You are on page 1of 12

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/51046062

Zahr NM, Kaufman KL, Harper CG. Clinical and


pathological features of alcohol-related brain
damage. Nat Rev Neurol ....

Article in Nature Reviews Neurology · May 2011


DOI: 10.1038/nrneurol.2011.42 · Source: PubMed

CITATIONS READS

116 1,581

3 authors, including:

Kimberley Louise Kaufman Clive Harper


University of Sydney University of Sydney
50 PUBLICATIONS 443 CITATIONS 226 PUBLICATIONS 8,996 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Microparticles as biomarkers for TB infection. View project

Proteomic Studies of Non-melanoma Skin Cancer View project

All content following this page was uploaded by Kimberley Louise Kaufman on 04 June 2014.

The user has requested enhancement of the downloaded file.


REVIEWS
Clinical and pathological features
of alcohol-related brain damage
Natalie M. Zahr, Kimberley L. Kaufman and Clive G. Harper
Abstract | One of the sequelae of chronic alcohol abuse is malnutrition. Importantly, a deficiency in thiamine
(vitamin B1) can result in the acute, potentially reversible neurological disorder Wernicke encephalopathy (WE).
When WE is recognized, thiamine treatment can elicit a rapid clinical recovery. If WE is left untreated, however,
patients can develop Korsakoff syndrome (KS), a severe neurological disorder characterized by anterograde
amnesia. Alcohol-related brain damage (ARBD) describes the effects of chronic alcohol consumption on
human brain structure and function in the absence of more discrete and well-characterized neurological
concomitants of alcoholism such as WE and KS. Through knowledge of both the well-described changes
in brain structure and function that are evident in alcohol-related disorders such as WE and KS and the
clinical outcomes associated with these changes, researchers have begun to gain a better understanding of
ARBD. This Review examines ARBD from the perspective of WE and KS, exploring the clinical presentations,
postmortem brain pathology, in vivo MRI findings and potential molecular mechanisms associated with these
conditions. An awareness of the consequences of chronic alcohol consumption on human behavior and brain
structure can enable clinicians to improve detection and treatment of ARBD.
Zahr, N. M. et al. Nat. Rev. Neurol. 7, 284–294 (2011); published online 12 April 2011; doi:10.1038/nrneurol.2011.42

Introduction
Alcoholism is an addictive disorder with multi­faceted bio- different genetic make-up remains to be determined.
logical underpinnings (Box 1). Frequent concomitants of Our objective here is to describe a potential continuum
alcoholism are liver disease (steatosis, hepatitis and cirrho- between ARBD, WE and KS with respect to changes in
sis),1 cardiovascular disease2 and mal­nutrition.3 The neuro- human behavior and brain structure. Note that while this
logical consequences associated with this addictive disorder Review is extensive, it is not intended to be exhaustive.
include hepatic encephalopathy, Wernicke encephalopathy
(WE), Korsakoff syndrome (KS), Marchiafava–Bignami The Wernicke–Korsakoff syndrome
disease (MBD) and central pontine myelinolysis (CPM). WE is an acute, potentially reversible neurological dis­
Each of these relatively well-characterized alcohol-related order caused by a deficiency in or severe depletion of thia-
CNS disorders is associated with a unique clinical pre- mine (vitamin B1) that can result from chronic alcoholism,
sentation and a discrete neuropathological and neuro­ poor nutrition, long-term parenteral feeding, hyper­emesis
radiological signature (Figure 1).4 The structural changes gravi­darum or bariatric surgery.7,8 Incidence rates of WE in
to the brain and functional consequences that occur with the general population—on the basis of autopsy findings
chronic alcohol consumption in the absence of diagnosable in Western countries—range from 0.1–2.8%, but can be as
neuro­logical concomitants of alcoholism (that is, in cases high as 12.5% in patients with alcoholism.9,10 Such indivi­
of uncomplicated alcoholism) are grouped under the term duals are at a high risk of thiamine deficiency because of
Department of ‘alcohol-related brain damage’ (ARBD). the poor diet associated with their lifestyle, and the fact
Psychiatry and
Behavioral Sciences,
Whether ARBD represents one end of a continuum of that chronic alcoholism compromises thiamine absorp-
401 Quarry Road, neurological deficits, with disorders such as KS and MBD tion from the gastro­intestinal tract, impairs thiamine
Stanford University,
at the other end,5 or one outcome in a range of dis­­con­ storage, and may reduce the phosphorylation of thiamine
Stanford, CA 94305,
USA (N. M. Zahr). tinuous, graded deficits occurring with chronic alcohol to its biologically active form, thiamine py­rophosphate
Neuropathology Unit, exposure and, for example, aging 6 remains unclear. In (TPP; Figure 2).11–15
Discipline of Pathology,
Blackburn Building, addition, whether people with certain genotypes (for Guidelines for the diagnosis, treatment and prevention
D06, Western Avenue, example, individuals who are genetically susceptible to of WE have been released by the European Federation of
The University of
Sydney, NSW 2006,
mal­nutrition or liver compromise) are at a greater risk of Neurological Societies (EFNS), and are based on three
Australia particular neuro­logical conditions and, consequently, are decades of research into this condition (Box 2).16 If WE
(K. L. Kaufman, more likely to express specific a­lcoholism-related neuro­ is recognized, treatment with thiamine can result in rapid
C. G. Harper).
psychological compromise than are individuals with a clinical improvement.10 Indeed, the prevalence of WE has
Correspondence to: been reduced in a number of countries (including the US,
C. G. Harper
clive.harper@ Competing interests the UK and Australia) that have instituted nationwide
sydney.edu.au The authors declare no competing interests. th­iamine supplementation in staple foods such as bread.17

284  |  MAY 2011  |  VOLUME 7  www.nature.com/nrneurol


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

When WE is left undiagnosed and untreated, ≈80% of Key points


patients with this condition develop KS, a severe, typically
■■ Alcohol can cause a spectrum of untoward structural and functional changes
permanent neurological disorder characterized by antero- in the brain
grade amnesia.18 The term Wernicke–Korsakoff syndrome
■■ The spectrum of disruption includes alcohol-related brain damage at one end
(WKS) is used to denote the range of brain and behavioral and complications such as hepatic encephalopathy, Wernicke encephalopathy,
impairments associated with thiamine deficiency.19,20 Korsakoff syndrome, Marchiafava–Bignami disease and central pontine
myelinolysis at the other
Clinical and psychological features ■■ The clinical diagnoses of alcohol-related brain damage and even Wernicke
Clinicians are often taught to diagnose WE on the basis encephalopathy can be difficult to make, and many cases of these conditions
of the presence of the classic clinical triad of ocular are missed
motor abnormalities, cerebellar dysfunction, and altered ■■ Changes to the brain associated with alcohol intake are regionally specific and
mental state. Ocular motor abnormalities occur in ≈30% can affect both gray and white matter; some of these changes are reversible
of patients with WE and may include nystagmus or with abstinence
ophthalmo­plegia, while cerebellar dysfunction can be ■■ Pathogenic mechanisms associated with alcoholism are under investigation,
found in ≈25% of patients with this disorder and may with neuroinflammation currently receiving particular attention
manifest as loss of equilibrium, incoordination of gait,
trunk ataxia, dysdiadochokinesia and, occasionally, limb
ataxia or dysarthria. Approximately 80% of patients with Box 1 | Prelude to alcoholism
WE exhibit an altered mental state, which may comprise
Alcoholism is the product of multiple interacting factors including complex
mental sluggishness, apathy, impaired awareness of an genetics, the environment, predisposing personality characteristics, and
immediate situation, an inability to concentrate, con­ psychiatric comorbidities.139 Variation in genes that modify the metabolism
fusion or agitation, hallucinations, behavioral disturbances of ethanol, such as those encoding alcohol dehydrogenase140 or aldehyde
mimicking an acute psychotic disorder, or coma.3,10,21 A dehydrogenase,140 has been shown to influence the risk of this condition.
retrospective analysis of the clinical signs and symptoms of Environmental factors that increase vulnerability to alcoholism include severe
patients diagnosed at autopsy as having WE revealed that childhood trauma (for example, emotional, physical or sexual abuse), maternal
depression, and lack of peer or family support.141 An example of a gene–
only 20% of patients with this disorder presented with the
environment interaction that may contribute to the development of alcoholism is
full triad of clinical features and ≈30% of such indivi­duals the influence of the environment on monoamine oxidase A (MAO‑A), an enzyme
exhibited only cognitive impairment.21 Thus, through the that is important for the normal functioning of the serotonergic system. Studies
requirement of the full triad for a positive diagnosis, WE have shown that young boys who experience a traumatic event can develop low
is missed by routine clinical examination in 75–80% of levels of MAO‑A expression, and that this decrease in MAO‑A levels correlates with
cases, even in teaching hospitals. By contrast, the pres- an increase in antisocial behavior, an antecedent of alcohol addiction.142,143
ence of just two of four signs (dietary deficiency, ocular The offspring of individuals with alcoholism can display mild dysfunction of
motor abnormality, cerebellar dysfunction, and either the frontal cortex,144 expressed as personality traits such as impulsivity,
altered mental state or mild memory impairment), which aggressiveness and perseveration, that increase their risk for alcoholism. 145,146
was first suggested by Caine and colleagues22 and is now Similarly, the mesolimbic dopaminergic system, which subserves reward-
recommended by the EFNS,16 can significantly improve dependent behaviors, may be dysfunctional (that is, it confers an attenuated
the diagnostic accuracy for WE. response to natural reward in certain individuals), thereby increasing the risk of
severe alcoholism.147 Common psychiatric illnesses associated with alcoholism
The most salient characteristic of KS is global amnesia.18
include schizophrenia,148,149 bipolar disorder,150 major depression,151 antisocial
Neuropsychological assessments of multi­ple functional
personality disorder,152,153 and general anxiety disorder.154
domains targeting executive functions, declarative and
procedural memory, visuospatial abilities and postural
stability have revealed that individuals with KS have alcoholism exhibit impairments in all the functions
severe deficits in memory for new material and in gait described above.33 Similarly, not all patients with KS have
and balance, despite sparing of general intelligence, short- permanent amnesia.35 This heterogeneity suggests that the
term memory and visuoperceptual implicit learning.23–26 functions affected by chronic alcohol consumption are
Patients with KS may also exhibit prefrontal neuro­ dissociable and supported by different neural systems.36
behavioral dysfunction, expressed as deficits on tests of One study examined the component processes of epi-
problem solving, working memory, cognitive flexibility, sodic memory (memory of auto­biographical events) and
perseverative responding, and self-regulation.27–29 working memory (the ability to hold information ‘online’
Over 80% of individuals with uncomplicated alcohol- while doing complex tasks) in individuals with uncompli-
ism (that is, ARBD) are estimated to show cognitive defi- cated alcoholism and patients with alcoholism and KS. The
cits in executive functions,30,31 although such deficits are former could generally be differentiated from the latter via
mild in comparison with those observed in patients with performance on tests of episodic memory (that is, the two
KS.32 Individuals with uncomplicated alcoholism also groups showed graded impairment in episodic memory),
demonstrate deficits in explicit memory, visuo­spatial despite significant overlap in performance on working-
processes and motor control (for example, speed, gait and memory tasks between the two groups (that is, continuous
balance; Figure 3).33,34 impairment was seen between the two groups).37 On the
The neuropsychological expression of ARBD is marked basis of these findings, the study’s researchers suggested
by heterogeneity in the extent (severity) and type (compo- that impairment of episodic memory was the result of the
nent) of deficit, and not all individuals with uncomplicated untoward effects of alcohol on the Papez (limbic) circuit

NATURE REVIEWS | NEUROLOGY VOLUME 7  |  MAY 2011  |  285


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Malnutrition Liver disease

Mamillary bodies Mamillary bodies

Inflammation Marchiafava–Bignami disease Central pontine myelinolysis

Frontal lobes Pons Putamen

Corpus callosum Hippocampus Amygdala

Cerebellum Caudate Hypothalamus

Thalamus
Figure 1 | Brain regions targeted by alcohol-related disease. Figure courtesy of A. Pfefferbaum, SRI International, CA, USA
and E. V. Sullivan, Stanford University, CA, USA.

and was exacerbated by thiamine deficiency, while the and motor deficits seen in patients with uncomplicated
observed impairment in working memory, which was not alcoholism can be accounted for, in part, by the number
specific to KS, may have reflected the effects of chronic of WE signs present.
alcohol consumption on frontocerebellar circuitry.38
In an attempt to explain the cognitive heterogeneity Postmortem pathological features
commonly seen in patients with alcoholism, a recent pro- The neuropathological changes observed in WKS, as des­
spective study applied the operational criteria of Caine cribed by Victor and Adams in the early 1970s, include
and colleagues to a group of individuals with uncompli- lesions in periventricular regions around the third and
cated alcoholism, so as to determine whether the pres- fourth ventricles, and atrophy of the mamillary bodies.40
ence of any of the four signs, determined by history or By contrast, traditional clinical patho­logical methods
current examination, could be used to predict perfor- have only been able to demonstrate mild cerebral atrophy
mance on a battery of neuropsychological tests. Among and lower mean brain weight in cases of uncomplicated
the 56 patients with uncomplicated alcoholism who were alcohol­ism.41–43 Thus, quantitative studies are required to
assessed, 16% displayed two or more signs, 57% showed charac­terize the relatively subtle structural abnormalities
only one sign, and 27% met no criteria. In this sample of in the brain that are caused by the direct effects of alcohol
sober, community-dwelling individuals, self-reported (Box 3).
dietary deficiency (n = 29) and cere­bellar dysfunction In one quantitative study, brain volume with respect to
(that is, ataxia; n = 20) were frequently described, while intracranial cavity volume was determined and the mean
oculomotor abnormalities (n = 2) and mental impairment pericerebral space was shown to rise from 8.3% of the total
(n = 0) were rarely observed.39 This study revealed a graded intracranial cavity volume in healthy controls to 11.3% in
effect in cognitive and motor performance among patient patients with ARBD and 14.7% in patients with WKS.44
subgroups: individuals with alcoholism who did not meet Stereometric studies have suggested that this reduction
any criteria performed at levels equivalent to healthy in brain volume is largely accounted for by the shrink-
controls, whereas patients with one sign showed mild-to- age of white matter.45–47 Cerebellar white matter volume
moderate neuro­psychological deficits, and patients with (especially in the vermis) is reduced 48 and the corpus
two or more signs showed the most severe deficits on each callo­sum area is significantly thinned in individuals with
neuro­psychological domain evaluated. This graded effect alcoholism,49,50 especially those with nutritional deficien-
suggests that the heterogeneity in the severity of cognitive cies,51 compared with healthy controls. This finding may

286  |  MAY 2011  |  VOLUME 7  www.nature.com/nrneurol


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

represent a dose effect of alcohol rather than an effect of


thiamine deficiency, as white matter volume was negatively Thiamine in diet owing
Alcohol consumption
correlated with maximum daily alcohol consumption.47 to poor nutrition
The nature of the white matter loss remains unknown;
however, this phenomenon probably involves changes in
both myelination and axonal integrity.52 Thiamine absorption Thiamine phosphorylation
from gastrointestinal tract in brain
In addition to atrophy of the mamillary bodies, WKS
reveals neuronal loss in the anterior principal and medio­
dorsal nuclei of the thalamus53 and in the basal forebrain.54 Thiamine pyrophosphate
Thiamine storage in liver
In patients with alcoholism and signs of WE, a reduc- (‘active thiamine’) in brain
tion in Purkinje cell density and molecular layer atrophy
are noted in the cerebellum, suggesting that this brain
region is selectively vulnerable to thiamine deficiency.55 Biosynthesis (e.g. biosynthesis
of amino acids and proteins) Energy (ATP)
In patients with uncomplicated alcoholism, micro-
scopic studies have revealed an ≈25% loss of pyramidal Figure 2 | Interactions between alcohol consumption and thiamine deficiency.
neurons in the superior frontal and frontal association
(dorsolateral portion) cortices.56,57 Little evidence exists
for neuronal loss in the primary motor cortex in ARBD. Box 2 | EFNS guidelines for diagnosis, therapy and prevention of WE
However, a silver impregnation technique has shown that ■■ The clinical diagnosis of WE should take into account the different
pyramidal neurons in both the superior frontal and motor presentations of clinical signs between individuals with and without
cortices have dendritic arbor shrinkage,58 indicating com- alcoholism; although the prevalence of WE is higher in the former than the
latter group, WE should be suspected in all clinical conditions that could lead
promise in interneuronal communication. Dendritic
to thiamine deficiency
shrinkage has been shown to be reversible in a rodent
model of alcoholism following a prolonged period of ■■ The clinical diagnosis of WE in patients with alcoholism requires the presence
of two of the following four signs: dietary deficiencies, eye signs, cerebellar
abstinence.59 Subcortical regions of brains from patients
dysfunction, and either an altered mental state or mild memory impairment
with uncomplicated alcoholism exhibit neuronal loss in
■■ Total thiamine levels in a blood sample should be measured immediately before
the supraoptic and para­ventricular nuclei of the hypo­
thiamine administration
thalamus that shows a positive correlation with maximum
■■ MRI should be used to support the diagnosis of acute WE in patients both
daily alcohol consumption.60 With respect to the cerebel­
with and without alcoholism
lum, pathological studies do not consistently show a
■■ Thiamine is indicated for the treatment of suspected or manifest WE, and
decrease in the number of neurons in cases of ARBD
should be administered before any carbohydrate at a dose of 200 mg three
compared with normal controls, suggesting that chronic times daily, preferably intravenously
alcohol consumption per se does not neces­sarily cause
■■ The overall safety of thiamine is very good
neuronal death in this region of the brain. No changes
■■ After bariatric surgery, thiamine status should be monitored for at least
have been documented in the number of neurons in the
6 months and be accompanied by parenteral thiamine supplementation
basal ganglia,61 hippo­campus57,62 or seroto­nergic raphe
nuclei63 in ARBD. ■■ Parenteral thiamine should be given to all at-risk individuals admitted to an
emergency room

In vivo MRI features ■■ Patients who die from symptoms suggesting WE should have an autopsy
MRI has shown that patients with KS have an increase in Derived from Galvin, R. et al. (2010).16 Abbreviations: EFNS, European Federation of
cerebrospinal fluid volume and widespread gray matter Neurological Societies; WE, Wernicke encephalopathy.
volume deficits.5,64 Moreover, group analysis has revealed
substantial volume changes in the mamillary bodies of
individuals with KS,65,66 although mamillary body shrink- patients, suggesting that the aging brain is especially sus-
age is not a necessary concomitant of this condition.3,67 ceptible to ARBD.70,73 Structural MRI has also demon-
MRI has also demonstrated volume losses in the orbito­ strated that indivi­duals with alcoholism have significant
frontal cortices and other hypothalamic nuclei in KS.5 The volume deficits in the corpus callosum74,75 and cerebellar
volume losses that best differentiate KS from uncompli­ white matter.48,76 In contrast to postmortem findings, MRI
cated alcoholism, however, involve the thalamus.68 In has provided in vivo evidence for volume deficits in the
acute WE, MRI can be used to detect symmetrical, an­terior hippocampus of patients with chronic alcohol-
bi­lateral hyperintense foci (clearly visible on T2-weighted ism.77,78 These deficits are potentially accounted for by a
and fluid-attenuated inversion recovery images) in peri­ loss of non-neuronal cells; that is, glia. Altogether, use of
aqueductal gray matter, the mamillary bodies, and the MRI to characterize WKS structural brain changes in the
tissue surrounding the third ventricle.5,69 context of the neuropathology of uncomplicated alcohol-
In patients with uncomplicated alcoholism, MRI ism has revealed a graded pattern of volume deficits (from
studies have generally confirmed postmortem studies mild deficits in ARBD to moderate or severe deficits in
by demonstrating that such patients have regional corti- WKS) in the mamillary bodies, hippocampus, thalamus,
cal volume deficits,64,70 especially in the frontal lobes.71,72 cerebellum and pons (Figure 4).5 As brain regions outside
Among individuals with alcoholism, cerebral shrinkage those traditionally associated with thiamine depletion
is more pronounced in older patients than in younger (for example, the frontal cortices, hippocampus and

NATURE REVIEWS | NEUROLOGY VOLUME 7  |  MAY 2011  |  287


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Executive functioning Memory

Mamillary bodies

Visuospatial abilities Upper and lower limb motor control Ocular motor control

Frontal lobes Pons Putamen


Corpus callosum Hippocampus Amygdala
Cerebellum Caudate Hypothalamus
Thalamus

Figure 3 | Functions and associated brain regions targeted by alcohol abuse and alcoholism. Figure courtesy of
A. Pfefferbaum, SRI International, CA, USA and E. V. Sullivan, Stanford University, CA, USA.

Box 3 | The New South Wales Tissue Resource Centre presenting signs and neuro­radiological profiles in patients
with alcoholism.79
For the past 25 years, the New South Wales Tissue In contrast to postmortem studies, in vivo magnetic
Resource Centre ‘brain bank’155 has provided much of the resonance (MR) modalities constitute safe, noninvasive
postmortem tissue (fresh-frozen and formalin-fixed) used by
methods for longitudinal examination of the condition of
research groups throughout the world to explore the effects
of alcohol on the brain.156–158 The validity of research using
the brain in patients with alcoholism during the natural
brain bank material largely depends on careful clinical course of chronic alcohol consumption, detoxification,
and pathological characterization of each case, precise abstinence or relapse. Such MR studies have demon-
matching to control cases, and appropriate storage. Brain strated that some structural brain changes are reversible
bank tissue has been used for structural and molecular with prolonged abstinence from alcohol.72,80–82 Indeed,
studies and to test hypotheses developed from animal ARBD may have two components, one of which is tran-
models and in vivo studies. To ensure the long-term sient and the other being permanent.83 If brain volume
success of the brain bank, their premortem, ‘in-life’ donor
loss is due to neuronal loss, brain volume recovery will
program carefully details the lifestyle and medical histories
of individuals who have committed to donating their tissue be incomplete with abstinence. For example, some MR
on their death.159,160 An important advantage of this tissue spectroscopy studies have shown that in spite of prolonged
is that it is not restricted to the small sample of patients abstinence, individuals who have chronically consumed
with alcoholism who are in treatment (estimated to be 25% alcohol demonstrate persistent N‑acetylaspartate (a puta-
of the total population of individuals with alcoholism in the tive marker of neuronal integrity) decreases in the frontal
USA), a limitation of in vivo studies, which typically rely on lobes,84–86 the thalamus86 and the cerebellum.86,87 Other
treatment-seeking patients.161
studies, however, have found improvements in the levels
of N‑acetylaspartate and choline—another metabolite
pons) are affected in both uncomplicated alcoholism that may indicate remyelination—with abstinence.88–90
and KS, alcohol­ism alone or in combination with nutri- Structural repair of myelin could explain the increase in
tional deficiencies may have roles in the mechanisms white matter volume that has been shown to occur after
under­lying these brain abnormalities. Indeed, multiple periods of abstinence from alcohol.91–93
subclinical episodes of thiamine deficiency or other nutri- Another advantage of in vivo MR tools is the facility to
tional deficiencies may contribute to the graded nature of conduct behavioral experiments concurrently with imaging,
brain regional volume deficits and to the heterogeneity in so as to determine brain structure–­function relationships.

288  |  MAY 2011  |  VOLUME 7  www.nature.com/nrneurol


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Combined neuropsychological and neuroimaging studies a


suggest that the amnesia observed in KS may be caused by
interruption of a complex d ­ iencephalic–hippocampal cir-
cuitry that includes thalamic nuclei and mamillary bodies,
rather than through an insult to a single node in the circuit
such as the hippo­campus.94 This hypothesis has received
support from a study using a novel ‘resting state’ functional
MRI analysis, which demonstrated that improvement in
memory function in patients recovering from WE parallels
the level of mammillothalamic ‘functional connec­tivity’.95 b
Diffusion tensor imaging (DTI), which is particularly
useful in the characterization of the integrity of white
matter microstructure, supports a positive correlation
between disruption of the microstructural integrity of the
corpus callosum and deficits in visuospatial performance,
gait and balance.81,96–98 In addition to confirming the con-
tribution of cerebellar white matter volume loss (especially
in the vermis) to ataxia in patients with chronic alcoholism,
combined brain imaging and neuropsychological methods c
have demonstrated the importance of frontocerebellar
connections99 to cognitive and sensory functioning,100–102
including perceptual motor tasks, executive functions, and
learning and memory.99,103,104 Improvements with absti-
nence in brain structure and biochemical status have been
demonstrated105 that correspond with improvement or
reversal of functional deficits in working memory, postur­al
stability and visuospatial ability.106,107
Figure 4 | Graded brain-volume deficits in alcoholism and its sequelae. T1-weighted
MRI scans from a | a 63-year-old healthy control male, b | a 59-year-old man with
Molecular features
alcoholism, and c | a 63-year-old man with WKS. Graded enlargement of the
A number of incompletely understood, mutually inclusive ventricles (indicating shrinkage of the surrounding tissue) can be observed from the
mechanisms have been proposed to explain how ethanol healthy control to the individual with WKS. Sagittal (left column), axial (middle
causes brain damage (Box 4). These mechanisms include column) and coronal (right column) brain images are shown. Abbreviation: WKS,
neurotoxicity of the ethanol molecule itself, and the con- Wernicke–Korsakoff syndrome. Figure courtesy of A. Pfefferbaum, SRI International,
sequences of nutritional deficiencies or liver dysfunction, CA, USA.
each of which can lead to the intriguing possibility of
alcohol-induced neuroinflammation. thereby interrupting energy production through the Krebs
Neuroinflammation is considered to be involved in the cycle and pentose phosphate pathway; lipid metabolism,
pathogenesis and progression of many neurodegenera- thereby interrupting the production and maintenance of
tive disorders,108,109 and a mechanistic role for this process myelin; and amino acid metabolism, thereby interrupt-
in ARBD has recently been proposed.110 The brain cells ing the production of glucose-derived neurotransmitters.10
that might mediate neuroinflammation are microglia, These metabolic deficits can contribute to neuronal and
which can exist in multiple states, with the activation white matter damage.
of these cells resulting in either anti-inflammatory or Bouts of thiamine deficiency may occur in upwards
pro­inflammatory responses.111 A recent study investi- of 80% of patients with alcoholism;114,115 however, only
gated several markers of brain inflammation in ‘brain ≈13% of such individuals develop WKS,116 raising the
bank’ tissue from patients with alcoholism and indivi­ possibility that a genetic predisposition to WKS may exist
duals who had moderate levels of alcohol consumption. in some individuals.15 Some studies have shown that tran-
Various patterns of positive signs of neuroinflammation sketolase binds TPP less effectively in patients with WKS
were identified in the ventral tegmental area, substantia than in healthy controls.117,118 No consistent correlation,
nigra, hippocampus and amygdala in the individuals with however, has been found between transketolase variants
alcoholism,110 providing some support for a mechanistic and thiamine deficiency.119 Other genetic loci or variants
role for inflammation in al­cohol-related alterations to the associated with WKS susceptibility include the X‑linked
brain reward system. transketolase-like 1 gene,120 the high-affinity thiamine
As described above, individuals with alcoholism have a transporter protein gene SLC19A2,121 the γ‑aminobutyric
high risk of thiamine deficiency because of poor nutrition, acid A receptor subunit gene cluster on chromosome
impaired absorption of thiamine from the gastrointestinal 5q33, 122 and the aldehyde dehydrogenase‑2 ADH21
tract, and reduced liver stores.112 Moreover, alcohol inter- allele.123 One possibility is that several genetic variants and
feres with the conversion of thiamine to its metabolically environmental factors must be present to generate a WKS
active form, namely TPP.113 A reduction in TPP levels dis- phenotype, which only becomes clinically relevant when
rupts the following processes: carbohydrate metabolism, an individual’s diet is deficient in thiamine.10

NATURE REVIEWS | NEUROLOGY VOLUME 7  |  MAY 2011  |  289


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 4 | Select pathophysiological mechanisms underlying ARBD In recent years, high-throughput genomic and pro-
teomic approaches have been used extensively to provide
Ethanol-specific effects
clues about the molecular mechanisms underlying ARBD.
■■ Toxic metabolites of ethanol such as acetaldehyde or fatty acid ethyl esters can
Oligonucleotide and complementary DNA microarray
accumulate and lead to adduct formation, which can disorder lipids, interrupt
mitochondrial function, and induce neuronal damage162,163
studies of samples of human frontal cortex from indivi­
duals with alcoholism have identified alcohol-responsive
■■ Ethanol increases the generation of reactive oxygen species (such as nitric
genes relating to several broad categories, namely myelina-
oxide and lipid peroxidation products), which can accumulate and cause DNA
damage, inhibition of gene expression, and neuronal death164–167
tion, synaptic structure, mitochondria, signal transduction,
intracellular metabolism, protein trafficking, apoptosis
■■ Ethanol lowers brain-derived neurotrophic factor levels and, hence, may
and transcriptional regulation.124–127 Moreover, samples
impair intracellular signaling pathways involved in cell survival, growth and
of human temporal cortex from patients with alcohol­
differentiation, thereby enhancing natural cell death168
ism have exhibited changes in the expression of genes
■■ Removal of ethanol causes brain disinhibition, dysregulation of glutamate encoding proteins related to mitochondria, the ubiqui­
release and uptake, and stimulation of NMDA receptors that mediate
tin system or signal transduction.128 Alcohol-responsive
excitotoxicity169
genes expressed in the nucleus accumbens and the ventral
Thiamine deficiency tegmental area are primarily associated with changes in
■■ Thiamine deficiency leads to low levels of thiamine pyrophosphate and, neurotransmission and signal transduction, suggesting
hence, impairment of several biochemical pathways in the brain, including neuroplastic changes that may contribute to changes in
carbohydrate metabolism (for energy production), lipid metabolism (for
reward response. The results of such genomic approaches
production and maintenance of myelin), and amino acid metabolism
(for production of glucose-derived neurotransmitters; for example,
suggest that multiple pathways may be involved in causing
glutamic acid and γ‑aminobutyric acid)10 altered neuronal function and structural changes in ARBD,
although changes in myelin-related genes seem particu-
Liver dysfunction larly important. Indeed, altered expression levels of proteo­
■■ A liver directly damaged by the toxic effects of ethanol is unable to remove lipid protein and myelin basic protein, both of which are
neurotoxic substances such as ammonia and manganese from blood;170
involved in stabilization and compaction of the myelin
accumulation of ammonia affects cerebral blood flow, metabolism and
astrocytic function,171,172 while manganese at high levels can affect the sheath,129,130 could explain the structural and functional
dopaminergic system, enhance oxidative stress, and induce neurotoxicity173,174 changes in white matter in patients with alcoholism.131
Protein expression studies have been conducted in
Synergistic effects
various brain regions including the occipital cortex,127
■■ Acetaldehyde protein adduct formation175 or oxidative stress176 may induce
hippo­campus132 and cerebellum.133 Again, while such
inflammatory liver damage,177 potentially resulting in a generalized immune
response178 and, consequently, central inflammation110,179,180 and neuronal
studies suggest that several pathways may be associated with
degeneration181 ARBD, relevant protein expression studies in patients
with uncomplicated alcoholism show dysregulation of
■■ Liver dysfunction182 or thiamine deficiency113 can contribute to astrocytic
pathology, which may compromise glutamate homeostasis and enhance
key energy-regulating and metabolic proteins, notably
NMDA-receptor-mediated excitoxicity those involved in thiamine-dependent cascades in pre­
frontal gray and white matter,134,135 cerebellar vermis133
Abbreviations: ARBD, alcohol-related brain damage; NMDA, N‑methyl‑d-aspartate. and corpus callosum.136,137 These findings lend weight to
the con­tinuum hypothesis of ARBD, WE and KS.

Review criteria
Conclusions
We have described a potential continuum between ARBD,
Articles were selected on the basis of their contribution WE and KS with respect to changes in human behavior
to the field of alcohol-related brain damage research, and brain structure. The clinical diagnosis of ARBD and
with a particular focus on neuropathological and WE remains difficult; however, an awareness of current
neuroimaging studies in humans. Referenced articles
research findings and a high index of suspicion can aid in
were mostly identified from MEDLINE using access
search engines PubMed and NLM Gateway. Articles were
the detection of these conditions. An intimate relation-
retrieved using keywords such as “alcohol”, “ethanol”, ship seems to exist between alcohol use and thiamine defi-
“alcoholism”, “brain damage”, “white matter loss”, ciency, and we hypothesize that both ARBD and WE may
“atrophy”, “neuropathology”, “Wernicke encephalopathy”, develop as a result of repeated episodes of subclinical thia-
“Korsakoff psychosis”, “Wernicke–Korsakoff sydrome”, mine deficiency.138 Neuroradiological examination (with
“thiamine deficiency”, “pathogenesis”, “neuroimaging”, MRI) is a valuable tool in the diagnosis of acute WE and
“genomics” and “proteomics”. All articles cited were enables in vivo tracking of the progression of brain patho­
published in English and most represent peer-reviewed
logy from ARBD to KS. An awareness of the facts pre-
original research articles. This Review aimed to include
the most recent pathological and radiological data; sented herein by clinicians and other health workers could
however, many articles date back to the 1970s and help minimize the overall burden of ARBD. Moreover,
1980s when much of the original neuropathology public education programs should be promoted so that
research was performed. Some review articles were also indivi­duals using alcohol become aware of the associated
cited and their reference lists scrutinized. Related citation risks and gain an understanding that components of the
lists generated by PubMed searches were also a useful structural and functional changes linked to alcohol use are
source of references.
potentially reversible with abstinence.

290  |  MAY 2011  |  VOLUME 7  www.nature.com/nrneurol


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

1. Lieber, C. S. Alcoholic fatty liver: its 21. Harper, C. G., Giles, M. & Finlay-Jones, R. Clinical 39. Pitel, A. L. et al. Signs of preclinical Wernicke’s
pathogenesis and mechanism of progression to signs in the Wernicke–Korsakoff complex: encephalopathy and thiamine levels as
inflammation and fibrosis. Alcohol 34, 9–19 a retrospective analysis of 131 cases diagnosed predictors of neuropsychological deficits in
(2004). at necropsy. J. Neurol. Neurosurg. Psychiatry 49, alcoholism without Korsakoff’s syndrome.
2. Hillbom, M., Juvela, S. & Karttunen, V. 341–345 (1986). Neuropsychopharmacology 36, 580–588 (2011).
Mechanisms of alcohol-related strokes. Novartis 22. Caine, D., Halliday, G. M., Kril, J. J. & 40. Victor, M., Adams, R. D. & Collins, G. H.
Found. Symp. 216, 193–204 (1998). Harper, C. G. Operational criteria for the The Wernicke–Korsakoff Syndrome (F. A. Davis,
3. Victor, M., Adams, R. D. & Collins, G. H. The classification of chronic alcoholics: identification Philadelphia, 1971).
Wernicke–Korsakoff Syndrome and Related of Wernicke’s encephalopathy. J. Neurol. 41. Harper, C. G. & Blumbergs, P. C. Brain weights in
Neurologic Disorders due to Alcoholism and Neurosurg. Psychiatry 62, 51–60 (1997). alcoholics. J. Neurol. Neurosurg. Psychiatry 45,
Malnutrition, 2nd edn (F. A. Davis, Philadelphia, 23. Fama, R., Marsh, L. & Sullivan, E. V. Dissociation 838–840 (1982).
1989). of remote and anterograde memory impairment 42. Skullerud, K. Variations in the size of the human
4. Zuccoli, G. et al. Neuroimaging findings in and neural correlates in alcoholic Korsakoff brain. Influence of age, sex, body length, body
alcohol-related encephalopathies. AJR Am. syndrome. J. Int. Neuropsychol. Soc. 10, mass index, alcoholism, Alzheimer changes, and
J. Roentgenol. 195, 1378–1384 (2010). 427–441 (2004). cerebral atherosclerosis. Acta Neurol. Scand.
5. Sullivan, E. V. & Pfefferbaum, A. Neuroimaging of 24. Fama, R., Pfefferbaum, A. & Sullivan, E. V. 102, 1–94 (1985).
the Wernicke–Korsakoff syndrome. Alcohol Visuoperceptual priming in alcoholic Korsakoff 43. Harper, C. G. & Kril, J. J. in Alcohol Induced Brain
Alcohol. 44, 155–165 (2009). syndrome. Alcohol. Clin. Exp. Res. 30, 680–687 Damage: NIAAA Research Monograph No. 22
6. Oscar-Berman, M. & Marinkovic, K. Alcoholism (2006). (eds Hunt, W. A. & Nixon, S. J.) 39–69 (NIH,
and the brain: an overview. Alcohol Res. Health 25. Sullivan, E. V., Deshmukh, A., Desmond, J. E., Rockville, 1993).
27, 125–133 (2003). Lim, K. O. & Pfefferbaum, A. Cerebellar volume 44. Harper, C. G., Kril, J. J. & Holloway, R. L. Brain
7. Harper, C. Thiamine (vitamin B1) deficiency and decline in normal aging, alcoholism, and shrinkage in chronic alcoholics: a pathological
associated brain damage is still common Korsakoff’s syndrome: relation to ataxia. study. Br. Med. J. 290, 501–504 (1985).
throughout the world and prevention is simple Neuropsychology 14, 341–352 (2000). 45. De la Monte, S. M. Disproportionate atrophy of
and safe! Eur. J. Neurol. 13, 1078–1082 (2006). 26. Kopelman, M. D., Thomson, A. D., Guerrini, I. & cerebral white matter in chronic alcoholics. Arch.
8. Burns, E. M. et al. Introduction of laparoscopic Marshall, E. J. The Korsakoff syndrome: clinical Neurol. 45, 990–992 (1988).
bariatric surgery in England: observational aspects, psychology and treatment. Alcohol 46. Harper, C. & Kril, J. J. Brain atrophy in chronic
population cohort study. BMJ 341, c4296 (2010). Alcohol. 44, 148–154 (2009). alcoholic patients: a quantitative pathological
9. Thomson, A. D., Cook, C. C., Touquet, R. & 27. Oscar-Berman, M. & Ellis, R. J. Cognitive deficits study. J. Neurol. Neurosurg. Psychiatry 48,
Henry, J. A. The Royal College of Physicians related to memory impairments in alcoholism. 211–217 (1985).
report on alcohol: guidelines for managing Recent Dev. Alcohol. 5, 59–80 (1987). 47. Kril, J. J. & Butterworth, R. F. Diencephalic and
Wernicke’s encephalopathy in the accident and 28. Dirksen, C. L., Howard, J. A., Cronin-Golomb, A. & cerebellar pathology in alcoholic and
emergency department. Alcohol Alcohol. 37, Oscar-Berman, M. Patterns of prefrontal nonalcoholic patients with end-stage liver
513–521 (2002). dysfunction in alcoholics with and without disease. Hepatology 26, 837–841 (1997).
10. Sechi, G. & Serra, A. Wernicke’s encephalopathy: Korsakoff’s syndrome, patients with Parkinson’s 48. Phillips, S. C., Harper, C. G. & Kril, J.
new clinical settings and recent advances in disease, and patients with rupture and repair of A quantitative histological study of the cerebellar
diagnosis and management. Lancet Neurol. 6, the anterior communicating artery. vermis in alcoholic patients. Brain 110,
442–455 (2007). Neuropsychiatr. Dis. Treat. 2, 327–339 (2006). 301–314 (1987).
11. Thomson, A. D., Jeyasingham, M. D., Pratt, O. E. 29. Oscar-Berman, M. in Review of NIAAA’s 49. Harper, C. G. & Kril, J. J. Corpus callosal
& Shaw, G. K. Nutrition and alcoholic Neuroscience and Behavioral Research Portfolio, thickness in alcoholics. Br. J. Addict. 83,
encephalopathies. Acta Med. Scand. 717 NIAAA Research Monograph No. 34 (eds 577–580 (1988).
(Suppl.), 55–65 (1987). Noronha, A., Eckardt, M. & Warren, K.) 437–472 50. Tarnowska-Dziduszko, E., Bertrand, E. &
12. Todd, K. G. & Butterworth, R. F. Early microglial (NIH, Bethesda, 2000). Szpak, G. Morphological changes in the corpus
response in experimental thiamine deficiency: 30. Giancola, P. R. & Moss, H. B. Executive cognitive callosum in chronic alcoholism. Folia
an immunohistochemical analysis. Glia 25, functioning in alcohol use disorders. Recent Dev. Neuropathol. 33, 25–29 (1995).
190–198 (1999). Alcohol. 14, 227–251 (1998). 51. Lee, S. T., Jung, Y. M., Na, D. L., Park, S. H. &
13. Lieber, C. S. Relationships between nutrition, 31. Bates, M. E., Bowden, S. C. & Barry, D. Kim, M. Corpus callosum atrophy in Wernicke’s
alcohol use, and liver disease. Alcohol Res. Neurocognitive impairment associated with encephalopathy. J. Neuroimaging 15, 367–372
Health 27, 220–231 (2003). alcohol use disorders: implications for (2005).
14. Thomson, A. D. Mechanisms of vitamin treatment. Exp. Clin. Psychopharmacol. 10, 52. Harper, C. et al. The pathophysiology of ‘brain
deficiency in chronic alcohol misusers and the 193–212 (2002). shrinkage’ in alcoholics structural and molecular
development of the Wernicke–Korsakoff 32. Oscar-Berman, M. & Marinkovic, K. Alcohol: changes and clinical implications. Alcohol. Clin.
syndrome. Alcohol Alcohol. 35 (Suppl.), 2–7 effects on neurobehavioral functions and the Exp. Res. 29, 1106–1115 (2005).
(2000). brain. Neuropsychol. Rev. 17, 239–257 (2007). 53. Harding, A., Halliday, G., Caine, D. & Kril, J.
15. Martin, P. R., Singleton, C. K. & Hiller- 33. Sullivan, E. V., Harris, R. A. & Pfefferbaum, A. Degeneration of anterior thalamic nuclei
Sturmhofel, S. The role of thiamine deficiency in Alcohol’s effects on brain and behavior. Alco. differentiates alcoholics with amnesia. Brain
alcoholic brain disease. Alcohol Res. Health 27, Res. Health 33, 127–143 (2010). 123, 141–154 (2000).
134–142 (2003). 34. Green, A. et al. The effect of moderate to heavy 54. Cullen, K. M., Halliday, G. M., Caine, D. &
16. Galvin, R. et al. EFNS guidelines for diagnosis, alcohol consumption on neuropsychological Kril, J. J. The nucleus basalis (Ch4) in the
therapy and prevention of Wernicke performance as measured by the repeatable alcoholic Wernicke–Korsakoff syndrome:
encephalopathy. Eur. J. Neurol. 17, 1408–1418 battery for the assessment of reduced cell number in both amnesic and non-
(2010). neuropsychological status. Alcohol. Clin. Exp. amnesic patients. J. Neurol. Neurosurg.
17. Harper, C. The neuropathology of alcohol-specific Res. 34, 443–450 (2010). Psychiatry 63, 315–320 (1997).
brain damage, or does alcohol damage the 35. Bowden, S. C. Separating cognitive impairment 55. Baker, K., Harding, A., Halliday, G., Kril, J. &
brain? J. Neuropathol. Exp. Neurol. 57, 101–110 in neurologically asymptomatic alcoholism from Harper, C. Neuronal loss in functional zones of
(1998). Wernicke–Korsakoff Syndrome: is the the cerebellum of chronic alcoholics with and
18. Butters, N. The Wernicke–Korsakoff syndrome: neuropsychological distinction justified? Psychol. without Wernicke’s encephalopathy.
a review of psychological, neuropathological and Bull. 107, 355–366 (1990). Neuroscience 91, 429–438 (1999).
etiological factors. Curr. Alcohol. 8, 205–232 36. Squire, L. & Butters, N. (eds) Neuropsychology of 56. Harper, C. & Kril, J. An introduction to alcohol-
(1981). Memory 2nd edn (Guilford Press, New York, induced brain damage and its causes. Alcohol
19. Feinberg, I., Fein, G., Price, L. J., Jernigan, T. L. & 1992). Alcohol. 2 (Suppl.), 237–243 (1994).
Floyd, T. C. in Aging in the 1980s: Psychological 37. Pitel, A. L. et al. Episodic and working memory 57. Kril, J. J., Halliday, G. M., Svoboda, M. D. &
Issues (ed. Poon, L. W.) 71–77 (American deficits in alcoholic Korsakoff patients: Cartwright, H. The cerebral cortex is damaged in
Psychological Association, Washington D. C., the continuity theory revisited. Alcohol. Clin. Exp. chronic alcoholics. Neuroscience 79, 983–998
1980). Res. 32, 1229–1241 (2008). (1997).
20. Butters, N. & Brandt, J. in Recent Developments 38. Sullivan, E. V. et al. Disruption of frontocerebellar 58. Harper, C. & Corbett, D. Changes in the basal
in Alcoholism Vol. 3 (ed. Galanter, M.) 207–226 circuitry and function in alcoholism. Alcohol. Clin. dendrites of cortical pyramidal cells from
(Plenum Publishing, New York, 1985). Exp. Res. 27, 301–309 (2003). alcoholic patients—a quantitative Golgi study.

NATURE REVIEWS | NEUROLOGY VOLUME 7  |  MAY 2011  |  291


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

J. Neurol. Neurosurg. Psychiatry 53, 856–861 75. Estruch, R. et al. Atrophy of the corpus callosum 92. Pfefferbaum, A. et al. Longitudinal changes in
(1990). in chronic alcoholism. J. Neurol. Sci. 146, magnetic resonance imaging brain volumes
59. McMullen, P. A., Saint-Cyr, J. A. & Carlen, P. L. 145–151 (1997). in abstinent and relapsed alcoholics. Alcohol.
Morphological alterations in the rat CA1 76. Sullivan, E. V. et al. Cerebellar volume deficits Clin. Exp. Res. 19, 1177–1191 (1995).
hippocampal pyramidal cell dendrites resulting and neuropsychological function in alcoholics 93. Gazdzinski, S., Durazzo, T. C. & Meyerhoff, D. J.
from chronic ethanol consumption and [abstract]. Alcohol. Clin. Exp. Res. 22, 63A (1998). Temporal dynamics and determinants of whole
withdrawal. J. Comp. Neurol. 225, 111–118 77. Agartz, I., Momenan, R., Rawlings, R. R., brain tissue volume changes during recovery
(1984). Kerich, M. J. & Hommer, D. W. Hippocampal from alcohol dependence. Drug Alcohol Depend.
60. Harding, A. J., Halliday, G. M., Ng, J. L., volume in patients with alcohol dependence. 78, 263–273 (2005).
Harper, C. G. & Kril, J. J. Loss of vasopressin- Arch. Gen. Psychiatry 56, 356–363 (1999). 94. Sullivan, E. V. & Marsh, L. Hippocampal volume
immunoreactive neurons in alcoholics is dose- 78. Sullivan, E. V., Marsh, L., Mathalon, D. H., deficits in alcoholic Korsakoff’s syndrome.
related and time-dependent. Neuroscience 72, Lim, K. O. & Pfefferbaum, A. Anterior Neurology 61, 1716–1719 (2003).
699–708 (1996). hippocampal volume deficits in nonamnesic, 95. Kim, E. et al. Mammillothalamic functional
61. Harper, C., Dixon, G., Sheedy, D. & Garrick, T. aging chronic alcoholics. Alcohol. Clin. Exp. Res. connectivity and memory function in Wernicke’s
Neuropathological alterations in alcoholic 19, 110–122 (1995). encephalopathy. Brain 132, 369–376 (2009).
brains. Studies arising from the New South 79. Blansjaar, B., Vielvoye, G., van Dijk, J. & 96. Schulte, T., Sullivan, E. V., Muller-Oehring, E. M.,
Wales Tissue Resource Centre. Prog. Rijnders, R. Similar brain lesions in alcoholics Adalsteinsson, E. & Pfefferbaum, A. Corpus
Neuropsychopharmacol. Biol. Psychiatry 27, and Korsakoff patients: MRI, psychometric and callosal microstructural integrity influences
951–961 (2003). clinical findings. Clin. Neurol. Neurosurg. 93, interhemispheric processing: a diffusion tensor
62. Harding, A. J., Wong, A., Svoboda, M., Kril, J. J. & 197–203 (1992). imaging study. Cereb. Cortex 15, 1384–1392
Halliday, G. M. Chronic alcohol consumption 80. O’Neill, J., Cardenas, V. A. & Meyerhoff, D. J. (2005).
does not cause hippocampal neuron loss in Effects of abstinence on the brain: quantitative 97. Pfefferbaum, A., Adalsteinsson, E. &
humans. Hippocampus 7, 78–87 (1997). magnetic resonance imaging and magnetic Sullivan, E. V. Supratentorial profile of white
63. Baker, K. G., Halliday, G. M., Kril, J. J. & resonance spectroscopic imaging in chronic matter microstructural integrity in recovering
Harper, C. G. Chronic alcoholics without alcohol abuse. Alcohol. Clin. Exp. Res. 25, alcoholic men and women. Biol. Psychiatry 59,
Wernicke–Korsakoff syndrome or cirrhosis do 1673–1682 (2001). 364–372 (2006).
not lose serotonergic neurons in the dorsal 81. Pfefferbaum, A., Adalsteinsson, E. & 98. Pfefferbaum, A., Adalsteinsson, E. &
raphe nucleus. Alcohol. Clin. Exp. Res. 20, 61–66 Sullivan, E. V. Dysmorphology and Sullivan, E. V. Dysmorphology and
(1996). microstructural degradation of the corpus microstructural degradation of the corpus
64. Jernigan, T. L. et al. Reduced cerebral grey callosum: interaction of age and alcoholism. callosum: interaction of age and alcoholism.
matter observed in alcoholics using magnetic Neurobiol. Aging 27, 994–1009 (2006). Neurobiol. Aging 27, 994–1009 (2006).
resonance imaging. Alcohol. Clin. Exp. Res. 15, 82. Schroth, G., Naegele, T., Klose, U., Mann, K. & 99. Sullivan, E. V. Compromised pontocerebellar and
418–427 (1991). Petersen, D. Reversible brain shrinkage in cerebellothalamocortical systems: speculations
65. Sullivan, E. V. et al. In vivo mammillary body abstinent alcoholics, measured by MRI. on their contributions to cognitive and motor
volume deficits in amnesic and nonamnesic Neuroradiology 30, 385–389 (1988). impairment in nonamnesic alcoholism. Alcohol.
alcoholics. Alcohol. Clin. Exp. Res. 23, 83. Carlen, P. L., Wilkinson, D. A., Wortzman, G. & Clin. Exp. Res. 27, 1409–1419 (2003).
1629–1636 (1999). Holgate, R. Partially reversible cerebral atrophy 100. Leiner, H. C., Leiner, A. L. & Dow, R. S. Cognitive
66. Sheedy, D., Lara, A., Garrick, T. & Harper, C. Size and functional improvement in recently abstinent and language functions of the human
of mamillary bodies in health and disease: alcoholics. Can. J. Neurol. Sci. 11, 441–446 cerebellum. Trends Neurosci. 16, 444–447
useful measurements in neuroradiological (1984). (1993).
diagnosis of Wernicke’s encephalopathy. Alcohol. 84. Schweinsburg, B. C. et al. Chemical pathology in 101. Chanraud, S. et al. Diffusion tensor tractography
Clin. Exp. Res. 23, 1624–1628 (1999). brain white matter of recently detoxified in mesencephalic bundles: relation to mental
67. Shear, P. K., Sullivan, E. V., Lane, B. & alcoholics: a 1H magnetic resonance flexibility in detoxified alcohol-dependent
Pfefferbaum, A. Mammillary body and cerebellar spectroscopy investigation of alcohol-associated subjects. Neuropsychopharmacology 34,
shrinkage in chronic alcoholics with and without frontal lobe injury. Alcohol. Clin. Exp. Res. 25, 1223–1232 (2009).
amnesia. Alcohol. Clin. Exp. Res. 20, 1489–1495 924–934 (2001). 102. Zahr, N. M., Pitel, A. L., Chanraud, S. &
(1996). 85. Fein, G., Meyerhoff, D. J. & Weiner, M. W. Sullivan, E. V. Contributions of studies on alcohol
68. Jernigan, T. L., Schafer, K., Butters, N. Magnetic resonance spectroscopy of the brain in use disorders to undrestanding cerebellar
& Cermak, L. S. Magnetic resonance imaging alcohol abuse. Alcohol Health Res. World 19, function. Neuropsychol. Rev. 20, 280–289
of alcoholic Korsakoff patients. 3056–3314 (1995). (2010).
Neuropsychopharmacology 4, 175–186 (1991). 86. Jagannathan, N. R., Desai, N. G. & 103. Parks, M. H. et al. Longitudinal brain metabolic
69. Lenz, V. et al. Value of MRI findings in Raghunathan, P. Brain metabolite changes in characterization of chronic alcoholics with proton
Gayet–Wernicke encephalopathy [French]. alcoholism: An in vivo proton magnetic magnetic resonance spectroscopy. Alcohol. Clin.
J. Neuroradiol. 29, 153–160 (2002). resonance spectroscopy (MRS) study. Magn. Exp. Res. 26, 1368–1380 (2002).
70. Pfefferbaum, A. et al. Brain gray and white matter Reson. Imaging 14, 553–557 (1996). 104. Schmahmann, J. & Sherman, J. The cerebellar
volume loss accelerates with aging in chronic 87. Seitz, D. et al. Localized proton magnetic cognitive affective syndrome. Brain 121,
alcoholics: a quantitative MRI study. Alcohol. resonance spectroscopy of the cerebellum in 561–579 (1998).
Clin. Exp. Res. 16, 1078–1089 (1992). detoxifying alcoholics. Alcohol. Clin. Exp. Res. 23, 105. Meyerhoff, D. J. Brain spectroscopic imaging,
71. Pfefferbaum, A., Sullivan, E. V., Mathalon, D. H. & 158–163 (1999). morphometry, and cognition in social drinkers
Lim, K. O. Frontal lobe volume loss observed 88. Durazzo, T. C., Gazdzinski, S., Rothlind, J. C., and recovering alcoholics. Alcohol. Clin. Exp. Res.
with magnetic resonance imaging in older Banys, P. & Meyerhoff, D. J. Brain metabolite 29, 153–154 (2005).
chronic alcoholics. Alcohol. Clin. Exp. Res. 21, concentrations and neurocognition during short- 106. Sullivan, E. V., Rosenbloom, M. J., Lim, K. O.
521–529 (1997). term recovery from alcohol dependence: & Pfefferbaum, A. Longitudinal changes in
72. Cardenas, V. A., Studholme, C., Gazdzinski, S., preliminary evidence of the effects of concurrent cognition, gait, and balance in abstinent and
Durazzo, T. C. & Meyerhoff, D. J. Deformation- chronic cigarette smoking. Alcohol. Clin. Exp. Res. relapsed alcoholic men: relationships to
based morphometry of brain changes in alcohol 30, 539–551 (2006). changes in brain structure. Neuropsychology 14,
dependence and abstinence. Neuroimage 34, 89. Martin, P. R. et al. Brain proton magnetic 178–188 (2000).
879–887 (2007). resonance spectroscopy studies in recently 107. Rosenbloom, M. J., Pfefferbaum, A. &
73. Pfefferbaum, A. et al. Increase in brain abstinent alcoholics. Alcohol. Clin. Exp. Res. 19, Sullivan, E. V. Recovery of short-term memory
cerebrospinal fluid volume is greater in older 1078–1082 (1995). and psychomotor speed but not postural
than in younger alcoholic patients: a replication 90. Bartsch, A. J. et al. Manifestations of early brain stability with long-term sobriety in alcoholic
study and CT/MRI comparison. Psychiatry Res. recovery associated with abstinence from women. Neuropsychology 18, 589–597 (2004).
50, 257–274 (1993). alcoholism. Brain 130, 36–47 (2007). 108. Shatz, C. J. MHC class I: an unexpected role in
74. Pfefferbaum, A., Lim, K. O., Desmond, J. E. & 91. Shear, P. K., Jernigan, T. L. & Butters, N. neuronal plasticity. Neuron 64, 40–45 (2009).
Sullivan, E. V. Thinning of the corpus callosum in Volumetric magnetic resonance imaging 109. Luna-Medina, R. et al. NP031112, a
older alcoholic men: a magnetic resonance quantification of longitudinal brain changes in thiadiazolidinone compound, prevents
imaging study. Alcohol. Clin. Exp. Res. 20, abstinent alcoholics. Alcohol. Clin. Exp. Res. 18, inflammation and neurodegeneration under
752–757 (1996). 172–176 (1994). excitotoxic conditions: potential therapeutic role

292  |  MAY 2011  |  VOLUME 7  www.nature.com/nrneurol


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

in brain disorders. J. Neurosci. 27, 5766–5776 128. Sokolov, B. P., Jiang, L., Trivedi, N. S. & Aston, C. pathway? Acta Paediatr. Suppl. 404, 9–13
(2007). Transcription profiling reveals mitochondrial, (1994).
110. He, J. & Crews, F. T. Increased MCP‑1 and ubiquitin and signaling systems abnormalities in 146. Whipple, S. C. & Noble, E. P. Personality
microglia in various regions of the human postmortem brains from subjects with a history characteristics of alcoholic fathers and their
alcoholic brain. Exp. Neurol. 210, 349–358 of alcohol abuse or dependence. J. Neurosci. sons. J. Stud. Alcohol 52, 331–337 (1991).
(2008). Res. 72, 756–767 (2003). 147. Blum, K. et al. Reward deficiency syndrome:
111. Barres, B. A. The mystery and magic of glia: 129. Weimbs, T. & Stoffel, W. Proteolipid protein (PLP) a biogenetic model for the diagnosis and
a perspective on their roles in health and of CNS myelin: positions of free, disulfide- treatment of impulsive, addictive, and
disease. Neuron 60, 430–440 (2008). bonded, and fatty acid thioester-linked cysteine compulsive behaviors. J. Psychoactive Drugs 32
112. Cook, C. C., Hallwood, P. M. & Thomson, A. D. residues and implications for the membrane (Suppl. i–iv), 1–112 (2000).
B vitamin deficiency and neuropsychiatric topology of PLP. Biochemistry 31, 12289–12296 148. Mathalon, D. H., Pfefferbaum, A., Lim, K. O.,
syndromes in alcohol misuse. Alcohol Alcohol. (1992). Rosenbloom, M. J. & Sullivan, E. V. Compounded
33, 317–336 (1998). 130. Boison, D. & Stoffel, W. Disruption of the brain volume deficits in schizophrenia–
113. Butterworth, R. F., Kril, J. J. & Harper, C. G. compacted myelin sheath of axons of the central alcoholism comorbidity. Arch. Gen. Psychiatry 60,
Thiamine-dependent enzyme changes in the nervous system in proteolipid protein-deficient 245–252 (2003).
brains of alcoholics—relationship to the mice. Proc. Natl Acad. Sci. USA 91, 149. Sullivan, E. V. et al. Contribution of alcohol abuse
Wernicke–Korsakoff syndrome. Alcohol. Clin. Exp. 11709–11713 (1994). to cerebellar volume deficits in men with
Res. 17, 1084–1088 (1993). 131. Lewohl, J. M., Dodd, P. R., Mayfield, R. D. & schizophrenia. Arch. Gen. Psychiatry 57,
114. Morgan, M. Y. Alcohol and nutrition. Br. Med. Bull. Harris, R. A. Application of DNA microarrays to 894–902 (2000).
38, 21–29 (1982). study human alcoholism. J. Biomed. Sci. 8, 150. Krystal, J. H. et al. The vulnerability to alcohol
115. Tallaksen, C. M., Bøhmer, T. & Bell, H. Blood and 28–36 (2001). and substance abuse in individuals diagnosed
serum thiamin and thiamin phosphate esters 132. Matsuda-Matsumoto, H., Iwazaki, T., with schizophrenia. Neurotox. Res. 10, 235–252
concentrations in patients with alcohol Kashem, M. A., Harper, C. & Matsumoto, I. (2006).
dependence syndrome before and after thiamin Differential protein expression profiles in the 151. Cargiulo, T. Understanding the health impact of
treatment. Alcohol. Clin. Exp. Res. 16, 320–325 hippocampus of human alcoholics. Neurochem. alcohol dependence. Am. J. Health Syst. Pharm.
(1992). Int. 51, 370–376 (2007). 64, S5–S11 (2007).
116. Harper, C., Rodriguez, M., Gold, J. & Perdices, M. 133. Alexander-Kaufman, K., Harper, C., Wilce, P. & 152. Ceballos, N. A., Nixon, S. J., Phillips, J. A. &
The Wernicke–Korsakoff syndrome in Sydney—a Matsumoto, I. Cerebellar vermis proteome of Tivis, R. Semantic processing in alcoholics with
prospective necropsy study. Med. J. Aust. 149, chronic alcoholic individuals. Alcohol. Clin. Exp. and without antisocial symptomatology. J. Stud.
718 (1988). Res. 31, 1286–1296 (2007). Alcohol 64, 286–291 (2003).
117. Blass, J. P. & Gibson, G. E. Abnormality of a 134. Alexander-Kaufman, K., Dedova, I., Harper, C. & 153. Grant, B. F. et al. Co-occurrence of 12-month
thiamine-requiring enzyme in patients with Matsumoto, I. Proteome analysis of the alcohol and drug use disorders and personality
Wernicke–Korsakoff syndrome. N. Engl. J. Med. dorsolateral prefrontal region from healthy disorders in the United States: results from the
297, 1367–1370 (1977). individuals. Neurochem. Int. 51, 433–439 national epidemiologic survey on alcohol and
118. Mukherjee, A. B. et al. Transketolase abnormality (2007). related conditions. Arch. Gen. Psychiatry 61,
in cultured fibroblasts from familial chronic 135. Alexander-Kaufman, K., James, G., Sheedy, D., 361–368 (2004).
alcoholic men and their male offspring. J. Clin. Harper, C. & Matsumoto, I. Differential protein 154. Kessler, R. C., Chiu, W. T., Demler, O.,
Invest. 79, 1039–1043 (1987). expression in the prefrontal white matter of Merikangas, K. R. & Walters, E. E. Prevalence,
119. Nixon, P. F., Kaczmarek, M. J., Tate, J., Kerr, R. A. human alcoholics: a proteomics study. Mol. severity, and comorbidity of 12-month DSM-IV
& Price, J. An erythrocyte transketolase Psychiatry 11, 56–65 (2006). disorders in the national comorbidity survey
isoenzyme pattern associated with the 136. Kashem, M. A., James, G., Harper, C., Wilce, P. & replication. Arch. Gen. Psychiatry 62, 617–627
Wernicke–Korsakoff syndrome. Eur. J. Clin. Invest. Matsumoto, I. Differential protein expression in (2005).
14, 278–281 (1984). the corpus callosum (splenium) of human 155. New South Wales Tissue Resource Centre.
120. Coy, J. F. et al. Molecular cloning of tissue- alcoholics: a proteomics study. Neurochem. Int. ‘Brain Bank’ [online], http://sydney.edu.au/
specific transcripts of a transketolase-related 50, 450–459 (2007). medicine/pathology/trc/index.php (2010).
gene: implications for the evolution of new 137. Kashem, M. A., Harper, C. & Matsumoto, I. 156. Harper, C. et al. How important are brain banks
vertebrate genes. Genomics 32, 309–316 Differential protein expression in the corpus for alcohol research? Alcohol. Clin. Exp. Res. 27,
(1996). callosum (genu) of human alcoholics. 310–323 (2003).
121. Guerrini, I. et al. Direct genomic PCR sequencing Neurochem. Int. 53, 1–11 (2008). 157. Pfefferbaum, A., Sullivan, E. V., Adalsteinsson, E.,
of the high affinity thiamine transporter 138. Lishman, W. A. Cerebral disorder in alcoholism: Garrick, T. & Harper, C. Postmortem MR imaging
(SLC19A2) gene identifies three genetic variants syndromes of impairment. Brain 104, 1–20 of formalin-fixed human brain. Neuroimage 21,
in Wernicke Korsakoff syndrome (WKS). Am. (1981). 1585–1595 (2004).
J. Med. Genet. B Neuropsychiatr. Genet. 137B, 139. Crabbe, J. C. Alcohol and genetics: new models. 158. Dedova, I. et al. The importance of brain banks
17–19 (2005). Am. J. Med. Genet. 114, 969–974 (2002). for molecular neuropathological research:
122. Loh, E. W. et al. Association between variants at 140. Chen, Y. C. et al. Pharmacokinetic and the New South Wales Tissue Resource Centre
the GABAAβ2, GABAAα6 and GABAAγ2 gene pharmacodynamic basis for overcoming experience. Int. J. Mol. Sci. 10, 366–384 (2009).
cluster and alcohol dependence in a Scottish acetaldehyde-induced adverse reaction in Asian 159. Garrick, T., Azizi, L., Merrick, J. & Harper, C. Brain
population. Mol. Psychiatry 4, 539–544 (1999). alcoholics, heterozygous for the variant donation for research, what do people say?
123. Singleton, C. K. & Martin, P. R. Molecular ALDH2*2 gene allele. Pharmacogenet. Genomics Intern. Med. J. 33, 475 (2003).
mechanisms of thiamine utilization. Curr. Mol. 19, 588–599 (2009). 160. Glaw, X. M. et al. Brain donation: who and why?
Med. 1, 197–207 (2001). 141. Enoch, M. A. Genetic and environmental Cell Tissue Bank 10, 241–246 (2009).
124. Liu, J. et al. Patterns of gene expression influences on the development of alcoholism: 161. Fein, G. & Landman, B. Treated and treatment-
in the frontal cortex discriminate alcoholic resilience vs. risk. Ann. NY Acad. Sci. 1094, naive alcoholics come from different
from nonalcoholic individuals. 193–201 (2006). populations. Alcohol 35, 19–26 (2005).
Neuropsychopharmacology 31, 1574–1582 142. Caspi, A. et al. Role of genotype in the cycle of 162. Nakamura, K. et al. Acetaldehyde adducts in the
(2006). violence in maltreated children. Science 297, brain of alcoholics. Arch. Toxicol. 77, 591–593
125. Mayfield, R. D. et al. Patterns of gene expression 851–854 (2002). (2003).
are altered in the frontal and motor cortices of 143. Foley, D. L. et al. Childhood adversity, 163. Bora, P. S. & Lange, L. G. Molecular mechanism
human alcoholics. J. Neurochem. 81, 802–813 monoamine oxidase a genotype, and risk for of ethanol metabolism by human brain to fatty
(2002). conduct disorder. Arch. Gen. Psychiatry 61, acid ethyl esters. Alcohol. Clin. Exp. Res. 17,
126. Iwamoto, K. et al. Decreased expression of NEFH 738–744 (2004). 28–30 (1993).
and PCP4/PEP19 in the prefrontal cortex of 144. Rangaswamy, M. et al. A functional MRI study of 164. Coyle, J. T. & Puttfarcken, P. Oxidative stress,
alcoholics. Neurosci. Res. 49, 379–385 (2004). visual oddball: evidence for frontoparietal glutamate, and neurodegenerative disorders.
127. Etheridge, N., Lewohl, J. M., Mayfield, R. D., dysfunction in subjects at risk for alcoholism. Science 262, 689–695 (1993).
Harris, R. A. & Dodd, P. R. Synaptic proteome Neuroimage 21, 329–339 (2004). 165. Tsai, G. E. et al. Increased glutamatergic
changes in the superior frontal gyrus and 145. Wiers, R. W., Sergeant, J. A. & Gunning, W. B. neurotransmission and oxidative stress after
occipital cortex of the alcoholic brain. Proteomics Psychological mechanisms of enhanced risk of alcohol withdrawal. Am. J. Psychiatry 155,
Clin. Appl. 3, 730–742 (2009). addiction in children of alcoholics: a dual 726–732 (1998).

NATURE REVIEWS | NEUROLOGY VOLUME 7  |  MAY 2011  |  293


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

166. Ikegami, Y. et al. Increased TUNEL positive cells 173. Mousseau, D. D., Perney, P., Layrargues, G. P. & endotoxin following ethanol treatment.
in human alcoholic brains. Neurosci. Lett. 349, Butterworth, R. F. Selective loss of pallidal J. Neuroinflammation 5, 10 (2008).
201–205 (2003). dopamine D2 receptor density in hepatic 180. Crews, F. T. & Nixon, K. Mechanisms of
167. Brooks, P. J. Brain atrophy and neuronal loss in encephalopathy. Neurosci. Lett. 162, 192–196 neurodegeneration and regeneration in
alcoholism: a role for DNA damage? Neurochem. (1993). alcoholism. Alcohol Alcohol. 44, 115–127
Int. 37, 403–412 (2000). 174. Donaldson, J., LaBella, F. S. & Gesser, D. (2009).
168. Climent, E., Pascual, M., Renau-Piqueras, J. & Enhanced autoxidation of dopamine as a 181. Phillips, S. C. Cytoprotective value of lysine,
Guerri, C. Ethanol exposure enhances cell death possible basis of manganese neurotoxicity. penicillamine, and pyridoxal phosphate against
in the developing cerebral cortex: role of brain- Neurotoxicology 2, 53–64 (1981). the neurotoxicity of acetaldehyde. Toxicol. Appl.
derived neurotrophic factor and its signaling 175. Niemela, O. et al. Antibodies against Pharmacol. 98, 553–560 (1989).
pathways. J. Neurosci. Res. 68, 213–225 (2002). acetaldehyde-modified protein epitopes in 182. Butterworth, R. F. Pathophysiology of alcoholic
169. Fadda, F. & Rossetti, Z. L. Chronic ethanol human alcoholics. Hepatology 7, 1210–1214 brain damage: synergistic effects of ethanol,
consumption: from neuroadaptation to (1987). thiamine deficiency and alcoholic liver disease.
neurodegeneration. Prog. Neurobiol. 56, 176. Albano, E. Alcohol, oxidative stress and free Metab. Brain Dis. 10, 1–8 (1995).
385–431 (1998). radical damage. Proc. Nutr. Soc. 65, 278–290
170. Butterworth, R. F. Hepatic encephalopathy—a (2006).
Acknowledgments
serious complication of alcoholic liver disease. 177. Yokoyama, H. et al. Experimental hepatitis
The authors would like to thank E. V. Sullivan for her
Alcohol Res. Health 27, 143–145 (2003). induced by ethanol after immunization with
invaluable support and advice in preparing this
171. Felipo, V. & Butterworth, R. F. Neurobiology of acetaldehyde adducts. Hepatology 17, 14–19
Review. The authors would also like to thank
ammonia. Prog. Neurobiol. 67, 259–279 (1993).
A. Pfefferbaum for contributing the illustrations.
(2002). 178. Horner, M., Behrens, U. J., Worner, T. &
172. Zahr, N. M. et al. Glutamate and glutamine Lieber, C. S. Humoral immune response to
changes induced by ethanol treatment in the rat acetaldehyde adducts in alcoholic patients. Res. Author contributions
brain detectable at 3T. In Proc. ISMRM 18th Comm. Chem. Pathol. Pharmacol. 54, 3–12 (1996). The authors all contributed to researching data for the
Annual Meeting 917 (Stockholm, Sweden, 179. Qin, L. et al. Increased systemic and brain article, discussion of content and writing. N. M. Zahr
2010). cytokine production and neuroinflammation by reviewed the manuscript before submission.

294  |  MAY 2011  |  VOLUME 7  www.nature.com/nrneurol


View publication stats
© 2011 Macmillan Publishers Limited. All rights reserved

You might also like