system.19 In one series, 30% of gnAChR-seropositive patients had other paraneoplastic antibodies in the setting of occult cancer, most com­ monly adenocarcinoma.19 In some cases, AAG coexists with other autoimmune disorders, such as myasthenia gravis, in patients with occult cancer.112,113 Several observational studies report that patients with gnAChR-seropositive AAG benefit from intra­ venous immunoglobulin, plasma exchange, prednisone, mycophenolate, azathioprine and rituximab, either individually or in combination.19,114–116 A substantial percentage of patients with AAG are seronegative for gnAChR and other autoantibodies;117 some of these patients may also show symptomatic improvement with ­immunomodulatory therapy.116 Paraneoplastic autonomic neuropathy A type of subacute autonomic neuropathy that is clinically indistinguishable from idiopathic AAG might occur as a manifestation of an occult neoplasm, most commonly small-cell lung carcinoma associated with anti-Hu antibodies or, less frequently, thymoma or other neo­ plasms.118,119 Like AAG, paraneoplastic autonomic neuro­ pathy may manifest with generalized autonomic failure, or with isolated enteric ganglionopathy leading to the intestinal pseudo-obstruction syndrome.120,121 Acute autonomic and sensory neuropathy Acute autonomic and sensory neuropathy is a rare dis­ order that differs from AAG in that profound autonomic failure is associated with various degrees of sensory REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 7. NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 7 impairment, including loss of pain and temperature sensation, followed by sensory ataxia due to impair­ ment of proprioception.109,122,123 The mean age of onset is in the late second decade, but can vary from child­ hood to the elderly. A triggering event, such as respira­ tory or gastrointestinal tract infections, is reported in two-thirds of patients. Gastrointestinal dysmotility is the most prominent autonomic symptom; the sensory loss affects the proximal regions of the limbs, face, scalp and trunk, and tends to be asymmetrical and segmen­ tal. Pain in the involved region is a common and serious symptom. Other features of this disorder are reduction of sensory nerve action potentials and increased T2 signal in the posterior columns of the cervical spinal cord.123 Autopsy studies have shown severe neuronal cell loss in the thoracic sympathetic and dorsal root ganglia, and in Auerbach’s plexus.123 Sjögren syndrome Primary Sjögren syndrome manifests with a wide variety of peripheral neuropathies or ganglionopathies.124 Dorsal root ganglionopathy may manifest with neuropathic pain or ataxic neuropathy; autonomic neuropathy might be prominent in some cases, but is the least frequent form of neuropathy overall.124 Autoantibodies against muscarinic M3 receptors have been identified in a subset of patients with Sjögren syn­ drome.125 These antibodies are particularly prevalent in juvenile cases126 and have cholinergic blocking actions in vitro.127 Some patients with Sjögren syndrome and chronic progressive autonomic failure have gnAChR antibodies, which may predict a positive response to immunomodulatory therapy.128 Peripheral neuropathies Peripheral neuropathies associated with autonomic failure present with various phenotypes, including typical distal symmetric sensorimotor neuropathy; dorsal root ganglionopathy manifesting with neuropathic pain or sensory ataxia; demyelinating, predominantly motor polyradiculopathy; or distal painful neuropathy. The spectrum of severity of autonomic failure is highly vari­ able in these disorders, ranging from being a major cause of disability, as can occur in neuropathies associated with diabetes or amyloidosis, to restricted distal vaso­ motor and sudomotor impairment in the lower limbs, as observed in painful SFN. Diabetic autonomic neuropathy Diabetic neuropathies are heterogeneous: they include typical diabetic sensorimotor polyneuropathy, painful diabetic neuropathy, and diabetic autonomic neuro­ pathy.129 Estimates of the prevalence of diabetic auto­ nomic neuropathy vary depending of the source of information (community, clinic or tertiary referral centre) and the type of tests performed to assess autonomic func­ tion.129 Using strict criteria based on autonomic testing abnormalities, the prevalence in one study was 16.8% for patients with type 1 and 34.3% for patients with type 2 diabetes mellitus.130 Diabetic autonomic neuropathy can affect cardio­ vascular, gastrointestinal, urogenital and sudomotor functions to various degrees. In most of the typical cases, a combination of cardiovascular tests and sudomotor tests allows its detection in otherwise asympto­ matic patients. An impaired heart rate response to deep breathing­ —an indicator of cardiovagal failure—has the greatest specificity (~80%).131,132 Characteristic mani­ festations of diabetic cardiovascular autonomic neuro­ pathy include resting tachycardia, exercise intolerance, orthostatic hypotension, intraoperative cardiovascular instability, and silent myocardial infarction–silent ischae­ mia syndrome. Cardiovascular autonomic neuropathy has been associated with increased morbidity and mor­ tality,133 so screening for this condition should be per­ formed at the time of diagnosis of type 2 diabetes and 5 years after the diagnosis of type 1 diabetes, particularly in high-risk patients.129 Upper gastrointestinal dysmotility, which is present in 40–50% of longstanding cases of diabetes, may affect postprandial blood glucose levels and glycaemic control. Constipation is the most frequent gastrointesti­ nal symptom in diabetes, and was reported by 60% of patients in one study.134 Profuse, watery and predomi­ nantly nocturnal diarrhoea135 and faecal incontinence136 may be disabling in some patients. Bladder symptoms occur in up to 50% of patients; 43–87% of type 1 and 25% of type 2 diabetic patients have abnormal urodynamic findings.137,138 Erectile dysfunction has a reported preva­ lence of 35–90% in patients with diabetes.139,140 Length- dependent loss of thermoregulatory sweating is common, and is occasionally associated with compensatory hyper­ hidrosis.22 Gustatory sweating can result from aberrant innervation of sweat glands by parasympathetic fibres.141 Some patients with diabetes develop an acute painful neuropathy associated with autonomic failure within a short time of commencing tight glucose control.142 The symptoms may be associated with abnormal autonomic test results and reduced IENFD on punch skin biopsy, and can all improve over time.142 Amyloid neuropathy Amyloidoses manifesting with autonomic neuro­ pathy include the AL type (which may be associated with myeloma or macroglobulinaemia), and familial amyloid polyneuropathy (FAP), which is most com­ monly linked to mutations in the transthyretin (TTR) gene. The types of amyloid neuropathy associated with autonomic failure are highly heterogeneous.143,144 In a retrospective study on 65 cases of AL amyloidosis or FAP, generalized autonomic failure was associated with painful (62%) or nonpainful (17%) sensorimotor poly­ neuropathy or distal SFN (5%); autonomic failure also occurred without neuropathy (11%), but polyneuro­ pathy without generalized autonomic failure was infre­ quent (6%).143 In AL amyloidosis, peripheral neuropathy may be the presenting feature or an incidental finding of the disease, and can occur in up to 20% of cases.145 The median survival of patients with AL amyloidosis who have peripheral neuropathy has improved with the REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 8. 8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol Table 1 | General management of autonomic failure Symptom Approach Comments Orthostatic hypotension Patient education: recognize atypical symptoms of 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 The most important aspect of management Sleep with the head elevated 4in (reverse Trendelenburg position) Avoids supine hypertension and nocturnal polyuria Postural countermanouevres (for example, leg crossing, squatting) Increases orthostatic tolerance Support garments (waist-high support stockings, abdominal binder) Reduces venous pooling Volume expansion (daily intake of 8–10g sodium, fluid intake of at least 2.0–2.5l) Initial step before pharmacological management Rapid ingestion of 500ml tap water Rapidly increases blood pressure independently of volume expansion Dietary changes; avoid alcohol; eat small, frequent meals (carbohydrate is best ingested with the last meal of the day) Prevents splanchnic vasodilation Florinef (0.1–0.4mg daily) Adverse effects include supine hypertension, ankle oedema, hypokalaemia, hypomagnesaemia, headache, and reduced effects of warfarin Midodrine (10–40mg daily) Adverse effects include scalp tingling or itching, goose bumps, supine hypertension, and urinary urgency or retention Pyridostigmine (60–240mg daily) Adverse effects include nausea, abdominal cramps, diarrhoea, increased salivation, urinary urgency and bradycardia Droxidopa (100–600mg TID) Main potential adverse effect is supine hypertension Erythropoietin (25–75U/kg SQ) Iron supplementation is usually required; supine hypertension may occur Desmopressin nasal spray (5–40μg) Risk of water intoxication and hyponatraemia Octreotide (25–200μg SQ) Adverse effects include nausea and abdominal cramps, hypertension, risk of gallstones and postprandial hyperglycaemia Neurogenic bladder Anticholinergics (for example, oxybutynin, tolterodine); clean intermittent self-catheterization Adverse effects include constipation, dry mouth, risk of glaucoma, and worsening of cognitive function Drooling Anticholinergics; botulinum toxin Anticholinergics may worsen confusion; botulinum toxin may worsen dysphagia Gastroparesis Metoclopramide (5–20mg QID); domperidone (10–30mg QID); erythromycin (50–250mg QID); pyridostigmine (30–60mg TID); bethanechol (25mg QID); nutritional support (low-fat, low-fibre oral supplementation; infusion of formula via a jejunal feeding tube) Gastroparesis may impair absorption of levodopa; metoclopramide is contraindicated in parkinsonian disorders; domperidone may cause QT prolongation, and is not available in the USA Constipation Fibre supplementation (15g per day); bulk agents (for example, psyllium [1tsp up to three times daily], methylcellulose); osmotic laxatives (for example, milk of magnesia [two tablets up to four times daily], polyethylene glycol [17g in 25ml once or twice daily], lactulose [15–30ml TID]); docusate (stool softener, 100mg BID); lubiprostone (24μg BID); pyridostigmine (up to 180–540mg); bisacodyl (10mg up to three times a week); antibiotics to prevent bacterial overgrowth; surgery for intractable colonic inertia Donepezil and other cholinesterase inhibitors may cause diarrhoea Erectile dysfunction Oral phosphodiesterase type 5 inhibitors (for example, sildenafil 25–100mg) Adverse effects include headache, flushing, nasal congestion, blue vision, and non-arteritic ischaemic optic neuropathy; these drugs may worsen orthostatic hypotension Intracavernous drugs (for example, alprostadil, papaverine) Adverse effects include penile pain, oedema and haematoma, palpable nodules or plaques, and priapism Abbreviations: BID, twice a day; QID, four times a day; SQ, subcutaneous; TID, three times a day. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 9. NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 9 combination of melphalan, prednisone and autologous stem cell transplantation.146 The clinical and pathological features of FAP have been reviewed in detail elsewhere.147,148 Over 100 muta­ tions in the TTR gene have been identified, the most common (~50% of cases of FAP worldwide) of which causes a Val30Met substitution. Different TTR variants contribute to variable age at diagnosis, degree of cardiac involvement, and survival.149 The first-line specific treat­ ment of choice for Val30Met TTR-FAP is liver transplan­ tation.150 In addition, tafamidis, a stabilizer of tetrameric transthyretin, has shown short-term effectiveness in slowing the progression of peripheral neuropathy at very early stages of Val30Met TTR-FAP.151 Painful small fibre neuropathies Selective involvement of postganglionic as well as noci­ cep­ tive fibres in painful SFN typically manifests with distal anhidrosis (or sometimes hyperhidrosis) and vasomotor dysfunction (generally erythema, but some­ times vasoconstriction and coldness) in the lower limbs. In some cases, SFN can result in more-generalized autonomic failure that may be symptomatic or detect­ able with autonomic testing. Important examples of painful SFN are those associated with HIV infections152 and Fabry disease,153 in addition to the types related to dia­ betes and amyloidosis, as highlighted above. Distal sympathetic sudomotor and vasomotor failure in con­ junction with neuropathic pain also occur in sodium channelopathies associated with gain-of-function SCNA9 mutations, both in familial syndromes154 and in a substantial proportion of idiopathic SFN cases.155 The SCNA9 gene encodes the Nav 1.7 voltage-gated sodium channel, which is expressed in nociceptive dorsal root ganglion and sympathetic neurons. These gain-of-­ function mutations cause increased excitability of the nociceptor and hypoexcitability of sympathetic ganglion neurons,156,157 which might explain the coexistence of neuropathic pain and postganglionic sympathetic failure. Management issues A detailed discussion of the management of autonomic failure is beyond the scope of this Review, but the general approach is summarized in this section and in Table 1. The principles of management include discontinua­ tion of potentially causative drugs; immunomodula­ tory therapy for autoimmune disorders; management of diabetes, amyloidosis or other potentially treatable cause of autonomic neuropathy; patient education; non-­ pharmacological approaches; and pathophysiologically based drug therapy. Patient education is a fundamen­ tal aspect of management. For example, patients with orthostatic hypotension should be instructed to recog­ nize atypical symptoms and avoid precipitating factors. Appropriate fluid and sodium intake, dietary adjust­ ments and regular exercise are beneficial in most cases of autonomic failure. Conclusions The wide spectrum of clinical manifestations and dis­ orders associated with autonomic failure requires a systematic clinical and laboratory approach to establish the diagnosis, particularly in cases where autonomic failure is disabling or life-threatening. A careful history and examination is the mainstay of diagnosis, and lab­ oratory and other ancillary tests must be prioritized to search for potentially treatable causes. Patient education, avoidance of precipitating factors, non-pharmacological approaches, and pathophysiologically based drug therapy form the basis of current treatment of these disorders. Further elucidation of the pathobiological mechanisms that lead to immune, metabolic or degenerative damage or dysfunction of central and peripheral autonomic neurons will provide more-specific therapeutic targets. Review criteria A PubMed search from 1st January 1975 to 15th December 2013 was performed using the following terms (individually and in various combinations): “autonomic failure”, “autonomic neuropathy”, “orthostatic hypotension”, “multiple system atrophy”, “Parkinson disease”, “Lewy body disease”, “dementia with Lewy bodies”, “autoimmune autonomic ganglionopathy”, “paraneoplastic”, “diabetic neuropathy”, “amyloid neuropathy”, “Sjögren syndrome”, “leukoencephalopathy”, “fragile X” and “prion”. The search was limited to full articles published in English. Other articles were identified from bibliographies of the retrieved articles. 1. Stefanova, N., Bucke, P ., Duerr, S. Wenning, G. K. Multiple system atrophy: an update. Lancet Neurol. 8, 1172–1178 (2009). 2. Cersosimo, M. G. Benarroch, E. E. Autonomic involvement in Parkinson’s disease: pathology, pathophysiology, clinical features and possible peripheral biomarkers. J. Neurol. Sci. 313, 57–63 (2012). 3. Vernino, S. Autoimmune and paraneoplastic channelopathies. Neurotherapeutics 4, 305–314 (2007). 4. Muppidi, S. Vernino, S. Autoimmune autonomic failure. Handb. Clin. Neurol. 117, 321–327 (2013). 5. Freeman, R. Autonomic peripheral neuropathy. Lancet 365, 1259–1270 (2005). 6. Freeman, R. et al. Consensus statement on the definition of orthostatic hypotension, neurally mediated syncope and the postural tachycardia syndrome. Auton. Neurosci. 161, 46–48 (2011). 7. Mathias, C. J. et al. Differential blood pressure and hormonal effects after glucose and xylose ingestion in chronic autonomic failure. Clin. Sci. (Lond.) 77, 85–92 (1989). 8. Mathias, C. J., Mallipeddi, R. Bleasdale- Barr, K. Symptoms associated with orthostatic hypotension in pure autonomic failure and multiple system atrophy. J. Neurol. 246, 893–898 (1999). 9. Freeman, R. Clinical practice. Neurogenic orthostatic hypotension. N. Engl. J. Med. 358, 615–624 (2008). 10. Benarroch, E. E. The arterial baroreflex: functional organization and involvement in neurologic disease. Neurology 71, 1733–1738 (2008). 11. Cheshire, W. P. Freeman, R. Disorders of sweating. Semin. Neurol. 23, 399–406 (2003). 12. Fowler, C. J., Griffiths, D. de Groat, W. C. The neural control of micturition. Nat. Rev. Neurosci. 9, 453–466 (2008). 13. Holstege, G. Micturition and the soul. J. Comp. Neurol. 493, 15–20 (2005). 14. Panicker, J. N. Fowler, C. J. The bare essentials: uro-neurology. Pract. Neurol. 10, 178–185 (2010). 15. Travagli, R. A., Hermann, G. E., Browning, K. N. Rogers, R. C. Brainstem circuits regulating gastric function. Annu. Rev. Physiol. 68, 279–305 (2006). 16. Furness, J. B. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 9, 286–294 (2012). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 10. 10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol 17. Kyle, R. A. Bayrd, E. D. Amyloidosis: review of 236 cases. Medicine (Baltimore) 54, 271–299 (1975). 18. Vernino, S. et al. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N. Engl. J. Med. 343, 847–855 (2000). 19. McKeon, A., Lennon, V. A., Lachance, D. H., Fealey, R. D. Pittock, S. J. Ganglionic acetylcholine receptor autoantibody: oncological, neurological, and serological accompaniments. Arch. Neurol. 66, 735–741 (2009). 20. Goldstein, D. S. Catecholamines 101. Clin. Auton. Res. 20, 331–352 (2010). 21. Robertson, D. et al. Isolated failure of autonomic noradrenergic neurotransmission. Evidence for impaired β‑hydroxylation of dopamine. N. Engl. J. Med. 314, 1494–1497 (1986). 22. Fealey, R. D., Low, P . A. Thomas, J. E. Thermoregulatory sweating abnormalities in diabetes mellitus. Mayo Clin. Proc. 64, 617–628 (1989). 23. Low, P . A. Autonomic nervous system function. J. Clin. Neurophysiol. 10, 14–27 (1993). 24. Gibbons, C. H., Illigens, B. M., Centi, J. Freeman, R. QDIRT: quantitative direct and indirect test of sudomotor function. Neurology 70, 2299–2304 (2008). 25. Bennett, T. et al. Assessment of vagal control of the heart in diabetes. Measures of R–R interval variation under different conditions. Br. Heart J. 39, 25–28 (1977). 26. Booth, R. W. Ryan, J. M. The clinical use of the Valsalva maneuver. Heart Bull. 10, 111–113 (1961). 27. Sandroni, P ., Benarroch, E. E. Low, P . A. Pharmacological dissection of components of the Valsalva maneuver in adrenergic failure. J. Appl. Physiol. 71, 1563–1567 (1991). 28. Cooke, J. et al. Sitting and standing blood pressure measurements are not accurate for the diagnosis of orthostatic hypotension. QJM 102, 335–339 (2009). 29. Low, P . A. Testing the autonomic nervous system. Semin. Neurol. 23, 407–421 (2003). 30. Freeman, R. Assessment of cardiovascular autonomic function. Clin. Neurophysiol. 117, 716–730 (2006). 31. Hilz, M. J. Dutsch, M. Quantitative studies of autonomic function. Muscle Nerve 33, 6–20 (2006). 32. Lauria, G. et al. European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. Eur. J. Neurol. 17, 903–912, e44–e49 (2010). 33. Donadio, V. et al. Peripheral autonomic neuropathy: diagnostic contribution of skin biopsy. J. Neuropathol. Exp. Neurol. 71, 1000–1008 (2012). 34. Gibbons, C. H., Illigens, B. M., Wang, N. Freeman, R. Quantification of sweat gland innervation: a clinical-pathologic correlation. Neurology 72, 1479–1486 (2009). 35. Nolano, M. et al. Quantification of pilomotor nerves: a new tool to evaluate autonomic involvement in diabetes. Neurology 75, 1089–1097 (2010). 36. Libbey, C. A., Skinner, M. Cohen, A. S. Use of abdominal fat tissue aspirate in the diagnosis of systemic amyloidosis. Arch. Intern. Med. 143, 1549–1552 (1983). 37. Kyle, R. A. Greipp, P . R. Amyloidosis (AL). Clinical and laboratory features in 229 cases. Mayo Clin. Proc. 58, 665–683 (1983). 38. Klein, C. J. et al. Mass spectrometric-based proteomic analysis of amyloid neuropathy type in nerve tissue. Arch. Neurol. 68, 195–199 (2011). 39. Gilman, S. et al. Second consensus statement on the diagnosis of multiple system atrophy. Neurology 71, 670–676 (2008). 40. Wenning, G. K., Ben Shlomo, Y., Magalhaes, M., Daniel, S. E. Quinn, N. P . Clinical features and natural history of multiple system atrophy. An analysis of 100 cases. Brain 117, 835–845 (1994). 41. Kollensperger, M. et al. Presentation, diagnosis, and management of multiple system atrophy in Europe: final analysis of the European multiple system atrophy registry. Mov. Disord. 25, 2604–2612 (2010). 42. Kaufmann, H. Biaggioni, I. Autonomic failure in neurodegenerative disorders. Semin. Neurol. 23, 351–363 (2003). 43. Chaudhuri, K. R. Schapira, A. H. Non-motor symptoms of Parkinson’s disease: dopaminergic pathophysiology and treatment. Lancet Neurol. 8, 464–474 (2009). 44. Graham, J. G. Oppenheimer, D. R. Orthostatic hypotension and nicotine sensitivity in a case of multiple system atrophy. J. Neurol. Neurosurg. Psychiatry 32, 28–34 (1969). 45. Papp, M. I., Kahn, J. E. Lantos, P . L. Glial cytoplasmic inclusions in the CNS of patients with multiple system atrophy (striatonigral degeneration, olivopontocerebellar atrophy and Shy–Drager syndrome). J. Neurol. Sci. 94, 79–100 (1989). 46. Iodice, V. et al. Autopsy confirmed multiple system atrophy cases: Mayo experience and role of autonomic function tests. J. Neurol. Neurosurg. Psychiatry 83, 453–459 (2012). 47. Multiple-System Atrophy Research Collaboration. Mutations in COQ2 in familial and sporadic multiple-system atrophy. N. Engl. J. Med. 369, 233–244 (2013). 48. Tada, M. et al. Early development of autonomic dysfunction may predict poor prognosis in patients with multiple system atrophy. Arch. Neurol. 64, 256–260 (2007). 49. O’Sullivan, S. S. et al. Clinical outcomes of progressive supranuclear palsy and multiple system atrophy. Brain 131, 1362–1372 (2008). 50. Wenning, G. K. et al. The natural history of multiple system atrophy: a prospective European cohort study. Lancet Neurol. 12, 264–274 (2013). 51. Wenning, G. K. et al. Time course of symptomatic orthostatic hypotension and urinary incontinence in patients with postmortem confirmed parkinsonian syndromes: a clinicopathological study. J. Neurol. Neurosurg. Psychiatry 67, 620–623 (1999). 52. Oppenheimer, D. R. Lateral horn cells in progressive autonomic failure. J. Neurol. Sci. 46, 393–404 (1980). 53. Benarroch, E. E., Smithson, I. L., Low, P . A. Parisi, J. E. Depletion of catecholaminergic neurons of the rostral ventrolateral medulla in multiple systems atrophy with autonomic failure. Ann. Neurol. 43, 156–163 (1998). 54. Winge, K. Fowler, C. J. Bladder dysfunction in Parkinsonism: mechanisms, prevalence, symptoms, and management. Mov. Disord. 21, 737–745 (2006). 55. Benarroch, E. E. Schmeichel, A. M. Depletion of corticotrophin-releasing factor neurons in the pontine micturition area in multiple system atrophy. Ann. Neurol. 50, 640–645 (2001). 56. Kennedy, P . G. Duchen, L. W. A quantitative study of intermediolateral column cells in motor neuron disease and the Shy–Drager syndrome. J. Neurol. Neurosurg. Psychiatry 48, 1103–1106 (1985). 57. Chalmers, D. Swash, M. Selective vulnerability of urinary Onuf motoneurons in Shy–Drager syndrome. J. Neurol. 234, 259–260 (1987). 58. Kirchhof, K., Apostolidis, A. N., Mathias, C. J. Fowler, C. J. Erectile and urinary dysfunction may be the presenting features in patients with multiple system atrophy: a retrospective study. Int. J. Impot. Res. 15, 293–298 (2003). 59. Sakakibara, R. et al. Colonic transit time, sphincter EMG, and rectoanal videomanometry in multiple system atrophy. Mov. Disord. 19, 924–929 (2004). 60. Sandroni, P ., Ahlskog, J. E., Fealey, R. D. Low, P . A. Autonomic involvement in extrapyramidal and cerebellar disorders. Clin. Auton. Res. 1, 147–155 (1991). 61. Lipp, A. et al. Prospective differentiation of multiple system atrophy from Parkinson disease, with and without autonomic failure. Arch. Neurol. 66, 742–750 (2009). 62. Iranzo, A. Sleep and breathing in multiple system atrophy. Curr.Treat. Options Neurol. 9, 347–353 (2007). 63. Ghorayeb, I., Bioulac, B. Tison, F. Sleep disorders in multiple system atrophy. J. Neural Transm. 112, 1669–1675 (2005). 64. Silber, M. H. Levine, S. Stridor and death in multiple system atrophy. Mov. Disord. 15, 699–704 (2000). 65. Glass, G. A., Josephs, K. A. Ahlskog, J. E. Respiratory insufficiency as the primary presenting symptom of multiple-system atrophy. Arch. Neurol. 63, 978–981 (2006). 66. Vetrugno, R. et al. Sleep-related stridor due to dystonic vocal cord motion and neurogenic tachypnea/tachycardia in multiple system atrophy. Mov. Disord. 22, 673–678 (2007). 67. Schwarzacher, S. W., Rub, U. Deller, T. Neuroanatomical characteristics of the human pre-Bötzinger complex and its involvement in neurodegenerative brainstem diseases. Brain 134, 24–35 (2011). 68. Benarroch, E. E., Schmeichel, A. M., Low, P . A. Parisi, J. E. Depletion of putative chemosensitive respiratory neurons in the ventral medullary surface in multiple system atrophy. Brain 130, 469–475 (2007). 69. Tada, M. et al. Depletion of medullary serotonergic neurons in patients with multiple system atrophy who succumbed to sudden death. Brain 132, 1810–1819 (2009). 70. Braak, H. et al. Stanley Fahn Lecture 2005: the staging procedure for the inclusion body pathology associated with sporadic Parkinson’s disease reconsidered. Mov. Disord. 21, 2042–2051 (2006). 71. Dickson, D. W. et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 115, 437–444 (2008). 72. Jellinger, K. A. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta Neuropathol. 116, 1–16 (2008). 73. Beach, T. G. et al. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol. 119, 689–702 (2010). 74. Shishido, T. et al. α‑Synuclein accumulation in skin nerve fibers revealed by skin biopsy in pure autonomic failure. Neurology 74, 608–610 (2010). 75. Wang, N., Gibbons, C. H., Lafo, J. Freeman, R. α‑Synuclein in cutaneous autonomic nerves. Neurology 81, 1604–1610 (2013). 76. Polinsky, R. J. Clinical autonomic neuropharmacology. Neurol. Clin. 8, 77–92 (1990). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 11. NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 11 77. Klein, C. M. et al. The spectrum of autoimmune autonomic neuropathies. Ann. Neurol. 53, 752–758 (2003). 78. Pfeiffer, R. F. Gastrointestinal dysfunction in Parkinson’s disease. Parkinsonism Relat. Disord. 17, 10–15 (2011). 79. Siddiqui, M. F., Rast, S., Lynn, M. J., Auchus, A. P . Pfeiffer, R. F. Autonomic dysfunction in Parkinson’s disease: a comprehensive symptom survey. Parkinsonism Relat. Disord. 8, 277–284 (2002). 80. Abbott, R. D. et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology 57, 456–462 (2001). 81. Savica, R. et al. Medical records documentation of constipation preceding Parkinson disease: a case–control study. Neurology 73, 1752–1758 (2009). 82. Proulx, M., de Courval, F. P ., Wiseman, M. A. Panisset, M. Salivary production in Parkinson’s disease. Mov. Disord. 20, 204–207 (2005). 83. Cersosimo, M. G. et al. Hyposialorrhea as an early manifestation of Parkinson disease. Auton. Neurosci. 150, 150–151 (2009). 84. Del Tredici, K., Hawkes, C. H., Ghebremedhin, E. Braak, H. Lewy pathology in the submandibular gland of individuals with incidental Lewy body disease and sporadic Parkinson’s disease. Acta Neuropathol. 119, 703–713 (2010). 85. Edwards, L. L., Quigley, E. M., Harned, R. K., Hofman, R. Pfeiffer, R. F. Characterization of swallowing and defecation in Parkinson’s disease. Am. J. Gastroenterol. 89, 15–25 (1994). 86. Castell, J. A. et al. Manometric abnormalities of the oesophagus in patients with Parkinson’s disease. Neurogastroenterol. Motil. 13, 361–364 (2001). 87. Wakabayashi, K., Takahashi, H., Takeda, S., Ohama, E. Ikuta, F. Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol. 76, 217–221 (1988). 88. Sakakibara, R., Uchiyama, T., Yamanishi, T., Shirai, K. Hattori, T. Bladder and bowel dysfunction in Parkinson’s disease. J. Neural Transm. 115, 443–460 (2008). 89. Sharabi, Y. Goldstein, D. S. Mechanisms of orthostatic hypotension and supine hypertension in Parkinson disease. J. Neurol. Sci. 310, 123–128 (2011). 90. Winge, K., Skau, A. M., Stimpel, H., Nielsen, K. K. Werdelin, L. Prevalence of bladder dysfunction in Parkinsons disease. Neurourol. Urodyn. 25, 116–122 (2006). 91. Blackett, H., Walker, R. Wood, B. Urinary dysfunction in Parkinson’s disease: a review. Parkinsonism Relat. Disord. 15, 81–87 (2009). 92. Winge, K. Nielsen, K. K. Bladder dysfunction in advanced Parkinson’s disease. Neurourol. Urodyn. 31, 1279–1283 (2012). 93. Sakakibara, R., Uchiyama, T., Yamanishi, T. Kishi, M. Sphincter EMG as a diagnostic tool in autonomic disorders. Clin.Auton. Res. 19, 20–31 (2009). 94. Seppi, K. et al. Progression of putaminal degeneration in multiple system atrophy: a serial diffusion MR study. Neuroimage 31, 240–245 (2006). 95. Feigin, A. et al. Tc‑99m ethylene cysteinate dimer SPECT in the differential diagnosis of parkinsonism. Mov. Disord. 17, 1265–1270 (2002). 96. Kwon, K. Y., Choi, C. G., Kim, J. S., Lee, M. C. Chung, S. J. Comparison of brain MRI and 18 F-FDG PET in the differential diagnosis of multiple system atrophy from Parkinson’s disease. Mov. Disord. 22, 2352–2358 (2007). 97. Varrone, A., Marek, K. L., Jennings, D., Innis, R. B. Seibyl, J. P . [123 I]β-CIT SPECT imaging demonstrates reduced density of striatal dopamine transporters in Parkinson’s disease and multiple system atrophy. Mov. Disord. 16, 1023–1032 (2001). 98. King, A. E., Mintz, J. Royall, D. R. Meta-analysis of 123 I-MIBG cardiac scintigraphy for the diagnosis of Lewy body-related disorders. Mov. Disord. 26, 1218–1224 (2011). 99. Goldstein, D. S., Holmes, C., Cannon, R. O. 3rd , Eisenhofer, G. Kopin, I. J. Sympathetic cardioneuropathy in dysautonomias. N. Engl. J. Med. 336, 696–702 (1997). 100. Treglia, G. et al. Diagnostic performance of iodine‑123‑metaiodobenzylguanidine scintigraphy in differential diagnosis between Parkinson’s disease and multiple-system atrophy: a systematic review and a meta-analysis. Clin. Neurol. Neurosurg. 113, 823–829 (2011). 101.Nagayama, H. et al. Abnormal cardiac [123 I]‑meta‑iodobenzylguanidine uptake in multiple system atrophy. Mov. Disord. 25, 1744–1747 (2010). 102.McKeith, I. G. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the Consortium on DLB International Workshop. J. Alzheimers. Dis. 9, 417–423 (2006). 103.Stubendorff, K., Aarsland, D., Minthon, L. Londos, E. The impact of autonomic dysfunction on survival in patients with dementia with Lewy bodies and Parkinson’s disease with dementia. PLoS ONE 7, e45451 (2012). 104.Kanazawa, M. et al. An autopsy case of dementia with Lewy bodies showing autonomic failure and dementia as the initial symptoms. Mov. Disord. 22, 1212–1213 (2007). 105.Thaisetthawatkul, P . et al. Autonomic dysfunction in dementia with Lewy bodies. Neurology 62, 1804–1809 (2004). 106.Leehey, M. A. Fragile X‑associated tremor/ataxia syndrome: clinical phenotype, diagnosis, and treatment. J. Investig. Med. 57, 830–836 (2009). 107.Guaraldi, P . et al. Isolated noradrenergic failure in adult-onset autosomal dominant leukodystrophy. Auton. Neurosci. 159, 123–126 (2011). 108.Mead, S. et al. A novel prion disease associated with diarrhea and autonomic neuropathy. N. Engl. J. Med. 369, 1904–1914 (2013). 109.Koike, H., Watanabe, H. Sobue, G. The spectrum of immune-mediated autonomic neuropathies: insights from the clinicopathological features. J. Neurol. Neurosurg. Psychiatry 84, 98–106 (2013). 110.Suarez, G. A., Fealey, R. D., Camilleri, M. Low, P . A. Idiopathic autonomic neuropathy: clinical, neurophysiologic, and follow-up studies on 27 patients. Neurology 44, 1675–1682 (1994). 111.Gibbons, C. H. Freeman, R. Antibody titers predict clinical features of autoimmune autonomic ganglionopathy. Auton. Neurosci. 146, 8–12 (2009). 112.Vernino, S., Cheshire, W. P . Lennon, V. A. Myasthenia gravis with autoimmune autonomic neuropathy. Auton. Neurosci. 88, 187–192 (2001). 113.Peltier, A. C. et al. Coexistent autoimmune autonomic ganglionopathy and myasthenia gravis associated with non‑small‑cell lung cancer. Muscle Nerve 41, 416–419 (2010). 114.Gibbons, C. H., Vernino, S. A. Freeman, R. Combined immunomodulatory therapy in autoimmune autonomic ganglionopathy. Arch. Neurol. 65, 213–217 (2008). 115.Imrich, R., Vernino, S., Eldadah, B. A., Holmes, C. Goldstein, D. S. Autoimmune autonomic ganglionopathy: treatment by plasma exchanges and rituximab. Clin.Auton. Res. 19, 259–262 (2009). 116.Iodice, V. et al. Efficacy of immunotherapy in seropositive and seronegative putative autoimmune autonomic ganglionopathy. Neurology 72, 2002–2008 (2009). 117.Sandroni, P . et al. Idiopathic autonomic neuropathy: comparison of cases seropositive and seronegative for ganglionic acetylcholine receptor antibody. Arch. Neurol. 61, 44–48 (2004). 118.Lucchinetti, C. F., Kimmel, D. W. Lennon, V. A. Paraneoplastic and oncologic profiles of patients seropositive for type 1 antineuronal nuclear autoantibodies. Neurology 50, 652–657 (1998). 119.Winkler, A. S., Dean, A., Hu, M., Gregson, N. Chaudhuri, K. R. Phenotypic and neuropathologic heterogeneity of anti-Hu antibody-related paraneoplastic syndrome presenting with progressive dysautonomia: report of two cases. Clin.Auton. Res. 11, 115–118 (2001). 120.Chinn, J. S. Schuffler, M. D. Paraneoplastic visceral neuropathy as a cause of severe gastrointestinal motor dysfunction. Gastroenterology 95, 1279–1286 (1988). 121.De Giorgio, R. et al. Inflammatory neuropathies of the enteric nervous system. Gastroenterology 126, 1872–1883 (2004). 122.Colan, R. V., Snead, O. C. 3rd , Oh, S. J. Kashlan, M. B. Acute autonomic and sensory neuropathy. Ann. Neurol. 8, 441–444 (1980). 123.Koike, H. et al. Clinicopathological features of acute autonomic and sensory neuropathy. Brain 133, 2881–2896 (2010). 124.Pavlakis, P . P . et al. Peripheral neuropathies in Sjögren syndrome: a new reappraisal. J. Neurol. Neurosurg. Psychiatry 82, 798–802 (2011). 125.Dawson, L., Tobin, A., Smith, P . Gordon, T. Antimuscarinic antibodies in Sjögren’s syndrome: where are we, and where are we going? Arthritis Rheum. 52, 2984–2995 (2005). 126.Nakamura, Y. et al. High prevalence of autoantibodies to muscarinic‑3 acetylcholine receptor in patients with juvenile-onset Sjögren syndrome. Ann. Rheum. Dis. 67, 136–137 (2008). 127.Park, K., Park, S. Jackson, M. W. The inhibitory effects of antimuscarinic autoantibodies in the sera of primary Sjogren syndrome patients on the gastrointestinal motility. Mol. Immunol. 56, 583–587 (2013). 128.Kondo, T. et al. Autoimmune autonomic ganglionopathy with Sjögren’s syndrome: significance of ganglionic acetylcholine receptor antibody and therapeutic approach. Auton. Neurosci. 146, 33–35 (2009). 129.Tesfaye, S. et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care 33, 2285–2293 (2010). 130.Ziegler, D., Gries, F. A., Spuler, M. Lessmann, F. The epidemiology of diabetic neuropathy. Diabetic Cardiovascular Autonomic Neuropathy Multicenter Study Group. J. Diabetes Complications 6, 49–57 (1992). 131.Low, P . A. et al. Autonomic symptoms and diabetic neuropathy: a population-based study. Diabetes Care 27, 2942–2947 (2004). 132.England, J. D. et al. Practice Parameter: evaluation of distal symmetric polyneuropathy: role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, American Association of Neuromuscular and Electrodiagnostic Medicine, and American Academy of Physical Medicine and Rehabilitation. Neurology 72, 177–184 (2009). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 12. 12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol 133.Vinik, A. I. Ziegler, D. Diabetic cardiovascular autonomic neuropathy. Circulation 115, 387–397 (2007). 134.Maleki, D. et al. Gastrointestinal tract symptoms among persons with diabetes mellitus in the community. Arch. Intern. Med. 160, 2808–2816 (2000). 135.Lysy, J., Israeli, E. Goldin, E. The prevalence of chronic diarrhea among diabetic patients. Am. J. Gastroenterol. 94, 2165–2170 (1999). 136.Schiller, L. R. et al. Pathogenesis of fecal incontinence in diabetes mellitus: evidence for internal‑anal‑sphincter dysfunction. N. Engl. J. Med. 307, 1666–1671 (1982). 137.Bradley, W. E. Diagnosis of urinary bladder dysfunction in diabetes mellitus. Ann. Intern. Med. 92, 323–326 (1980). 138.Kaplan, S. A., Te, A. E. Blaivas, J. G. Urodynamic findings in patients with diabetic cystopathy. J. Urol. 153, 342–344 (1995). 139.Bacon, C. G. et al. Association of type and duration of diabetes with erectile dysfunction in a large cohort of men. Diabetes Care 25, 1458–1463 (2002). 140.Malavige, L. S. Levy, J. C. Erectile dysfunction in diabetes mellitus. J. Sex. Med. 6, 1232–1247 (2009). 141. Watkins, P . J. Facial sweating after food: a new sign of diabetic autonomic neuropathy. Br. Med. J. 1, 583–587 (1973). 142.Gibbons, C. H. Freeman, R. Treatment- induced diabetic neuropathy: a reversible painful autonomic neuropathy. Ann. Neurol. 67, 534–541 (2010). 143.Wang, A. K., Fealey, R. D., Gehrking, T. L. Low, P . A. Patterns of neuropathy and autonomic failure in patients with amyloidosis. Mayo Clin. Proc. 83, 1226–1230 (2008). 144.Cappellari, M. et al. Variable presentations of TTR-related familial amyloid polyneuropathy in seventeen patients. J. Peripher. Nerv. Syst. 16, 119–129 (2011). 145.Rajkumar, S. V., Gertz, M. A. Kyle, R. A. Prognosis of patients with primary systemic amyloidosis who present with dominant neuropathy. Am. J. Med. 104, 232–237 (1998). 146.Gertz, M. A. et al. Stem cell transplantation for the management of primary systemic amyloidosis. Am. J. Med. 113, 549–555 (2002). 147.Planté-Bordeneuve, V. Said, G. Familial amyloid polyneuropathy. Lancet Neurol. 10, 1086–1097 (2011). 148.Said, G. Planté-Bordeneuve, V. TTR-familial amyloid polyneuropathy—neurological aspects. Amyloid 19 (Suppl. 1), 25–27 (2012). 149.Arruda-Olson, A. M. et al. Genotype, echocardiography, and survival in familial transthyretin amyloidosis. Amyloid 20, 263–268 (2013). 150.Yamashita, T. et al. Long-term survival after liver transplantation in patients with familial amyloid polyneuropathy. Neurology 78, 637–643 (2012). 151.Coelho, T. et al. Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology 79, 785–792 (2012). 152.Robinson-Papp, J., Sharma, S., Simpson, D. M. Morgello, S. Autonomic dysfunction is common in HIV and associated with distal symmetric polyneuropathy. J. Neurovirol. 19, 172–180 (2013). 153.Dutsch, M. et al. Small fiber dysfunction predominates in Fabry neuropathy. J. Clin. Neurophysiol. 19, 575–586 (2002). 154.Dib-Hajj, S. D., Cummins, T. R., Black, J. A. Waxman, S. G. From genes to pain: Nav 1.7 and human pain disorders. Trends Neurosci. 30, 555–563 (2007). 155.Faber, C. G. et al. Gain of function Nav 1.7 mutations in idiopathic small fiber neuropathy. Ann. Neurol. 71, 26–39 (2012). 156.Rush, A. M. et al. A single sodium channel mutation produces hyper- or hypoexcitability in different types of neurons. Proc. Natl Acad. Sci. USA 103, 8245–8250 (2006). 157.Han, C. et al. Functional profiles of SCN9A variants in dorsal root ganglion neurons and superior cervical ganglion neurons correlate with autonomic symptoms in small fibre neuropathy. Brain 135, 2613–2628 (2012). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved