Você está na página 1de 13

J Neurol (2009) 256:320–332

DOI 10.1007/s00415-009-0144-7 REVIEW

Joseph Ferrara Acquired hepatocerebral degeneration


Joseph Jankovic

Received: 31 August 2008 ■ Abstract Cirrhosis and its other movement disorders. This
Accepted: 20 October 2008 co-morbidities may cause a variety article reviews the clinical spec-
Published online: 17 February 2009 of neurological complications, the trum, pathophysiology, neuroimag-
most common being bouts of toxic ing features and differential diag-
J. Ferrara, MD · J. Jankovic, MD (쾷)
Dept. of Neurology metabolic encephalopathy. A pro- nosis of AHD along with emerging
Baylor College of Medicine portion of patients with chronic treatment options.
6550 Fannin, Suite 1801 liver disease develop acquired
Houston, Texas 77030, USA hepatocerebral degeneration ■ Key words acquired hepatocere-
Tel.: 713/798-5998
Fax: 713/798-6808 (AHD), a chronic progressive bral degeneration · portosystemic
E-Mail: josephj@bcm.tmc.edu neurological syndrome character- shunt · cirrhosis · movement
Web: www.jankovic.org ized by parkinsonism, ataxia and disorder

lowing evidence that, as with Wilson disease, metal in-


Introduction toxication may contribute to its pathogenesis [25, 29, 41,
68, 69, 109, 115].
Acquired hepatocerebral degeneration (AHD) is an of- The exact prevalence of AHD is not known, but cir-
ten debilitating neurological disorder characterized by rhosis affects approximately 5.5 million Americans [4],
parkinsonism, ataxia and other movement disorders, and a broad spectrum of movement disorders occur in
which may accompany various forms of advanced liver 20 to 90 % (8, 56, 70, 121, 138, 139) of the nearly 17,000
disease, especially those characterized by portosystemic [152] patients who are currently awaiting liver trans-
shunting. plantation. Including patients with less severe degrees of
AHD was first reported by W. van Woerkem in 1914 cirrhosis, the prevalence of AHD may be more in the
[153], two years following S. A. Kinnear Wilson’s seminal range of 2 % [114].
manuscript on the hereditary form of hepatolenticular Symptoms of AHD usually begin in adulthood, often
degeneration that now bears his name [159]. Over the in the fifth to sixth decade of life [8, 67, 114, 154]; how-
past century, progress in understanding and treating ever, AHD occasionally affects children [51, 108], so the
Wilson disease has been considerable. We now recognize age at onset alone cannot distinguish AHD from Wilson
that Wilson disease is a hereditary disorder of biliary disease (Table 1).
copper excretion and insights into its etiopathogenesis AHD arises insidiously or subacutely after several
have yielded effective treatments for this once devastat- weeks [2] to decades [67] of hepatic dysfunction. While
ing neurological condition, although failure to diagnose generally progressive, its course is highly variable: grad-
the disease is still the principal cause of death [155]. ual deterioration may be punctuated by years of clinical
AHD, by contrast, lacks proven medical therapy and has stability [8, 30, 154], and symptoms may not parallel
received relatively little attention despite its greater other features of liver disease including hepatic enceph-
prevalence. Indeed, AHD went virtually unrecognized alopathy [8, 55]. Survival following the onset of AHD
until Victor, Adams and Cole’s landmark review in 1965 ranges from several weeks [154] to an excess of 30 years
JON 3144

[154], and even today the disorder remains under-diag- [52]. Most patients eventually die from the systemic
nosed. Recently, however, interest in AHD has grown fol- complications of cirrhotic liver failure including infec-
321

Table 1 Features distinguishing AHD from Wilson disease

Acquired hepatocerebral degeneration Wilson disease

Typical age at onset Middle age and beyond Childhood, adolescence or young adulthood
Family history Typically unrevealing Consistent with autosomal recessive inheritance
Kayser-Fleisher rings Not present on slit-lamp examination Almost always accompany neurological disease
Urinary copper excretion Normal except in cases of severe cholestasis Elevated in symptomatic patients
Serum ceruloplasmin Normal or reduced in proportion to albumin Usually reduced, rarely in excess of 30 mg/dL
Typical MRI findings Increased T1 signal in the pallidum and nigra Increased T2 signal in the basal ganglia, white matter, thalamus or
brainstem
Response to copper-lowering therapies Not formally studied but theoretically poor Often excellent but not universally successful

tion, coagulopathy, hepatorenal syndrome, hepatocellu- universal feature of AHD, even in patients with promi-
lar carcinoma, and hepatic coma [114, 154]. In one study nent motor difficulties [30, 154]. A recent study, which
of 16 patients with AHD, after a median follow-up of 29 utilized formal psychometric testing, showed that im-
months, 75 % of patients had died prior to liver trans- pairments in visuo-spatial attention best separate pa-
plantation, 12.5 % underwent liver transplantation, and tients with AHD from normal controls; however, the
12.5 % were awaiting surgery [114]. authors found little difference in cognitive parameters
between patients with AHD versus those with cirrhosis
without a movement disorder [143]. Accordingly, in pa-
Illustrative case tients with cirrhosis, movement and cognitive dysfunc-
tion may have distinct causes. In contrast to hepatic en-
A 65-year-old woman presented with a six-month his- cephalopathy, however, movement disorders associated
tory of subacute-onset, severe, limb-flinging movements with AHD are generally not accompanied by a reduced
accompanied by instability, slurred speech and difficulty level of consciousness, nor responsive to ammonia-low-
swallowing. The patient’s medical history was notable ering therapies AHD may give rise to various types of
for cryptogenic cirrhosis, Child-Pugh Score 6 (Class A), abnormal movement including parkinsonism, ataxia,
with portal hypertension, esophageal varices, and a prior dystonia, chorea and orobuccolingual stereotypy, phe-
bout of mild hepatic encephalopathy. nomenologically similar to tardive dyskinesias. In pro-
Her neurological examination revealed impaired ori- spective studies of patients attending liver clinics, par-
entation and concentration. She had generalized chorea, kinsonism and action tremor are most prevalent [8, 56,
limb and gait ataxia, cerebellar dysarthria, and asterixis. 121, 138, 139], and about one-fifth of patients hospital-
Magnetic resonance imaging (MRI) showed homoge- ized as potential candidates for liver transplant have
nously increased T1 signal within the globus pallidus definite parkinsonism (mean United Parkinson Disease
and subthalamic region, while T2-weighted and gado- Rating Scale Part 3 score of 38) [8]. In case series derived
linium enhanced sequences were normal. The patient from neurology clinics, patients with ataxia, chorea and
had no relevant family history, had not used medications orobuccolingual dyskinesias are as commonly reported,
known to precipitate dyskinesias, and laboratory testing likely reflecting referral patterns among hepatologists
excluded Wilson disease, Huntington disease, dentato- [30, 55, 67, 122, 148, 154].
rubropallidoluysian atrophy, neuroacanthocytosis, an- AHD-related parkinsonism, like idiopathic Parkin-
tiphospholipid antibody syndrome, systemic lupus ery- son disease, is characterized by tremor, rigidity, bradyki-
thematosus and endocrinopathies. nesia, postural instability and shuffling gait [8]; however,
Treatment with lactulose and metronidozole yielded in most cases other clinical features readily distinguish
no benefit. Tetrabenazine lessened the patient’s chorea the two disorders. In contrast to idiopathic Parkinson
but dose titration was limited by sedation. disease, AHD-related parkinsonism is often [8], though
not always [55, 134], bilateral and symmetrical at disease
onset. Compared to Parkinson disease, AHD progresses
Clinical manifestations more rapidly, with peak severity occurring on average
within 7 months of symptom onset [8] and postural in-
AHD causes a variety of cognitive and behavioral stability or cognitive impairment, early in the course of
changes including prominent apathy, bradyphrenia, and disease [8, 67, 134]. Some patients with AHD may have
deficits in attention, although language function, mem- resting tremor [55, 71], but action tremor is far more
ory and praxis are usually well preserved [30, 67, 71, 114, common [8, 30, 41, 154]. The diagnosis of AHD is also
154]. Overt cognitive dysfunction is a frequent but not suggested by the presence of additional neurological ab-
322

normalities not seen in Parkinson disease, such as cho- lular failure alone may contribute to AHD [124], AHD in
rea and orobuccolingual dyskinesias, ataxia or myelopa- the absence of portosystemic shunting is unusual.
thy [41, 118, 122, 142]. Finally,AHD-related parkinsonism Portosystemic shunting may predispose patients to
and Parkinson disease differ in their response to ther- AHD by allowing neuroactive substances contained
apy; although levodopa benefits some patients with within the portal circulation to circumvent hepatic me-
AHD [8, 55, 67, 83, 122], the response is far less consistent tabolism or biliary elimination through a portacaval
than in Parkinson disease [2, 68, 134, 164]. AHD-related anastomosis, and then enter the brain via the systemic
parkinsonism, however, may improve following liver circulation. While a specific neurotoxin has not been
transplantation [77, 134, 139, 142]. identified, ammonia, which plays a role in hepatic en-
Ataxia is a ubiquitous feature of AHD in most series, cephalopathy, and aromatic amino acids, which may dis-
but alcoholic cerebellar degeneration is a potential con- rupt dopaminergic pathways, are candidates [55]. Over
founder for patients with a history of alcohol-related the past two decades, research into the pathogenesis of
cirrhosis. In this population, features supporting the di- AHD has increasingly focused on the role of heavy met-
agnosis of AHD include: appendicular as opposed to als, particularly manganese.
truncal ataxia, a long latency between alcohol cessation Couper first described the neurotoxic properties of
and symptom onset, and imaging findings or other neu- manganese in 1837, and subsequent studies of manga-
rological deficits suggestive of AHD. nese ore-workers in Africa [123], Asia [45–48] and the
AHD rarely causes a metabolic myelopathy resem- Americas [15, 91] have defined the clinical features of
bling subacute combined degeneration [14, 114], but intoxication, now termed “manganism” [106]. Improved
otherwise does not produce pyramidal tract signs or environmental health regulations have reduced the inci-
sensory loss [8, 30, 67]. Other neurological deficits, such dence of manganism in miners, but historically 1 to 4
as myoclonus and asterixis, have been tentatively as- (and perhaps as much as 25) % of that population devel-
cribed to AHD but may be attributable to co-morbid oped symptoms [106]. Recently, manganism unexpect-
hepatic encephalopathy. edly re-emerged as an important medical topic because
The most characteristic motor phenotype of AHD is of scientific and legal debate surrounding a potential
cranial dyskinesia, which resembles a medication-in- connection between welding, manganese, and neuro-
duced tardive stereotypy and is often intermixed with logical disease [54]. Also, recent reports of outbreaks of
dystonic spasms that produce forced grimacing, jaw manganism in persons exposed to recreational drugs
opening, blepharospasm and ocular deviations [8, 29, tainted with manganese have drawn more attention to
30]. Dysarthria is a common feature of AHD and may the role of this metal in neurological disorders [21,
reflect combinations of slow, high-pitched speech asso- 141].
ciated with cranial dystonia [30], scanning speech repre- When reviewing evidence that links manganese to
senting cerebellar dysfunction, and hypophonia from AHD, it is important to note the similarities and differ-
parkinsonism. Cranial dyskinesias may be part of more ences in clinical phenotype between AHD and occupa-
generalized hyperkinetic movement disorder, particu- tional manganism. Both disorders may cause cranial
larly chorea [30, 55, 107, 148] or dystonia [67, 158]. dystonia and a form of atypical parkinsonism that is
subacute in onset, symmetrical, poorly responsive to do-
pamine replacement therapy and characterized by ac-
Pathogenesis tion tremor, and an early impairment in postural reflexes
[54]. When compared to occupational manganism, AHD
AHD may accompany any form of chronic liver disease is much more likely to cause ataxia and chorea, but less
associated with portosystemic shunting and has been likely to produce limb dystonia, psychosis or compulsive
described in the setting of viral hepatitis, alcoholic cir- behaviors. So-called “manganese madness,” the initial
rhosis, alpha-1 antitrypsin deficiency, nonalcoholic ste- period of psychomotor excitement that often precedes
atohepatitis, autoimmune hepatitis, primary and sec- motor symptoms in miners, has not been observed in
ondary biliary cirrhosis [67, 114], biliary atresia [51], patients with AHD. The speech difficulties and tremor of
Byler’s disease [108] and cryptogenic forms of cirrhosis occupational manganism may lessen with time, but gait
[8]. Occasionally, AHD occurs in patients who have por- dysfunction rarely improves, and neurological deficits
tosystemic shunts without underlying hepatocellular do not wax and wane [15]. In contrast, AHD may be in-
disease, such as those with hereditary hemorrhagic te- terrupted by spontaneous remissions and sudden re-
langiectasias, surgeries or portal vein thrombosis [9, 30, lapses [55, 148].
93, 100, 127, 144, 164]. The report of a patient who devel- A relationship between liver disease and manganese
oped reversible levodopa-unresponsive parkinsonism became apparent when, in 1989, two research groups re-
several weeks following an episode of fulminant hepati- ported that cirrhotic patients have unusual findings on
tis-E infection suggests that the disorder may arise as a brain MRI – specifically, homogenous pallidal hyperin-
consequence of acute liver diseases [2]. While hepatocel- tensities on T1-weighted sequences with normal T2-
323

weighted sequences [25, 79]. This abnormal MRI signal, ligands reveals a variable reduction of D2 receptor den-
suggestive of deposition of a paramagnetic substance sity, which indicates that manganese disturbs postsyn-
within brain tissue, is identical to MRI changes seen in aptic nigrostriatal pathways [62, 132]. Fluorodeoxyglu-
nonhuman primates intoxicated with manganese [99, cose PET, a technique that measures regional metabolic
133], patients with occupational manganism [41], and activity, shows reduced uptake in the globus pallidum
persons exposed to total parenteral nutrition [97] or in- [132], as one might expect considering the pallidal le-
travenous drugs contaminated with manganese [21, sions on MRI. Likewise, autopsy studies of patients with
141]. Subsequent studies have confirmed that patients occupational manganism confirm that neurodegenera-
with advanced liver disease have excessive amounts of tion occurs predominantly in the globus pallidum – es-
manganese in their blood [8] and have shown that blood pecially the medial segment – followed by the striatum
levels correlated with imaging abnormalities [11, 69, 100, and substantia nigra par reticularis [163]. In two pa-
138]. Cerebrospinal fluid [8, 59] and brain tissue manga- tients with action tremor and parkinsonism from AHD,
nese concentrations [49, 68, 84, 115, 124] have been found functional neuroimaging using dopamine transporter
to be elevated, demonstrating that the metal can traverse ligands revealed normal uptake in the striatum, indicat-
the blood-brain barrier of patients with chronic liver ing integrity of the presynaptic dopaminergic pathway
disease. Indeed, patients who expire from liver failure similar to manganism [64, 109].
have pallidal manganese levels that are between 3- and In patients with AHD, neuropathological examina-
9-fold higher than in controls [49, 68, 84, 115, 124], while tion often shows diffuse brain atrophy and regions of
the brain concentrations of other common metals are translucent discoloration within the cortex and basal
normal or only modestly elevated [68, 84, 116]. ganglia [30, 154]. On light microscopy, small vacuoles
Most [33, 53, 70, 73, 121, 156], though not all [138, are present (polymicrocavitation) corresponding to the
146], studies indicate that increased pallidal signal, and translucent lesions seen on gross inspection. Vacuolar
thus brain manganese levels, parallel portosystemic change originates in the deep cortical layers and basal
shunting. Such a correlation is not surprising consider- ganglia. Within the cortex, polymicrocavitation is often
ing the liver’s role in the metabolism of manganese and most severe in the posterior frontal, parietal and occipi-
other metals. In healthy persons, the gastrointestinal tal areas [30], but the temporal lobes may also be affected
tract provides the most important means of manganese [66, 137]. In some patients, polymicrocavitation is great-
uptake and elimination. Typically, dietary manganese est adjacent to sulci [148, 154], while in others, it is more
intake exceeds physiological requirements in excess of prominent at the gyral summits [30, 66, 137, 144]. Within
95 %, and most manganese that enters the portal blood the basal ganglia, the superior pole of the putamen, cau-
is promptly excreted into bile before reaching the sys- date heads, pallidum and subthalamic nuclei are most
temic circulation [17, 24, 54]. In patients with liver dis- involved [36, 66, 154]. Characteristically, polymicrocavi-
ease, however, normal manganese excretion is impaired tation extends into adjacent white matter tracts where it
and the metal enters the systemic circulation and brain. appears to displace rather than destroy surrounding
The transfer of manganese from the portal to systemic neurons. In some cases, however, white matter vacu-
circulation appears to be the critical pathogenic event, olization is accompanied by myelinolysis [66, 78, 137].
as patients who have obstructive jaundice without por- Polymicrocavitation does not involve the cerebellum,
tosystemic shunts are less prone to increased pallidal and ataxia in patients with AHD may be related to de-
signal [6]. Despite strong evidence that portosystemic generation of the dentate nuclei, Purkinje cell loss, and
shunting results in brain manganese accumulation, evi- Bergmann’s gliosis. Though pathologically striking, the
dence directly linking manganism with neurological relevance of polymicrocavitation is unsettled because of
symptoms in this population is scarce. Furthermore, questions regarding clinicopathological correlation. For
manganese blood levels, correlate only with pallidal sig- example, most patients with AHD lack weakness and
nal and do not predict the presence of neurological dis- sensory loss despite profound vacuolization of the fron-
ease [138]. toparietal cortices, while some patients with clinically
In contrast to patients with idiopathic Parkinson dis- typical AHD lack polymicrocavitation entirely [154]. In-
ease, those with occupational manganism have normal deed, the only consistent neuropathological feature of
fluorodopa positron emission tomography (PET) imag- AHD is the presence Alzheimer type II glia, reactive as-
ing, suggesting that the metal does not cause loss of pre- trocytes exhibiting large pale nuclei with one or more
synaptic nigrostriatal dopaminergic nerve terminals small basophilic nucleoli (Fig. 1). Alzheimer type II glia
[45, 132, 161]. However, recent work using 11-C-raclo- are abundant in, but not restricted to, regions of vacu-
pride/amphetamine PET in a non-human primate model olization [68, 154], and although universally present in
of manganism showed impaired striatal dopamine re- patients with AHD, they are not pathognomonic for this
lease in the absence of dopaminergic nerve terminal disorder. Alzheimer type II glia also arise in patients
loss, leaving open the possibility of more subtle presyn- with uncomplicated cirrhosis, various metabolic en-
aptic dysfunction [38]. PET using dopamine receptor cephalopathies, Wilson disease, and curiously, in both a
324

within the basal ganglia. Areas of manganese accumula-


tion, however, do not reflect the brain concentration of
transferrin receptors [27] and neuronal [5] as well as as-
trocytic [26] transport may facilitate redistribution. It is
also unclear how the manganese causes neural damage.
Some studies have found that the metal accumulates in
high concentrations within the mitochondria of glial
cells and that symptoms may be the result of disrupted
energy metabolism [101], but data in this arena remain
discordant [39, 57].

Diagnosis
AHD has protean manifestations and may mimic a broad
range of diseases. Even AHD’s most characteristic pre-
Fig. 1 Photomicrograph (hematoxylin and eosin staining) of the cerebral hemi- sentation, craniofacial dyskinesias, has an extensive dif-
sphere, centrum semiovale from a patient with liver failure showing numerous ferential diagnosis (Table 2) [28, 35, 76]. In patients who
Alzheimer type 2 astrocytes (arrows). Image is courtesy of Dr. Adekunle Adesina present with cognitive impairment, parkinsonism or
ataxia, clinicians might suspect AHD only when there is
clinical evidence of liver disease or signs of AHD on
primate and rodent model of manganism [42, 103, 113]. brain MRI. Even in patients with established liver dis-
Alzheimer type II glia, however, have not yet been re- ease, the diagnosis of AHD remains challenging because
ported in humans with occupational manganism, and there are multiple potential associations between he-
brain polymicrocavitation also has not been observed in patic and neurological disease. Apart from AHD, other
manganese ore workers [113]. neurological diseases to consider in persons with cir-
Although autopsy and PET studies of individuals rhosis include hepatic (toxic-metabolic) encephalopa-
with manganism have localized the predominant sites of thy, central pontine myelinolysis [74], alcoholic cerebel-
neurotoxicity to the globus pallidum, it is not clear why lar degeneration [10] and Wernicke’s encephalopathy
manganese preferentially accumulates in this brain re- [72].
gion. Like iron, manganese appears to transverse the Sporadic case reports suggest that hereditary hemo-
blood-brain barrier through receptor-mediated endo- chromatosis, which is known to cause cirrhosis, may
cytosis in complex with transferrin and via the divalent also precipitate various movement disorders [23]; how-
metal transporter 1, though other mechanisms likely ever, the connection remains unproven [126]. An asso-
contribute [32]. One might speculate that shared metal ciation between hereditary hemochromatosis and
transport systems explain why various metals, including movement disorders seems plausible given the fact that
iron, copper and manganese, accumulate predominantly abnormal regulation of brain iron homeostasis is an es-

Table 2 Differential diagnosis of orobuccolingual dyskinesias

Drug-induced 1. Acute forms due to dopaminergic drugs, stimulants, oral contraceptives, anabolic steroids, anticholingerics, anticonvulsants
and antimalarials
2. Tardive forms due to dopamine receptor-blocking drugs
Hereditary Huntington disease, neuroacanthocytosis, Lesch-Nyhan disease, Wilson disease, neurodegeneration with brain iron accumula-
tion, dentatorubropallidoluysian atrophy, Machado-Joseph disease, benign familial chorea, porphyria, various inborn errors of
metabolism including mitochondrial disorders, GM1 gangliosidosis, neuronal ceroid lipofuscinoses and others
Toxin-induced Carbon monoxide, manganese, mercury, thallium, toluene, kernicterus
Metabolic Hyperthyroidism, hyper- or hypoparathyroidism, striopallidodentate calcinosis, hyperglycemia, nutritional deficiencies,
electrolyte imbalances, chorea gravidarum, acquired hepatocerebral degeneration
Immune-mediated Sydenham disease, antiphospholipid antibody syndrome, systemic lupus erythematosus, post-vaccinal chorea, paraneoplastic
disorders such as anti-NMDA receptor encephalitis [50]
Cerebrovascular or hematological Basal ganglia or brainstem stroke or hemorrhage, anoxia, polycythemia vera, arteriovenous malformation, vasculitides, cerebral
palsy
Other Idiopathic and secondary craniocervical dystonias, tic disorders (Gilles de la Tourette syndrome or secondary tourettism), spon-
taneous buccolingual dyskinesias of the elderly, edentulous dyskinesias, stereotyped movements in schizophrenia, encephalitis
lethargica, oculomasticatory myorhythmia from Whipple’s disease, facial myoclonus of central origin, and other orolingual
rhythmical movements [135]
325

strate spinal cord dysfunction, usually at the thoracic


level [14, 80].

Treatment
Little has been published concerning the medical man-
agement of AHD, and available data are derived from
case-reports or small retrospective case series. Accord-
ingly, treatment recommendations for AHD, in terms of
disease prevention, modification, and symptomatic
therapies, are tenuous pending well-designed prospec-
tive studies.
AHD-related chorea and orobuccolingual dyskine-
sias often respond to dopamine receptor antagonist
drugs; however, these medications introduce the risk of
an irreversible tardive drug syndrome, and their long-
term safety in patients with AHD is uncertain. Further-
Fig. 2 MRI showing increased T1 signal within the pallidal nuclei of a patient with more, phenotypic similarities between AHD and tardive
parkinsonism associated with AHD. Image is courtesy of Dr. Jeffrey Guptill stereotypy may encumber monitoring for this complica-
tion. Tetrabenazine, a presynaptic dopamine-depleting
drug [61], is an alternative medication which is preferred
tablished cause of several movement disorders, now by some clinicians [107] as it poses no risk of a tardive
known collectively as neurodegeneration with brain syndrome and is effective for a broad range of hyperki-
iron accumulation (NBIA) [75, 87]. However, unlike pa- netic movement disorders [60]. Tetrabenazine has only
tients with NBIA, those with hereditary hemochromato- recently been approved for use in the United States, and
sis have not been shown to have basal ganglia iron like dopamine receptor antagonists, can cause parkin-
deposition on autopsy. Furthermore, while hereditary sonism, sedation, mood changes and other abnormali-
hemochromatosis is a relatively common disorder, very ties, but in contrast to the dopamine receptor blocking
few patients with movement disorders have been re- drugs (neuroleptics), the adverse effects of tetrabenazine
ported, and these patients are heterogeneous in terms of are usually dose-related and reversible.
their clinical, imaging and genetic findings [126]. For patients with parkinsonism related to AHD, in-
Although rare, Wilson disease is unquestionably the formation regarding the utility of dopamine replace-
most important diagnosis to exclude because it is fatal, ment therapy is conflicting. Some patients exhibit a sub-
yet is reversible when treated with copper lowering ther- stantive response to levodopa and dopamine agonists,
apies such as zinc, chelating agents and tetrathiomolyb- approaching the effect seen in idiopathic Parkinson dis-
date (Table 1). ease [8, 67, 83, 94], while in others, symptoms are recal-
Patients with AHD develop a variety of laboratory citrant to therapy [2, 68, 109, 134, 164]. In view of the
and neuroimaging abnormalities that are typical of cir- possible relationship between AHD and manganese, it is
rhotic liver disease; however, it is unclear which of these, notable that, contrary to initial reports [18, 89, 125], do-
if any, are related to the neurological phenotype. The pamine replacement therapy is minimally beneficial for
most characteristic finding in this population is in- occupational manganism [15, 48, 82]. Beta-blockers and
creased T1 signal on MRI within the pallidal nuclei, anticholinergics also have yielded little improvement in
sometimes extending into adjacent basal ganglia struc- patients with tremor [109].
tures and the thalami [95] (Fig. 2). T2-weighted and Thus far, no medications appear to retard the pro-
contrast-enhanced MRI sequences are typically normal, gression of AHD, and development of future neuropro-
but rarely show abnormalities in the pallidi [67, 139, 147, tective therapies is hampered by clinical heterogeneity,
151, 156] or lesions in other structures, such as the uncertainties regarding the pathogenesis and relative
brachium pontis [40, 78]. Several other disorders pro- scarcity of cases thus preventing large clinical trials
duce T1 hyperintensities within the basal ganglia; how- powered to show efficacy of active treatment over pla-
ever, apart from other forms of manganese deposition, cebo. Lactulose, antibiotics and short-term dietary pro-
all are easily differentiated from AHD based upon their tein restriction may minimize co-morbid toxic-meta-
appearance on T2-weighted sequences, contrast en- bolic encephalopathy and may reduce symptoms of
hancement or clinical features (Table 3). In patients with AHD in some patients [117, 154]; however, AHD-related
evidence of myelopathy, MR imaging of the spine is usu- movement disorders seldom respond well to ammonia-
ally normal; however, motor evoked potentials demon- reducing therapies [55, 83, 110, 114, 122, 131, 148].
326

Table 3 Differential diagnosis of T1 hyperintensities in the basal ganglia on MRI

Diagnosis Notes Reference

Acquired hepatocerebral T1 hyperintensities are homogenous and symmetrical. Abnormal signal is located predominantly in the pallidum; lesions do [95]
degeneration (AHD) not enhance
T2 sequences and CT are normal
Manganese intoxication MRI findings are identical to AHD [21, 41, 43,
Causes include: (1) exposure to manganese ore, (2) injection of manganese-tainted total parenteral nutrition or intravenous 65, 97, 141]
drugs, and (3) iron deficiency
Hyperglycemic T1 hyperintensities are typically asymmetrical and are located in the striatum more often than the pallidum; lesions do not [12, 102]
hyperosmolar state enhance
T2 sequences may show low, iso-, or high signal intensity lesions with restricted diffusion in the basal ganglia
CT may be normal or show high-attenuation lesions in the striatum
Wilson disease T1 sequences more often show low or iso-, signal intensity lesions in the basal ganglia, but hyperintensities have been [105, 130, 136]
reported; lesions do not enhance
T2 sequences may show low or high signal intensity lesions with restricted diffusion
FLAIR sequences may show the “giant panda face“ sign
CT may show low-attenuation lesions in the basal ganglia and thalamus
Cortical, subcortical white matter, thalamic, cerebellar and brainstem lesions often accompany basal ganglia disease
Neurodegeneration Four NBIA subtypes include: Pantothenate kinase associated neurodegeneration (PKAN), infantile neuroaxonal dystrophy, [75, 87]
with brain iron neuroferritinopathy and aceruloplasminemia
accumulation (NBIA) NBIA may cause increased T1 signal in the basal ganglia, but changes on T2-weighted imaging are the more common and
prominent finding; lesions do not enhance
T2 and fast spin echo sequences distinguish NBIA subtypes. All 4 subtypes cause T2 hypointensities within the globus pallidus
and substantia nigra. Infantile neuroaxonal dystrophy typically results in no additional abnormalities.
PKAN universally causes an area of central T2 hyperintensity within the globus pallidus – so called “eye of the tiger sign.”
Aceruloplasminemia causes T2 hypointensities within the putamen, caudate and thalamus (all three). Like aceruloplas-
minemia, neuroferritinopathy also may causes additional T2 hypointensities within the putamen, caudate, thalamus,
dentate or cerebral cortex. In addition, neuroferritinopathy sometimes results in either eye of the tiger sign or confluent
cavitary lesions involving the putamen or globus pallius.
Basal ganglia Common causes include parathyroid disease and Fahr syndrome [3, 128]
calcification T1 sequences more often show low or iso-, signal intensity lesions in the basal ganglia, but hyperintensities have been
reported; lesions do not enhance
T2 sequences usually show corresponding iso- or hypointense lesions
CT universally shows high-attenuation lesions and is the preferred imaging modality
Hypoxic-ischemic Prominent T1- hyperintensities of the basal ganglia may be seen following hypoxic-ischemic injury, sometimes with [16, 44, 149, 162]
injury gadolinium enhancement, but T2 lesions and restricted diffusion commonly co-occur
Associated imaging abnormalities are commonly found outside the basal ganglia, e.g., cortical laminar necrosis
In neonates with hypoxic ischemic injury, metabolic disorders such as perinatal hypoglycemia, bilirubin encephalopathy, non-
ketotic hyperglycinemia, and urea-cycle disorders also may cause basal ganglia hyperintensities on T1-weighted imaging
CT may show low-attenuation basal ganglia lesions
Carbon monoxide Basal ganglia T1- hyperintensities are possible, but T1 sequences more often show low signal intensity lesions in the medial [81]
poisoning globus pallidus, which may enhance
T2 sequences usually show corresponding hyperintense lesions and apparent diffusion coefficient maps show restricted
diffusion
CT may show low-attenuation basal ganglia lesions
Langerhans cell T1 pallidal hyperintensities are common and gadolinium enhancing lesions may be present T2 sequences may show low, iso-, [22, 119, 160]
histiocytosis or high signal intensity lesions
co-morbid extra-axial (osseous and dura-based) lesions always accompany parenchymal changes and other brain sites such as
the pineal, pituitary, white matter, dentate and brainstem are commonly involved
CT may show lytic lesions in the skull or skull base
Neurofibromatosis Basal ganglia T1- iso- or hyperintensities are possible, but T2 hyperintensities are more common; some lesions may enhance [92]
type I Lesions may be bilateral but are less homogenous than AHD
Other MRI lesions may co-occur including astrocytomas, optic gliomas, sphenoid dysplasia, and plexiform neurofibromas
CT typically shows normal basal ganglia
Fucosidosis T1 sequences may show pallidal hyperintensities similar to AHD but T2 sequences usually show corresponding basal ganglia [34, 104, 120]
hypointensities
T2 sequences may show prominent white matter abnormalities and thalamic lesions
CT may show low-attenuation lesions within the globus pallidus
Microhemorrhage Bilateral basal ganglia microhemorrhage may result from hypoxia, infections such as Japanese encephalitis, or microangiopa- [1, 58, 88, 129,
of the basal ganglia thies such as hemolytic-uremic syndrome or AIDS-related microangiopathy 140]
T1 and T2 sequences show low, iso-, or high signal intensity lesions depending on the oxidation state of degraded
hemoglobin
Apparent diffusion coefficient maps may show restricted diffusion from concurrent infarction
CT may show low-attenuation basal ganglia lesions from ischemia or high-attenuation lesions from hemorrhage
327

Ethylenediaminetetraacetic acid (EDTA) is a chelator netic resonance spectroscopy (MRS) of the brain is an
of manganese [20], and a proven therapy for some forms alternate means of tracking recovery following liver
of heavy metal poisoning, but its efficacy in AHD is transplantation, since MRS abnormalities resolve in uni-
uncertain. In one report, EDTA yielded unsatisfactory son with clinical improvement and in advance of changes
results in a series of patients with occupational man- on standard MRI [77]. In this population, the changes on
ganism [47]. Furthermore, intravenous EDTA poses po- MRS that correlate with the presence of parkinsonism or
tentially serious risks, including nephrotoxicity, provid- encephalopathy include reduced choline and myoinosi-
ing grounds for caution in its off-label use. A recent tol resonance [139] and increased glutamate/glutamine
report indicates that trientine, an oral chelating agent resonance [96].
used to treat Wilson disease, decreased neurological Recovery following liver transplantation is usually
deficits, MRI abnormalities and manganese blood levels gradual over the course of several months [114, 118, 134],
in a patient with AHD-related parkinsonism [109]. This yet remarkably, some patients improve within hours of
observation requires confirmation, especially since tri- transplant surgery [67, 107, 131]. Such a precipitous re-
entine has not been proven to effectively chelate manga- covery has potentially important implications regarding
nese [7]. the pathophysiology of AHD, as it provides support for
The effects of dietary manganese restriction has yet the role of reversible metabolic factors in AHD. Further-
to be studied for patients with AHD or cirrhotic patients more, it is unlikely that the effects of chronic manganese
who are at risk; however, low manganese intake is pru- accumulation could improve so rapidly. In patients with
dent for such patients on theoretical grounds. Major di- occupational manganism deficits will initially worsen
etary sources of manganese include grains, nuts, tea, and then remain static for decades following removal
fruits, legumes and other vegetables [111, 112]. Since the from the exposure [46]. This observation, in conjunction
proportion of manganese absorbed from the intestine with previously noted phenomenological differences
and retained in tissue varies inversely with iron stores between AHD and occupational manganism have not
[31, 65, 90], iron deficiency anemia may contribute to the been addressed by those who attribute AHD to manga-
progression of AHD and should be corrected when pres- nese intoxication.
ent. If symptoms of AHD result from false neurotrans- The durability of clinical benefit in patients who do
mitters derived from aromatic amino acids, supplement- respond to transplantation remains unknown. Some pa-
ing branched-chain amino acids may be useful, as they tients eventually develop re-emergent neurological defi-
compete with aromatic amino acids to cross the blood- cits either secondary to graft rejection [131] or for other
brain barrier [63]. There is scant evidence supporting reasons [107], some do not respond to transplantation
this therapy for patients with AHD [151], but further [19], and some suffer from peri-operative complications
studies are needed. [80]. Nonetheless, it should be stressed that AHD is not
Since AHD is related to portosystemic anastomoses a contraindication to liver transplantation and in some
[8, 14, 30, 127, 154, 164], deferring placement of endovas- cases may be the only effective therapy.
cular or surgical shunts (used to minimize variceal hem-
orrhage) may limit risk of disease. In selected patients
who suffer neurological deterioration following shunt Conclusion
placement, it may be possible to obstruct an existing
shunt [13, 14, 85, 145]; however, shunt occlusion is poten- AHD remains an underrecognized entity with high
tially risky as it could worsen portal hypertension and morbidity and no proven pharmacological therapies,
precipitate variceal hemorrhage. although liver transplantation is remarkably helpful in
Although long assumed to be irreversible, mounting some patients. More research is needed to better under-
evidence suggests that even long-standing AHD may im- stand the pathogenesis of AHD, define which patients
prove remarkably following liver transplantation [118, are at risk and develop effective strategies to arrest dis-
143], including hyperkinetic movement disorders [107, ease progression and minimize disability.
117, 131, 142], parkinsonism [77, 118, 121, 134, 139, 142],
ataxia [110, 131], myelopathy [98, 114, 150, 157] and cog- ■ Conflict of interest The authors have no association or vested inter-
est in the subject matter or products mentioned in the manuscript.
nitive dysfunction [67, 86, 142]. Blood manganese levels
and MRI changes also improve following restoration of ■ Acknowledgments The authors would like to thank Drs. Adekunle
hepatic function, although normalization of these pa- Adesina and Jeffrey Guptill for providing neuropathological and ra-
rameters is gradual and sometimes incomplete. Mag- diological images, respectively.
328

References
1. Abe T, Kojima K, Shoji H, Tanaka N, 12. Chu K, Kang DW, Kim DE, Park SH, 26. Dringen R, Bishop GM, Koeppe M,
Fujimoto K, Uchida M, Nishimura H, Roh JK (2002) Diffusion-weighted and Dang TN, Robinson SR (2007) The
Hayabuchi N, Norbash AM (1998) gradient echo magnetic resonance pivotal role of astrocytes in the metab-
Japanese encephalitis. J Magn Reson findings of hemichorea-hemiballismus olism of iron in the brain. Neurochem
Imaging 8:755–761 associated with diabetic hyperglyce- Res 32:1884–1890
2. Aggarwal A, Vaidya S, Shah S, Singh J, mia. Arch Neurol 59:448–452 27. Erikson KM, Thompson K, Aschner J,
Desai S, Bhatt M (2006) Reversible 13. Condat B, Dusoleil A, Bernardeau M, Aschner M (2007) Manganese neuro-
Parkinsonism and T1W pallidal hyper- Roche A, Pelletier G, Buffet C (1999) toxicity: a focus on the neonate.
intensities in acute liver failure. Mov Chronic acquired hepatocerebral Pharmacol Ther 113:369–377
Disord 21:1986–1990 degeneration: the role of manganese 28. Fahn S (2007) The Tardive Syndromes:
3. Ahn JH, Yoo CI, Lee CR, Lee JH, Lee H, and treatment by endovascular occlu- Phenomenology, Concepts on
Kim CY, Park JK, Sakai T, Yoon CS, Kim sion of a porto-systemic shunt. Gastro- Pathophysiology and Treatment, and
Y (2003) Calcification mimicking enterol Clin Biol 23:268–270 Other Neruoleptic-Induced Syn-
manganese-induced increased signal 14. Conn HO, Rossle M, Levy L, Glocker dromes. In: Fahn S, Jankovic J (eds)
intensities in T1-weighted MR images FX (2006) Portosystemic myelopathy: The Principles and Practice of Move-
in a patient taking herbal medicine: spastic paraparesis after portosystemic ment Disorders. 1st ed. Philadelphia,
case report. Neurotoxicology 24: shunting. Scand J Gastroenterol 41: PA: Churchill Livingstone, pp 479–518
835–838 619–625 29. Ferrara J, Gupta D, Foster E, Garman K,
4. American Gastroenterological Associ- 15. Cook DG, Fahn S, Brait KA (1974) Stacy M (2008) Extraocular muscle
ation (2001) The Burden of Gastroin- Chronic manganese intoxication. Arch dystonia due to acquired (non-Wilso-
testinal Diseases. at: www.gastro.org/ Neurol 30:59–64 nian) hepatocerebral degeneration.
user-assets/Documents/burden-report. 16. Coskun A, Yikilmaz A, Kumandas S, Mov Disord 23:875–878
pdf Karahan OI, Akcakus M, Manav A 30. Finlayson MH, Superville B (1981)
5. Aschner M, Guilarte TR, Schneider JS, (2005) Hyperintense globus pallidus Distribution of cerebral lesions in
Zheng W (2007) Manganese: recent on T1-weighted MR imaging in acute acquired hepatocerebral degeneration.
advances in understanding its trans- kernicterus: is it common or rare? Eur Brain 104:79–95
port and neurotoxicity. Toxicol Appl Radiol 15:1263–1267 31. Finley JW, Davis CD (1999) Manganese
Pharmacol 221:131–147 17. Cotzias GC, Horiuchi K, Fuenzalida S, deficiency and toxicity: are high or low
6. Bang SJ, Choi SH, Park NH, Shin JW, Mena I (1968) Chronic Manganese dietary amounts of manganese cause
Choy N, Kim SH, Kim Y (2007) High poisoning: clearance of tissue manga- for concern? Biofactors 10:15–24
pallidal T1 signal is rarely observed in nese concentrations with persistence 32. Fitsanakis VA, Piccola G, Marreilha dos
obstructive jaundice, but is frequently of the neurological picture. Neurology Santos AP, Aschner JL, Aschner M
observed in liver cirrhosis. J Occup 18:376–382 (2007) Putative proteins involved in
Health 49:268–272 18. Cotzias GC (1969) Metabolic modifica- manganese transport across the blood-
7. Blanusa M, Varnai VM, Piasek M, tion of some neurologic disorders. brain barrier. Hum Exp Toxicol 26:
Kostial K (2005) Chelators as antidotes JAMA 210:1255–1262 295–302
of metal toxicity: therapeutic and 19. Counsell C, Warlow C (1996) Failure of 33. Fukuzawa T, Matsutani S, Maruyama
experimental aspects. Curr Med Chem presumed hepatic myelopathy to H, Akiike T, Saisho H, Hattori T (2006)
12:2771–2794 improve after liver transplantation. Magnetic resonance images of the
8. Burkhard PR, Delavelle J, Du Pasquier J Neurol Neurosurg Psychiatry 60:590 globus pallidus in patients with idio-
R, Spahr L (2003) Chronic parkin- 20. Crossgrove J, Zheng W (2004) Manga- pathic portal hypertension: a quantita-
sonism associated with cirrhosis: a nese toxicity upon overexposure. NMR tive analysis of the relationship be-
distinct subset of acquired hepatocere- Biomed 17:544–553 tween signal intensity and the grade of
bral degeneration. Arch Neurol 60: 21. de Bie RM, Gladstone RM, Strafella AP, portosystemic shunt. J Gastroenterol
521–528 Ko JH, Lang AE (2007) Manganese- Hepatol 21:902–907
9. Callaghan N, McCoy DG (1967) induced Parkinsonism associated with 34. Galluzzi P, Rufa A, Balestri P, Cerase A,
Acquired chronic hepatocerebral methcathinone (Ephedrone) abuse. Federico A (2001) MR brain imaging
degeneration with splenic vein occlu- Arch Neurol 64:886–889 of fucosidosis type I. AJNR Am J Neu-
sion. J Ir Med Assoc 60:118–120 22. Demaerel P, Van Gool S (2008) Paediat- roradiol 22:777–780
10. Charness ME (1993) Brain lesions in ric neuroradiological aspects of 35. Gershanik O (2004) Drug-induced
alcoholics. Alcohol Clin Exp Res 17: Langerhans cell histiocytosis. Neuro- dyskinesias. In: Jankovic J, Tolosa E
2–11 radiology 50:85–92 (eds) Parkinson’s Disease and Move-
11. Choi Y, Park JK, Park NH, Shin JW, Yoo 23. Demarquay G, Setiey A, Morel Y, Trepo ment Disorders. 4rd ed. Baltimore,
CI, Lee CR, Lee H, Kim HK, Kim SR, C, Chazot G, Broussolle E (2000) Clini- MD: Lippincott Williams & Wilkins,
Jung TH, Park J, Yoon CS, Kim Y (2005) cal report of three patients with hered- pp 365–379
Whole blood and red blood cell man- itary hemochromatosis and movement 36. Graham DI, Adams JH, Caird FI,
ganese reflected signal intensities of disorders. Mov Disord 15:1204–1209 Lawson JW (1970) Acquired hepato-
T1-weighted magnetic resonance 24. Dobson AW, Erikson KM, Aschner M cerebral degeneration: report of an
images better than plasma manganese (2004) Manganese neurotoxicity. Ann atypical case. J Neurol Neurosurg
in liver cirrhotics. J Occup Health 47: N Y Acad Sci 1012:115–128 Psychiatry 33:656–662
68–73 25. Drayer BP, Fram EK, Bird CR, Williams 37. Grossman RI, Yousem DM (2003) Neu-
KD, Keller PJ (1989) Systemic meta- rodegenerative diseases and hydro-
bolic disease and the globus pallidus: cephalus. In: Neuroradiology: The
An MRI approach [abstract]. AJNR Am Requisites. 2nd ed. St. Louis, Mosby,
J Neuroradiol 10:902 pp 391–396
329

38. Guilarte TR, Chen MK, McGlothan JL, 51. Ikeda S, Yamaguchi Y, Sera Y, Ohshiro 65. Kim Y, Park JK, Choi Y, Yoo CI, Lee CR,
Verina T, Wong DF, Zhou Y, Alexander H, Uchino S, Yamashita Y, Ogawa M Lee H, Lee JH, Kim SR, Jeong TH, Yoon
M, Rohde CA, Syversen T, Decamp E, (2000) Manganese deposition in the CS, Park JH (2005) Blood manganese
Koser AJ, Fritz S, Gonczi H, Anderson globus pallidus in patients with biliary concentration is elevated in iron defi-
DW, Schneider JS (2006) Nigrostriatal atresia. Transplantation 69:2339–2343 ciency anemia patients, whereas
dopamine system dysfunction and 52. Inose T (1952) Hepatocerebral degen- globus pallidus signal intensity is
subtle motor deficits in manganese- eration, a special type. J neuropath Exp minimally affected. Neurotoxicology
exposed non-human primates. Exp Neurol 11:401–408 26:107–111
Neurol 202:381–390 53. Inoue E, Hori S, Narumi Y, Fujita M, 66. Kleinschmidt-DeMasters BK, Filley
39. Gunter TE, Gavin CE, Aschner M, Kuriyama K, Kadota T, Kuroda C CM, Rojiani AM (2006) Overlapping
Gunter KK (2006) Speciation of man- (1991) Portal-systemic encephalop- features of extrapontine myelinolysis
ganese in cells and mitochondria: a athy: presence of basal ganglia lesions and acquired chronic (non-Wilsonian)
search for the proximal cause of man- with high signal intensity on MR hepatocerebral degeneration. Acta
ganese neurotoxicity. Neurotoxicology images. Radiology 179:551–555 Neuropathol (Berl) 112:605–616
27:765–776 54. Jankovic J (2005) Searching for a rela- 67. Klos KJ, Ahlskog JE, Josephs KA, Fealey
40. Hanner JS, Li KC, Davis GL (1988) tionship between manganese and RD, Cowl CT, Kumar N (2005) Neuro-
Acquired hepatocerebral degeneration: welding and Parkinson’s disease. logic spectrum of chronic liver failure
MR similarity with Wilson disease. Neurology 64:2021–2028 and basal ganglia T1 hyperintensity on
J Comput Assist Tomogr 12:1076–1077 55. Jog MS, Lang AE (1995) Chronic magnetic resonance imaging: probable
41. Hauser RA, Zesiewicz TA, Rosemurgy acquired hepatocerebral degeneration: manganese neurotoxicity. Arch Neurol
AS, Martinez C, Olanow CW (1994) case reports and new insights. Mov 62:1385–1390
Manganese intoxication and chronic Disord 10:714–722 68. Klos KJ, Ahlskog JE, Kumar N,
liver failure. Ann Neurol 36:871–875 56. Jover R, Compañy L, Gutiérrez A, Cambern S, Butz J, Burritt M, Fealey
42. Hazell AS, Normandin L, Norenberg Lorente M, Zapater P, Poveda MJ, Such RD, Cowl CT, Parisi JE, Josephs KA
MD, Kennedy G, Yi JH (2006) Alz- J, Pascual S, Palazón JM, Carnicer F, (2006) Brain metal concentrations in
heimer type II astrocytic changes fol- Ferrandis F, Pérez-Mateo M (2005) chronic liver failure patients with
lowing sub-acute exposure to manga- Clinical significance of extrapyramidal pallidal T1 MRI hyperintensity.
nese and its prevention by antioxidant signs in patients with cirrhosis. Neurology 67:1984–1989
treatment. Neurosci Lett 396:167–171 J Hepatol 42:659–665 69. Krieger D, Krieger S, Jansen O, Gass P,
43. Herrero Hernandez E, Valentini MC, 57. Kalia K, Jiang W, Zheng W (2008) Theilmann L, Lichtnecker H (1995)
Discalzi G (2002) T1-weighted hyper- Manganese accumulates primarily in Manganese and chronic encephalopa-
intensity in basal ganglia at brain mag- nuclei of cultured brain cells. Neuro- thy. Lancet 346:270–274
netic resonance imaging: are different toxicology 29:466–470 70. Krieger S, Jauss M, Jansen O, Stiehl A,
pathologies sharing a common mecha- 58. Kalita J, Misra UK (2000) Comparison Sauer P, Geissler M, Theilmann L,
nism? Neurotoxicology 23:669–674 of CT scan and MRI findings in the Krieger D (1997) MRI findings in
44. Huang BY, Castillo M (2008) Hypoxic- diagnosis of Japanese encephalitis. chronic hepatic encephalopathy de-
ischemic brain injury: imaging find- J Neurol Sci 174:3–8 pend on portosystemic shunt: results
ings from birth to adulthood. Radio- 59. Katsuragi T, Iseki E, Kosaka K, Koyano of a controlled prospective clinical
graphics 28:417–439 S, Iwabuchi K (1999) Cerebrospinal investigation. J Hepatol 27:121–126
45. Huang CC, Chu NS, Lu CS, Chen RS, fluid manganese concentrations in 71. Krieger S, Jauss M, Jansen O, Theil-
Calne DB (1998) Long-term progres- patients with symmetric pallidal mann L, Geissler M, Krieger D (1996)
sion in chronic manganism: ten years hyperintensities on T1 weighted MRI. Neuropsychiatric profile and hyperin-
of follow-up. Neurology 50:698–700 J Neurol Neurosurg Psychiatry 66: tense globus pallidus on T1-weighted
46. Huang CC, Chu NS, Lu CS, Chen RS, 551–552 magnetic resonance images in liver
Schulzer M, Calne DB (2007) The natu- 60. Kenney C, Hunter C, Jankovic J (2007) cirrhosis. Gastroenterology 111:
ral history of neurological manganism Long-term tolerability of tetra- 147–155
over 18 years. Parkinsonism Relat benazine in the treatment of hyper- 72. Kril JJ, Butterworth RF (1997) Dien-
Disord 13:143–145 kinetic movement disorders. Mov cephalic and cerebellar pathology in
47. Huang CC, Chu NS, Lu CS, Wang JD, Disord 22:193–197 alcoholic and nonalcoholic patients
Tsai JL, Tzeng JL, Wolters EC, Calne DB 61. Kenney C, Jankovic J (2006) Tetra- with end-stage liver disease. Hepatol-
(1989) Chronic manganese intoxica- benazine in the treatment of hyper- ogy 26:837–841
tion. Arch Neurol 46:1104–1106 kinetic movement disorders. Expert 73. Kulisevsky J, Pujol J, Balanzó J, Junqué
48. Huang CC, Lu CS, Chu NS, Hochberg F, Rev Neurother 6:7–17 C, Deus J, Capdevilla A, Villanueva C
Lilienfeld D, Olanow W, Calne DB 62. Kessler KR, Wunderlich G, Hefter H, (1992) Pallidal hyperintensity on mag-
(1993) Progression after chronic man- Seitz RJ (2003) Secondary progressive netic resonance imaging in cirrhotic
ganese exposure. Neurology 43: chronic manganism associated with patients: clinical correlations. Hepatol-
1479–1483 markedly decreased striatal D2 recep- ogy 16:1382–1388
49. Ihara H (1998) High signal intensity of tor density. Mov Disord 18:217–218 74. Kumar S, Fowler M, Gonzalez-Toledo
globus pallidus on T1-weighted MRI in 63. Khanna S, Gopalan S (2007) Role of E, Jaffe SL (2006) Central pontine
liver cirrhosis patients: clinical and branched-chain amino acids in liver myelinolysis, an update. Neurol Res
pathological study. Nippon Igaku disease: the evidence for and against. 28:360–366
Hoshasen Gakkai Zasshi 58:479–488 Curr Opin Clin Nutr Metab Care 10: 75. Kurian MA, Morgan NV, MacPherson
50. Iizuka T, Sakai F, Ide T, Monzen T, 297–303 L, Foster K, Peake D, Gupta R, Philip
Yoshii S, Iigaya M, Suzuki K, Lynch DR, 64. Kim J, Kim JM, Kim YK, Shin JW, Choi SG, Hendriksz C, Morton JE, Kingston
Suzuki N, Hata T, Dalmau J (2008) SH, Kim SE, Kim Y (2007)Dopamine HM, Rosser EM, Wassmer E, Gissen P,
Anti-NMDA receptor encephalitis in transporter SPECT of a liver cirrhotic Maher ER (2008) Phenotypic spectrum
Japan: long-term outcome without with atypical parkinsonism. Ind of neurodegeneration associated with
tumor removal. Neurology 70:504–511 Health 45:497–500 mutations in the PLA2G6 gene
(PLAN). Neurology 70:1623–1629
330

76. Kurlan R, Shoulson I (1988) Differen- 88. Meltzer CC, Wells SW, Becher MW, 100. Nolte W, Wiltfang J, Schindler CG,
tial diagnosis of facial chorea. Adv Flanigan KM, Oyler GA, Lee RR (1998) Unterberg K, Finkenstaedt M, Nied-
Neurol 49:225–237 AIDS-related MR hyperintensity of the mann PD, Hartmann H, Ramadori G
77. Lazeyras F, Spahr L, DuPasquier R, basal ganglia. AJNR Am J Neuroradiol (1998) Bright basal ganglia in T1-
Delavelle J, Burkhard P, Hadengue A, 19:83–89 weighted magnetic resonance images
Hochstrasser D, Mentha G, Giostra E, 89. Mena I, Court J, Fuenzalida S, are frequent in patients with portal
Terrier F, Vingerhoets F (2002) Persis- Papavasiliou PS, Cotzias GC (1970) vein thrombosis without liver cirrho-
tence of mild parkinsonism 4 months Modification of chronic manganese sis and not suggestive of hepatic en-
after liver transplantation in patients poisoning. Treatment with L-dopa or cephalopathy. J Hepatol 29:443–449
with preoperative minimal hepatic 5-0H tryptophane. N Engl J Med 282: 101. Normandin L, Hazell AS (2002) Man-
encephalopathy: a study on neuro- 5–10 ganese neurotoxicity: an update of
radiological and blood manganese 90. Mena I, Horiuchi K, Burke K, Cotzias pathophysiologic mechanisms. Metab
changes. Transpl Int 15:188–195 GC (1969) Chronic manganese poison- Brain Dis 17:375–387
78. Lee J, Lacomis D, Comu S, Jacobsohn J, ing. Individual susceptibility and 102. Oh SH, Lee KY, Im JH, Lee MS (2002)
Kanal E (1998) Acquired hepatocere- absorption of iron. Neurology 19: Chorea associated with non-ketotic
bral degeneration: MR and pathologic 1000–1006 hyperglycemia and hyperintensity
findings. AJNR Am J Neuroradiol 19: 91. Mena I, Marin O, Fuenzalida S, Cotzias basal ganglia lesion on T1-weighted
485–487 GC (1967) Chronic manganese poison- brain MRI study: a meta-analysis of
79. Levy LM, Yang A, Kumar AJ, Hennigar ing: clinical picture and manganese 53 cases including four present cases.
R, Rothstein J, Bryan RN (1989) The turnover. Neurology 17:128–136 J Neurol Sci 200:57–62
brain and hepatic encephalopathy MR 92. Mentzel HJ, Seidel J, Fitzek C, Eichhorn 103. Olanow CW, Good PF, Shinotoh H,
abnormalities (abstract). AJNR Am J A, Vogt S, Reichenbach JR, Zintl F, Hewitt KA, Vingerhoets F, Snow BJ,
Neuroradiol 10:904 Kaiser WA (2005) Pediatric brain MRI Beal MF, Calne DB, Perl DP (1996)
80. Lewis MB, MacQuillan G, Bamford JM, in neurofibromatosis type I. Eur Radiol Manganese intoxication in the rhesus
Howdle PD (2000) Delayed myelo- 15:814–822 monkey: a clinical, imaging, patho-
pathic presentation of the acquired 93. Mizoguchi N, Nishimura Y, Ono H, logic, and biochemical study. Neurol-
hepatocerebral degeneration syn- Sakura N (2001) Manganese elevations ogy 46:492–498
drome. Neurology 54:1011 in blood of children with congenital 104. Oner AY, Cansu A, Akpek S, Serdaro-
81. Lo CP, Chen SY, Lee KW, Chen WL, portosystemic shunts. Eur J Pediatr glu A (2007) Fucosidosis: MRI and
Chen CY, Hsueh CJ, Huang GS (2007) 160:247–250 MRS findings. Pediatr Radiol 37:
Brain injury after acute carbon mon- 94. Morgan MY, Jakobovits AW, James IM, 1050–1052
oxide poisoning: early and late compli- Sherlock S (1980) Successful use of 105. Page RA, Davie CA, MacManus D,
cations. AJR Am J Roentgenol 189: bromocriptine in the treatment of Miszkiel KA, Walshe JM, Miller DH,
W205–W211 chronic hepatic encephalopathy. Lees AJ, Schapira AH (2004) Clinical
82. Lu CS, Huang CC, Chu NS, Caine DB Gastroenterology 78:663–670 correlation of brain MRI and MRS
(1994) Levodopa failure in chronic 95. Morgan MY (1998) Cerebral magnetic abnormalities in patients with Wilson
manganism. Neurology 44:1600–1602 resonance imaging in patients with disease. Neurology 63:638–643
83. Lunzer M, James IM, Weinman J, chronic liver disease. Metab Brain Dis 106. Pal PK, Samii A, Calne DB (1999)
Sherlock S (1974) Treatment of chronic 13:273–290 Manganese neurotoxicity: a review of
hepatic encephalopathy with levodopa. 96. Naegele T, Grodd W, Viebahn R, Seeger clinical features, imaging and pathol-
Gut 15:555–561 U, Klose U, Seitz D, Kaiser S, Mader I, ogy. Neurotoxicology 20:227–238
84. Maeda H, Sato M, Yoshikawa A, Kimura Mayer J, Lauchart W, Gregor M, Voigt K 107. Papapetropoulos S, Singer C (2005)
M, Sonomura T, Terada M, Kishi K (2000) MR imaging and (1)H spectros- Management of the extrapyramidal
(1997) Brain MR imaging in patients copy of brain metabolites in hepatic syndrome in chronic acquired hepa-
with hepatic cirrhosis: relationship encephalopathy: time-course of renor- tocerebral degeneration (CAHD).
between high intensity signal in basal malization after liver transplantation. Mov Disord 20:1088–1089
ganglia on T1-weighted images and Radiology 216:683–691 108. Papapetropoulos S, Tzakis A, Sengun
elemental concentrations in brain. 97. Nagatomo S, Umehara F, Hanada K, C, Reddy C, Boukas K, Zitser J, Singer
Neuroradiology 39:546–550 Nobuhara Y, Takenaga S, Arimura K, C (2008) Case of pediatric acquired
85. Matsumoto S, Mori H, Yoshioka K, Osame M (1999) Manganese intoxica- chronic hepatocerebral degeneration.
Kiyosue H, Komatsu E (1997) Effects of tion during total parenteral nutrition: Pediatr Neurol 38:67–70
portal-systemic shunt embolization on report of two cases and review of the 109. Park HK, Kim SM, Choi CG, Lee MC,
the basal ganglia: MRI. Neuroradiol- literature. Neurol Sci 162:102–105 Chung SJ (2008) Effect of trientine on
ogy 39:326–328 98. Nardone R, Buratti T, Oliviero A, Loch- manganese intoxication in a patient
86. Mattarozzi K, Stracciari A, Vignatelli L, mann A, Tezzon F (2006) Corticospinal with acquired hepatocerebral degen-
D’Alessandro R, Morelli MC, Guarino involvement in patients with a porto- eration. Mov Disord 23:768–770
M (2004) Minimal hepatic encephalop- systemic shunt due to liver cirrhosis: a 110. Parkes JD, Murray-Lyon IM, Williams
athy: longitudinal effects of liver trans- MEP study. J Neurol 253:81–85 R (1970) Neuropsychiatric and elec-
plantation. Arch Neurol 61:242–247 99. Newland MC, Ceckler TL, Kordower tro-encephalographic changes after
87. McNeill A, Birchall D, Hayflick SJ, JH, Weiss B (1989) Visualizing manga- transplantation of the liver. Q J Med
Gregory A, Schenk JF, Zimmerman EA, nese in the primate basal ganglia with 39:515–527
Shang H, Miyajima H, Chinnery PF magnetic resonance imaging. Exp 111. Pennington JA, Schoen SA (1996)
(2008) T2 and FSE MRI distinguishes Neurol 106:251–258 Contributions of food groups to
four subtypes of neurodegeneration estimated intakes of nutritional
with brain iron accumulation. Neurol- elements: results from the FDA total
ogy 70:1614–1619 diet studies, 1982–1991. Int J Vitam
Nutr Res 66:342–349
331

112. Pennington JA, Young BE, Wilson DB, 124. Rose C, Butterworth RF, Zayed J, 136. Sinha S, Taly AB, Prashanth LK,
Johnson RD, Vanderveen JE (1986) Normandin L, Todd K, Michalak A, Ravishankar S, Arunodaya GR,
Mineral content of foods and total Spahr L, Huet PM, Pomier-Layrargues Vasudev MK (2007) Sequential MRI
diets: the Selected Minerals in Foods G (1999) Manganese deposition in changes in Wilson’s disease with
Survey, 1982 to 1984. J Am Diet Assoc basal ganglia structures results from de-coppering therapy: a study of 50
86:876–891 both portal-systemic shunting and patients. Br J Radiol 80:744–749
113. Perl DP, Olanow CW (2007) The neu- liver dysfunction. Gastroenterology 137. Soffer D, Sherman Y, Tur-Kaspa R, Eid
ropathology of manganese-induced 117:640–644 A (1995) Acquired hepatocerebral
Parkinsonism. J Neuropathol Exp 125. Rosenstock HA, Simons DG, Meyer JS degeneration in a liver transplant
Neurol 66:675–682 (1971) Chronic manganism: neuro- recipient. Acta Neuropathol (Berl)
114. Pinarbasi B, Kaymakoglu S, Matur Z, logic and laboratory studies during 90:107–111
Akyuz F, Demir K, Besisik F, Ozdil S, treatment with levodopa. JAMA 217: 138. Spahr L, Butterworth RF, Fontaine S,
Boztas G, Cakaloglu Y, Mungan Z, 1354–1358 Bui L, Therrien G, Milette PC, Lebrun
Okten A (2008) Are acquired hepato- 126. Russo N, Edwards M, Andrews T, LH, Zayed J, Leblanc A, Pomier-
cerebral degeneration and hepatic O’Brien M, Bhatia KP (2004) Heredi- Layrargues G (1996) Increased blood
myelopathy reversible? J Clin Gastro- tary haemochromatosis is unlikely to manganese in cirrhotic patients: rela-
enterol Aug 11 [Epub ahead of print] cause movement disorders – a critical tionship to pallidal magnetic reso-
115. Pomier-Layrargues G, Spahr L, But- review. J Neurol 251:849–852 nance signal hyperintensity and
terworth RF (1995) Increased manga- 127. Saporta MA, André C, Bahia PR, neurological symptoms. Hepatology
nese concentrations in pallidum of Chimelli L, Pais PH, Silva TS, Correia 24:1116–1120
cirrhotic patients. Lancet 345:735 AH, Nobre AB (2004) Acquired hepa- 139. Spahr L, Vingerhoets F, Lazeyras F,
116. Pomier-Layrargues GP, Shapcott D, tocerebral degeneration without Delavelle J, DuPasquier R, Giostra E,
Spahr L, Butterworth RF (1995) Accu- overt liver disease. Neurology 63: Mentha G, Terrier F, Hadengue A
mulation of manganese and copper 1981–1982 (2000) Magnetic resonance imaging
in pallidum of cirrhotic patients: role 128. Sarmiento de la Iglesia MM, Lecum- and proton spectroscopic alterations
in the pathogenesis of hepatic berri Cortés G, Lecumberri Cortés I, correlate with parkinsonian signs in
encephalopathy? Metab Brain Dis Oleaga Zufiria L, Isusi Fontan M, patients with cirrhosis. Gastroenter-
10:353–356 Grande Icaran D (2006) Intracranial ology 119:774–781
117. Pomier-Layrargues GP (2001) Move- calcifications on MRI. Radiologia 140. Steinborn M, Leiz S, Rüdisser K,
ment dysfunction and hepatic 48:19–26 Griebel M, Harder T, Hahn H (2004)
encephalopathy. Metab Brain Dis 129. Schmidt S, Gudinchet F, Meagher- CT and MRI in haemolytic uraemic
16:27–35 Villemure K, Maeder P (2001) Brain syndrome with central nervous
118. Powell EE, Pender MP, Chalk JB, involvement in haemolytic-uraemic system involvement: distribution of
Parkin PJ, Strong R, Lynch S, Kerlin P, syndrome: MRI features of coagula- lesions and prognostic value of
Cooksley WG, Cheng W, Powell LW tive necrosis. Neuroradiology 43: imaging findings. Pediatr Radiol
(1990) Improvement in chronic hepa- 581–585 34:805–810
tocerebral degeneration following 130. Sener RN (2003) Diffusion MR Imag- 141. Stepens A, Logina I, Liguts V, Aldins P,
liver transplantation. Gastroenterol- ing Changes Associated with Wilson Eksteina I, Platkājis A, Mārtinsone I,
ogy 98:1079–1082 Disease. AJNR Am. J. Neuroradiol 24: Tērauds E, Rozentāle B, Donaghy M
119. Prayer D, Grois N, Prosch H, Gadner 965–967 (2008) Parkinsonian syndrome in
H, Barkovich AJ (2004) Magnetic res- 131. Servin-Abad L, Tzakis A, Schiff ER, methcathinone users and the role of
onance imaging presentation of (in- Regev A (2006) Acquired hepatocere- manganese. N Engl J Med 358:
tra) cranial disease associated with bral degeneration in a patient with 1009–1017
Langerhans cell histiocytosis. Am J HCV cirrhosis: complete resolution 142. Stracciari A, Guarino M, Pazzaglia P,
Neuroradiol 25:880–891 with subsequent recurrence after Marchesini G, Pisi P (2001) Acquired
120. Provenzale JM, Barboriak DP, Sims K liver transplantation. Liver Transpl hepatocerebral degeneration: full
(1995) Neuroradiologic findings in 12:1161–1165 recovery after liver transplantation.
fucosidosis, a rare lysosomal storage 132. Shinotoh H, Snow BJ, Chu NS, Huang J Neurol Neurosurg Psychiatry 70:
disease. AJNR Am J Neuroradiol 16 CC, Lu CS, Lee C, Takahashi H, Calne 136–137
(4 Suppl):809–813 DB (1997) Presynaptic and postsyn- 143. Stracciari A, Mattarozzi K, D’Alessan-
121. Pujol A, Pujol J, Graus F, Rimola A, aptic striatal dopaminergic function dro R, Baldin E, Guarino M (2008)
Peri J, Mercader JM, García-Pagan JC, in patients with manganese intoxica- Cognitive functioning in chronic
Bosch J, Rodés J, Tolosa E (1993) tion: a positron emission tomography acquired hepatocerebral degenera-
Hyperintense globus pallidus on T1- study. Neurology 48:1053–1056 tion. Metab Brain Dis 23:155–160
weighted MRI in cirrhotic patients is 133. Shinotoh H, Snow BJ, Hewitt KA, Pate 144. Sussman EB, Sternberg SS (1975)
associated with severity of liver fail- BD, Doudet D, Nugent R, Perl DP, Hereditary hemorrhagic telangiecta-
ure. Neurology 43:65–69 Olanow W, Calne DB (1995) MRI and sia. A case with hepatocellular carci-
122. Read AE, Sherlock S, Laidlaw J, PET studies of manganese-intoxi- noma and acquired hepatocerebral
Walker JG (1967) The neuro-psychi- cated monkeys. Neurology 45: degeneration. Arch Pathol 99:95–100
atric syndromes associated with 1199–1204 145. Taguchi Y, Takashima S, Hirade S,
chronic liver disease and an extensive 134. Shulman LM, Minagar A, Weiner WJ Inoue H (1999) Disappearance of
portal-systemic collateral circulation. (2003) Reversal of parkinsonism fol- globus pallidus hyperintensity in a
Q J Med 36:135–150 lowing liver transplantation. Neurol- patient with portal-systemic enceph-
123. Rodier J (1955) Manganese poisoning ogy 60:519 alopathy after occlusion of the shunt
in Moroccan miners. Br J Indust Med 135. Silverdale MA, Schneider SA, Bhatia vessel. Rinsho Shinkeigaku 39:
12:21–35 KP, Lang AE (2008) The spectrum of 565–569
orolingual tremor – A proposed clas-
sification system. Mov Disord 23:
159–167
332

146. Taylor-Robinson SD, Oatridge A, 152. United Network for Organ Sharing. 159. Wilson S (1912) Progressive lenticu-
Hajnal J, Burroughs A, McIntyre N, Data. (Accessed September 1, 2007) lar degeneration: a familial nervous
Desouza N (1995) MR imaging of the at: www.unos.org/data/ disease associated with cirrhosis of
basal ganglia in chronic liver disease: 153. vanWoerkem W (1914) La cirrhose the liver. Brain 34:295–509
correlation of T1-weighted and mag- hepatique avec alterations dans les 160. Wnorowski M, Prosch H, Prayer D,
netisation transfer contrast measure- centres nerveux evoluant chez des Janssen G, Gadner H, Grois N (2008)
ments with liver dysfunction and sujets d’age moyen. Nouvelle Icono- Pattern and course of neurodegenera-
neuropsychiatric status. Metab Brain graphie de la Salpetriere, Clinique des tion in langerhans cell histiocytosis.
Dis 10:175–188 Maladies du Systeme Nerveux 7: J Pediatr 153:127–132
147. Thobois S, Giraud P, Debat P, Gout- 41–51 161. Wolters EC, Huang CC, Clark C,
tard M, Maurizi A, Perret-Liaudet A, 154. Victor M, Adams RD, Cole M (1965) Peppard RF, Okada J, Chu NS, Adam
Kopp N, Broussolle E (2002) Orofacial The acquired (non-Wilsonian) type MJ, Ruth TJ, Li D, Calne DB (1989)
dyskinesias in a patient with primary of chronic hepatocerebral degenera- Positron emission tomography in
biliary cirrhosis: a clinicopathological tion. Medicine (Baltimore) 44: manganese intoxication. Ann Neurol
case report and review. Mov Disord 345–396 26:647–651
17:415–419 155. Walshe JM (2007) Cause of death in 162. Wolz M, Junghanns S, Löhle M, von
148. Toghill P, Johnston A, Smith J (1967) Wilson disease. Mov Disord 22: Kummer R, Storch A (2008) Dystonia
Choreoathetosis in portosystemic 2216–2220 associated with hyperintense basal
encephalopathy. J Neurol Neurosurg 156. Weissenborn K, Ehrenheim C, Hori A, ganglia lesions on T1-weighted brain
Psychiatry 30:358–363 Kubicka S, Manns MP (1995) Pallidal MRI. Mov Disord 23:1618–1619
149. Triulzi F, Parazzini C, Righini A lesions in patients with liver cirrho- 163. Yamada M, Ohno S, Okayasu I, Okeda
(2006) Patterns of damage in the sis: clinical and MRI evaluation. R, Hatakeyama S, Watanabe H, Ushio
mature neonatal brain. Pediatr Radiol Metab Brain Dis 10:219–231 K, Tsukagoshi H (1986) Chronic
36:608–620 157. Weissenborn K, Tietge UJ, Bokemeyer manganese poisoning: a neuropatho-
150. Troisi R, Debruyne J, de Hemptinne B M, Mohammadi B, Bode U, Manns logical study with determination of
(1999) Improvement of hepatic myel- MP, Caselitz M (2003) Liver trans- manganese distribution in the brain.
opathy after liver transplantation. plantation improves hepatic myelopa- Acta Neuropathol (Berl) 70:273–278
N Engl J Med 340:151 thy: evidence by three cases. Gastro- 164. Yoshikawa K, Matsumoto M,
151. Ueki Y, Isozaki E, Miyazaki Y, Koide R, enterology 124:346–351 Hamanaka M, Nakagawa M (2003) A
Shimizu T, Yagi K, Hirai S (2002) Clin- 158. Wijdicks EF, Wiesner RH (2003) case of manganese induced parkin-
ical and neuroradiological improve- Acquired (non-Wilsonian) hepato- sonism in hereditary haemorrhagic
ment in chronic acquired hepatocere- cerebral degeneration: complex telangiectasia. J Neurol Neurosurg
bral degeneration after management decisions. Liver Transpl Psychiatry 74:1312–1314
branched-chain amino acid therapy. 9:993–994
Acta Neurol Scand 106:113–116

Você também pode gostar