Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Handbook of Clinical Neurology, Vol.

100 (3rd series)


Hyperkinetic Movement Disorders
W.J. Weiner and E. Tolosa, Editors
# 2011 Elsevier B.V. All rights reserved

Chapter 2

Huntington’s disease
KAREN E. ANDERSON*
Movement Disorders Division, Department of Neurology, School of Medicine, University of Maryland,
Baltimore, MD, USA

INTRODUCTION GENETICS AND DISEASE MECHANISM


Huntington’s disease (HD) is an autosomal-dominant The genetic abnormality underlying HD is caused by
neurodegenerative disorder caused by the abnormal an abnormal expansion of trinucleotide repeats coding
expansion of a nucleic acid triplet repeat. Chorea for glutamine at the N-terminus of the huntingtin pro-
and dystonia are the most prominent neurological fea- tein. The increase occurs in sequences of cytosine, ade-
tures. Cognitive impairments and psychiatric symp- nine, and guanine (CAG) in exon 1 of the HD gene,
toms are present in virtually all HD patients. These located on chromosome 4. The gene is expressed
behavioral symptoms are often more challenging than throughout the body (Strong et al., 1993). The normal
the movement disorder, increasing caregiver burden. function of huntingtin is unknown. Excitotoxicity,
Treatment is available for many problematic beha- mitochondrial dysfunction, and dysregulation of gene
viors, and can positively impact patient and caregiver expression have all been suggested as pathological
quality of life. mechanisms (Difiglia, 1990; Beal, 1992; Nucifora et al.,
2001). The HD gene is transmitted in an autosomal-
dominant pattern of inheritance; each child of an
EPIDEMIOLOGY
affected individual has a 50% chance of inheriting the
Prevalence of HD is estimated at approximately 5 per gene. Normal individuals have 9–29 CAG repeats. Those
100 000; it is less common among those of non-Euro- who develop clinically apparent symptoms of HD have a
pean ancestry (Hayden et al., 1980; Folstein et al., higher repeat number, usually 39 or over. Expansions
1987). Diagnosis is most often made in the third or and, rarely, contractions, in CAG repeat number in
fourth decade of life, but onset may occur from successive generations occur, and should also be
childhood through the eighth decade (Folstein, 1989; addressed in counseling (Wheeler et al., 2007; Nahhas
Gilstad and Reich, 2003). With the availability of et al., 2009). The CAG repeat number is inversely corre-
genetic testing, the diagnosis of HD is now made lated with age of symptom onset.
more frequently in those who have onset of chorea Genetic counseling at a certified testing program is
in their 60s or even 70s without a known family his- recommended by the Huntington’s Disease Society of
tory of HD. Previously, these patients with later-onset America (HDSA) for those seeking presymptomatic at-
chorea would often have been told they had “senile” risk testing for the HD gene. The HDSA advises against
or “benign” chorea in the absence of a family history. testing for the HD gene in minors. Adverse outcomes,
Late-onset HD may be confused with Parkinson’s dis- such as severe depression and suicide attempts, have
ease, hydrocephalus, or other neurodegenerative con- occurred following genetic testing. Concerns about genetic
ditions in the absence of a known family history of discrimination due to HD family history are also important
HD (Reuter et al., 2000; Caserta and Sullivan, 2009). to address during counseling (Bombard et al., 2009).

*Correspondence to: Karen E. Anderson, M.D., Assistant Professor, Psychiatry & Neurology, Department of Neurology, Move-
ment Disorders Division, University of Maryland, School of Medicine, Baltimore, MD 21201, USA. Tel: 410 328-7810, Fax: 410
328-0167, E-mail: kanderson@psych.umaryland.edu
16 K.E. ANDERSON
The distinctive neuropathological change in HD is the becomes more debilitated, bradykinetic, and rigid. Dysto-
decline in the number of medium spiny striatal neurons. nia worsens as the disease progresses. Late-onset disease
The first neuropathological change in HD is in the asso- is characterized by a slower course. Older patients are
ciative areas of the striatum (Vonsattel and Difiglia, sometimes misdiagnosed with a parkinsonian syndrome
1998; Middleton and Strick, 2000). Caudate changes (Reuter et al., 2000). Patients with juvenile-onset HD,
predominate, but cell death also occurs in the globus whose symptoms began prior to age 20, have a distinctly
pallidus. As the disease advances, widespread atrophy different clinical picture. Parkinsonism is their predomi-
occurs (Vonsattel et al., 1985; Hedreen et al., 1991). nant neurological feature, often accompanied by seizures
and myoclonus. Chorea is minimal, making diagnosis
BRAIN IMAGING FINDINGS challenging in cases lacking a family history, e.g., adop-
More advanced basal ganglia atrophy correlates tions or mistaken paternity. Psychiatric and cognitive
positively with longer CAG repeat length in magnetic res- changes may be the most prominent features, further
onance imaging studies (Harris et al., 1992; Aylward et al., obscuring the diagnosis (Ribaı̈ et al., 2007). Deterioration
1997; Rosas et al., 2001). Generalized atrophy occurs of function in juvenile-onset cases is rapid compared with
(Aylward et al., 1998). Cortical thinning and volume adult-onset disease. Although juvenile cases are usually
reductions in other regions are present even in early stages attributed to CAG expansions greater than 60 repeats,
of HD (Rosas et al., 2002, 2003, 2008). there are reported cases of juvenile-onset HD in indivi-
Striatal neuropathology is evident on structural duals with lower numbers of repeats (Ribaı̈ et al., 2007).
imaging before a clinical diagnosis can be made. The Dysphagia and dysarthria are two common problems
PREDICT-HD study seeks to identify markers of HD in HD. Communication becomes severely impaired in
in individuals who have undergone predictive genetic advanced stages, compounding patient and caregiver frus-
testing, are gene-positive, but have not been given a tration. Dysphagia and choking are common in HD and
clinical diagnosis of HD. In this premanifest popula- worsen as the disease progresses. Impulsivity and cogni-
tion, subtle motor signs were associated with a higher tive impairment often add to this problem, with patients
probability of HD diagnosis and smaller striatal stuffing more food into their mouths than they are reason-
volumes (Aylward et al., 2000; Biglan et al., 2009). ably able to swallow or forgetting to cut food into small
Functional studies show caudate and putamenal hypo- pieces if they are not supervised. Kagel and Leopold
metabolism, along with some frontal metabolic reduc- (1992) studied dysphagia in HD and found many abnorm-
tions that correlate with decline in function (Kuhl et al., alities, including swallow incoordination, repetitive swal-
1982; Young et al., 1986; Berent et al., 1988; Bartenstein lows, prolonged laryngeal elevation, inability to stop
et al., 1997). Positron emission tomography studies of respiration, coughing on foods, and choking on liquids.
dopamine function have found both striatal and cortical Videofluoroscopic swallowing studies confirmed the
receptor abnormalities (Bäckman et al., 1997; Pavese abnormalities, and identified aspiration. Another study
et al., 2003). Longitudinal diffusion tensor imaging found increased solid food dsyphagia in people with HD
studies show that a significant reduction of fractional (Trejo et al., 2004). Lanska et al. (1988) found choking
anisotropy, a measure of white-matter integrity, between to be commonly associated with death in HD. Speech
baseline and follow-up was evident throughout the brain. and swallowing evaluation and treatment are helpful in
In addition, a diffusion tensor imaging indicator of axon managing these symptoms (Bonelli et al., 2004).
stability showed significant longitudinal decreases in HD
subjects. These results suggest there is white-matter COGNITIVE SYMPTOMS
degeneration in HD, and support the concept that axonal
injury may occur in HD. Such changes may serve as Cognitive deficits are prominent symptoms of HD.
future biomarkers in HD (Weaver et al., 2009). Impairment of attention, visuospatial processing, and
memory occur early. HD patients also exhibit deficits
in executive function on tests requiring planning,
NEUROLOGICAL SYMPTOMS
problem solving, and cognitive flexibility, even in early
Abnormal involuntary movements (chorea and dystonia) disease. Decline in cognitive ability relates to the num-
and impairment of gait, saccades, smooth pursuit, dysar- ber of years the patient has been affected by HD and,
thria, and swallowing are seen in all patients. These symp- even more closely, to the severity of motor symptoms,
toms progress over 10–15 years, leading to severe particularly deficits in voluntary movement (Brandt
disability and death. Chorea, the involuntary writhing, et al., 1984; Jason et al., 1997; Klempı́r et al., 2009).
nonstereotyped, nonrepetitive movement that is the hall- Due to the varying definitions of “dementia,” it is not
mark of the disease, typically worsens in the middle surprising that research on the prevalence of dementia
stages of the illness and then decreases as the patient in HD is varied. Dementia prevalence is reported at rates
HUNTINGTON’S DISEASE 17
of 15–95% (Heathfield, 1967; Oliver, 1970). Other work show specific impairment in person-centered spatial
states that 90% of patients with HD develop dementia judgment. For example, people with HD (but not Parkin-
over the course of the illness (Gontkovsky, 1998). This son’s disease) experience difficulty with map reading and
wide variability is also undoubtedly due to differing popu- typically misjudge distances and the relationship of their
lations and cognitive assessments. At least in the early body to walls and other potential obstacles.
stages, HD dementia does not fit the standard Diagnostic
and Statistical Manual IV definition of dementia as a syn- EXECUTIVE MEMORY DEFICITS
drome of acquired intellectual impairment, with memory
Among the most consistently reported cognitive impair-
impairment and an additional cognitive impairment
ments in HD are deficits in executive memory function
(aphasia, apraxia, agnosia, or deficits in executive func-
(Josiassen et al., 1983; Brandt, 1991; Pillon et al., 1991;
tioning; American Psychiatric Association, 1994).
Lange et al., 1995). This domain encompasses perfor-
Since cognitive changes in HD are gradual, any acute
mance in many crucial areas including reasoning,
change in cognition or overall mental status should
planning, judgment, decision-making, attention, learning,
warrant a search for neurological compromise. Due to
memory, flexibility, and timing. Impairment in executive
generalized brain atrophy, which can begin relatively
memory has wide-ranging implications not only on cogni-
early in the condition, clinicians should have a low
tive assessments, but also on practical functions in
threshold for obtaining brain imaging to rule out a sub-
everyday life.
dural hematoma or other intracranial injury after even
HD patients demonstrate impairment on various
mild head trauma if there is a change in mental status.
standard executive function tests, including the Wis-
Cognitive dysfunction is already present in some
consin Card Sorting Test, the Stroop Color Word Test,
individuals who carry the HD gene expansion but do
and the Tower of London (Paulsen et al., 1996a;
not show neurological signs or symptoms. Neuropsy-
Savage, 1997; Watkins et al., 2000). In fact, brief tests
chological testing of these people shows gradual
of executive function have shown utility as tools for
decline in cognitive function, although not all cases
differential diagnosis. For example, the Serial Sevens
follow a steady path of functional loss (Snowden
item on the Mini-Mental Status Exam, and an abbre-
et al., 2003). The insidious nature of the changes adds
viated battery of frontal lobe tests have been demon-
to the difficulty in distinguishing cognitive symptoms
strated to be sensitive to HD cognitive deficits
from the types of deficits any individual may show
(Rothlind and Brandt, 1993; Paulsen et al., 1995). The
if fatigued, depressed, or anxious. Questions about
Montreal Cognitive Assessment, another brief test with
possible early cognitive changes can lead to heightened
an emphasis on executive function, has also been
anxiety in those at risk for HD and should be
found to be sensitive to HD-related cognitive change
addressed during evaluation.
(Nasreddine et al., 2005; Mickes et al., 2008).
Simulated gambling paradigms have shown that HD
Visuospatial deficits patients consistently make fewer advantageous choices
than healthy controls or patients with Parkinson’s
Deficits in the ability to copy simple geometric designs,
disease, even when level of cognitive impairment is
to copy block designs, and put together puzzles are
similar across disease populations (Stout et al., 2001).
evident in HD. Some of these problems reflect motor
Several HD patients indicated that one or more of the
symptoms, but performance is also impaired on
card decks were “bad” but continued to make selections
untimed perceptual tasks that do not depend on motor
from the identified “bad” decks. Deficits on tasks
skills. Visuospatial performance deficits may be some
requiring decision-making may be due to various
of the earliest cognitive changes seen in HD (Josiassen
cognitive impairments, including learning, attention,
et al., 1983; Hodges et al., 1990; Pillon et al., 1991).
inhibition, or diminished appreciation for consequences.
Memory deficits in HD include slowed rates of
HD patients may be less able to learn from feedback
learning and impaired recall (Weingartner et al., 1979;
and to modulate future responses based on prior experi-
Butters, 1984), which improves with cuing (Butters
ence. The impact of these impairments in daily life is
et al., 1988; Massman et al., 1990; Pillon et al., 1991).
far-ranging and severe.
Impairment has been reported in the perception of per-
sonal space (Potegal, 1971). Mohr et al. (1991) examined
ATTENTION AND WORKING MEMORY
whether visuospatial deficits in basal ganglia disease are
a nonspecific function of dementia severity. Findings The initial studies of cognition in HD consistently
suggested that general visuospatial processing capacity found the most severe performance deficits on
is impaired as a nonspecific effect of cognitive decline assessments of attention and working memory, such
in both HD and Parkinson’s disease; only HD patients as Digit Span (Boll et al., 1974; Josiassen et al., 1983;
18 K.E. ANDERSON
Strauss and Brandt, 1986). More recent studies have PSYCHIATRIC SYMPTOMS
emphasized dysfunction in unique aspects of atten-
Psychiatric symptoms may occur at any point in HD,
tion, including resource allocation, set shifting, and
including prior to the onset of motor abnormalities (Duff
vigilance (Hanes et al., 1995; Lange et al., 1995; Lawr-
et al., 2007). Depression is commonly reported as a pro-
ence et al., 1996). Sprengelmeyer et al. (1995) reported
dromal symptom. Subtle personality changes have also
that HD patients are able to maintain alertness with
been reported prior to neurological diagnosis, although
external direction, but fail when internal monitoring
many of these studies are problematic since data were
is required. HD patients perseverate on prior
retrospective. Changes reported include impulsivity, emo-
responses and demonstrate difficulty in revising
tional lability, and lack of empathy for others (Brothers
information regarding task rules during performance
and Meadows, 1955; Heathfield, 1967; Dewhurst et al.,
(Lange et al., 1995). Thompson and others (2010) sug-
1969; Lishman, 1998). Overall prevalence of psychiatric
gest the attentional deficits in HD lead to poor ability
symptoms in HD has been reported from 30% to over
to automate daily tasks, resulting in decreased
70% (van Duijn et al., 2007). Craufurd and colleagues
efficiency.
(2001) found that loss of energy and initiative, poor perse-
Learning and memory deficits verance and quality of work, impaired judgment, poor
self-care, and emotional blunting were all reported with
Even early HD patients show verbal learning deficits high frequency by HD patients. A study of outpatients
on sensitive tests (Hamilton, 1998). The deficit is one using careful evaluation with standard measures and
of encoding and retrieval, because recognition memory information from caregivers found that 98% of HD
is often preserved (Butters et al., 1986; Delis et al., patients had at least one psychiatric symptom, demon-
1991). HD patients perform near normal when a less strating that as more information becomes available and
challenging memory assessment is used, such as offer- evaluations are conducted, most patients are likely to
ing choices of, or recognizing, items. Recognition have some behavioral disturbances (Paulsen et al., 2001).
memory may not be preserved in moderate to Virtually all psychiatric symptoms described in the gen-
advanced disease (Kramer et al., 1988; Lang et al., eral population may be seen in persons with HD, although
2000). Retention is relatively normal in HD as distinct some appear to be more particular to the illness.
from “cortical dementias” such as Alzheimer’s disease Unlike the somewhat predictable progression of
(Butters et al., 1988; Massman et al., 1990; Delis et al., motor and cognitive symptoms, no generalized pattern
1991). HD patients demonstrate no temporal gradient for psychiatric disease has been demonstrated. Apathy
in memory performance; memory performance is is the one exception; it may worsen as the illness
mildly impaired for all periods of their lives (Butters progresses (Levy et al., 1998; Paulsen et al., 2001;
and Albert, 1982). Thompson et al., 2002). CAG repeat length has not been
Skill learning is dependent upon normal function of shown to correlate positively with presence or severity
the basal ganglia. Sequence skill learning deficits may of psychiatric symptoms (Zappacosta et al., 1996;
be due to working memory limitations. Paulsen et al. Berrios et al., 2001; Vassos et al., 2008). Severe psychi-
(1993) reported that skill learning was only impaired atric symptoms are one of the main factors in the
when external immediate feedback was unavailable. decision to institutionalize a patient (Wheelock et al.,
Willingham and his colleagues (1996) showed intact 2003). Since they are often amenable to treatment with
skill learning when visual feedback was available to standard psychopharmacological agents, providing
HD patients, but showed defective skill learning when relief from these symptoms can impact greatly on qual-
patients were required to learn without immediate ity of life for both patients and caregivers.
feedback.
Irritability and aggression
Cognitive deficits prior to diagnosis
Irritability and, at times, accompanying agitation in
In the earliest stages of HD, memory deficits, visuo- HD patients is commonly observed and can be a source
spatial skill decline, and an attention disorder often of significant disability to patients and troublesome to
lead to changes in performance at work or in the home. their caregivers. Uncontrolled irritability can poten-
Performance deficits on neuropsychological testing are tially increase the likelihood of nursing home place-
seen in carriers of the HD CAG expansion before a ment. Irritability is defined as a deficit in filtering
clinical diagnosis can be made (Langbehn et al., environmental stimuli combined with a faulty inhibi-
2007). Further complicating the picture, cognitive tory capacity, leading to impulsive and exaggerated
domains are not affected in a uniform fashion in reactions to common situations. It is reported in 40–
preclinical and early HD (Snowden et al., 2002). 80% of patients (Paulsen et al., 1996b, 2001; Murgod
HUNTINGTON’S DISEASE 19
et al., 2001; van Duijn et al., 2007). Aggression, when it Pharmacological interventions are not particularly
occurs in HD, usually accompanies irritability. Aggres- helpful for apathy. Structuring daily activities may be
sive behavior in HD ranges from threatening verbal useful, as with irritability and aggression. Education of
outbursts to property damage and physical harm to family members about differences between apathy and
others. A study of 960 patients found that over 60% depression, and how to cope with apathetic individuals,
of patients or caregivers reported aggressive patient may be the most useful intervention.
behavior (Marder et al., 2000). Chatterjee et al. (2005)
found that patients report a lower level of irritability
Affective disorders
than that in proxy reports by caregivers. The discrep-
ancy suggests impairment of patient insight with Changes in mood are quite common in HD. Thirty to
respect to this symptom. A retrospective study found 70% of patients report depressed mood (van Duijn
over a third of HD patients in nursing homes were et al., 2007). Depression frequently occurs either with
aggressive (Nance and Sanders, 1996). Irritability and or following onset of motor abnormalities, arguing
hostility measures have been shown to be significantly against simple reactive mood changes (Baxter et al.,
elevated in those who are presymptomatic but known 1992; Paulsen et al., 2001; Berrios et al., 2002).
to carry the HD expansion, compared to those who Suicide rates in HD are increased fourfold compared
are at risk but do not have the expansion (Berrios with the general population. Completed suicide rates
et al., 2002; Kirkwood et al., 2002; Vassos et al., 2007). from 3% to 7% have been reported. Over 25% of patients
Behavioral techniques, such as adherence to a attempt suicide at some point in the illness, particularly in
schedule to minimize the unexpected, may be helpful their fifth or sixth decade (Schoenfeld et al., 1984; Farrer,
(Moskowitz and Marder, 2001). Education of care- 1986). Paulsen et al. (2005a) found rates of depression in
givers in how to identify and avoid situations that trig- HD were twice that in the general population and 10% of
ger irritability, and how to minimize its effects if it patients reported at least one past suicide attempt. Risk
does occur, is extremely important. Combined with factors for suicidal behavior in HD patients are similar
the impulsivity seen in HD, irritability can quickly esca- to those in the general population: depression, access to
late into physical aggression with minimal or no provo- weapons, living alone, childlessness, recent loss (e.g., a
cation. Evaluation to rule out medical illness, delirium, divorce or termination from a job), and being unmarried.
medication toxicity, or physical discomfort should be HD patients who express suicidal ideation should be
conducted in patients whose communication may be evaluated immediately, since risk of suicide attempts
impaired or when a behavioral change is particularly and completion is high. Findings by Paulsen and others
uncharacteristic. Irritability and aggression both (2005b) suggest two critical periods for increased risk
respond to pharmacotherapy in many cases, sometimes of suicide in HD. The first is immediately before receiv-
in combination with behavioral interventions. If medi- ing a formal diagnosis of HD, and the second is when
cations are used to control these behaviors the lowest independence first diminishes.
effective dose should be administered. Exceptions Increased levels of mood symptoms are seen in
may occur in patients who are particularly aggressive; those individuals who choose to undergo genetic test-
individual patients may require high doses of medica- ing for HD (Codori et al., 2004). Suicidal ideation
tion to control these behavioral problems. may occur during the genetic testing process in at-risk
persons, regardless of final outcome (Almqvist et al.,
1999; Robins Wahlin et al., 2000). The involvement of
Apathy
a psychiatrist in the care of those who seek predictive
Apathy, which is decreased motivation, is common in testing is particularly important, due to these issues
HD. Those with mild apathy may not initiate novel (Scourfield et al., 1997).
activities but participate in them if engaged; for The prevalence of mania and hypomania is increased
patients with severe apathy, even routine activities in HD. From the limited data available, mania appears
require prompting. Up to half of all patients had apa- to occur in approximately 5% of patients. Including
thy in various studies (Pflanz et al., 1991; Craufurd hypomania, 10% of patients are affected with these
et al., 2001; Paulsen et al., 2001). It has been shown symptoms (Folstein, 1989).
to increase with duration of illness (Caine and Treatment of mood disorders in HD is particularly
Shoulson, 1983; Levy et al., 1998; Thompson et al., important, given the high rate of suicide. Many
2002). Apathy is often difficult to differentiate from patients will improve greatly with standard doses of
depression, especially as the disease progresses and antidepressants. If irritability or psychosis accompanies
communication diminishes due to dysarthria or the depression, addition of a small dose of an atypical
impaired cognition. neuroleptic can be helpful.
20 K.E. ANDERSON
Anxiety disorders TREATMENT
Anxiety disorders have received little study in HD. Older There is no proven treatment to prevent symptom onset
work describes anxiety as a frequent prodromal symp- or slow disease progression in people with the HD gene
tom (Dewhurst et al., 1969). Newer work demonstrates expansion. Agents such as omega fatty acids and mino-
anxiety is an issue in manifest disease (Craufurd et al., cycline have been studied following promising work in
2001). Obsessive and compulsive symptoms occur with animal models, without definitive benefits in human
high frequency in HD patients (Marder et al., 2000; clinical trials (Huntington Study Group TREND-HD
Anderson et al., 2001; Paulsen et al., 2001). Beglinger Investigators, 2008). In a recent Cochrane review, phar-
and colleagues (2007) found the probability of obsessive macological interventions were appraised and none
and compulsive symptoms is more than three times proved to be effective as a disease-modifying therapy
greater by the time a patient shows clearly manifest dis- for HD (Mestre et al., 2009). There are numerous treat-
ease than in at-risk people with no apparent motor symp- ment trials in progress or about to begin and the hope
toms. The same group also reported obsessive worrying remains that this work will lead to therapies that delay
and checking in at-risk individuals, prior to diagnosis symptom onset or modify the course of manifest HD
compared to normal controls (Beglinger et al., 2008). (see Huntington’s Study Group website for ongoing
Anxiety disorders usually respond to standard treat- trials of creatine, coenzyme Q, ACR-16, and other
ment. A selective serotonin reuptake inhibitor (SSRI) is agents; see Hersch and Rosas, 2008, for recent review
first-line treatment. The tricyclic clomipramine is par- and commentary on neuroprotective trials for HD).
ticularly useful in the treatment of obsessive and com- In 2008, tetrabenazine became the first therapy
pulsive symptoms, but often is poorly tolerated due to approved for symptomatic treatment of HD in the USA.
side-effects. As in the general population, when antide- Tetrabenazine selectively depletes central monoamines
pressants are used, especially SSRIs, higher doses may by reversibly binding to the type 2 vesicular monoamine
be needed to achieve maximal control of these symp- transporter. It has been used in Europe for many years
toms. Augmentation with an atypical neuroleptic may to treat chorea and other movement disorders, and was
be helpful in some cases. There have been no studies shown to be effective in reducing chorea severity in a ran-
of behavior therapy for anxiety in HD. These techni- domized clinical trial in the USA (Huntington Study
ques are highly effective in the general population Group, 2006). Tetrabenazine is not without significant
for treatment of anxiety disorders. side-effects, including sedation, akathisia, depressed
mood, and anxiety. It carries a special Food and Drug
Psychotic symptoms
Administration “black box” warning in the USA; due to
Psychotic symptoms are seen in 3–11% of patients (van its mechanism of action, it may exacerbate underlying
Duijn et al., 2007). Psychosis is more prevalent in psychiatric conditions; its use has been linked to suicidal
patients with younger onset of disease (Paulsen et al., behavior and completed suicide (Huntington Study
2001). Delusions of persecution are probably the most Group, 2006). It is important to stress to patients and
common psychotic symptoms. Most HD patients with families that tetrabenazine has not been shown to modify
psychosis do not meet diagnostic criteria for schizophre- disease course, but is rather a symptomatic treatment
nia (Berrios et al., 2002), although a few HD families for reduction of chorea. It is beneficial in a select group
with schizophrenic-like psychosis have been reported of patients who are functionally or socially impaired
(Tsuang et al., 1998; Corrêa et al., 2006). Delusions by involuntary movements, and requires expertise in
and auditory hallucinations, although rare, are among dosing escalation and monitoring at maintenance
the many psychiatric symptoms in HD associated with dose. Side-effects, including severe depression, can be
nursing home placement (Wheelock et al., 2003). delayed in onset. Individual patient tolerance of side-
Psychosis and paranoia in HD respond to standard effects may decrease as the disease progresses. Since
pharmacotherapy, usually atypical or typical neurolep- side-effects of tetrabenazine can mimic HD symptoms
tics. It is usually preferable to start with an atypical (including worsening psychiatric symptoms, gait and bal-
neuroleptic, due to the more favorable side-effect pro- ance impairment, and sedation), reassessment of treat-
file. Standard, high-potency agents such as haloperidol ment benefits and reduction or discontinuation of
may be the first choice in patients who are in need medication should be considered periodically.
of rapid treatment or would benefit from suppression There are no medications approved for treatment of
of chorea. This is particularly important for patients cognitive decline in HD. Agents used for treatment of
who are agitated and psychotic, which is a dangerous Alzheimer’s disease and other potential treatments for
combination that must be treated swiftly, usually with HD dementia have not been shown to be useful. A
sedating medications. novel antihistamine, latrepirdine (Dimebon), is being
HUNTINGTON’S DISEASE 21
studied by the Huntington Study Group as a possible Aylward EH, Li Q, Stine OC et al. (1997). Longitudinal change
therapy for cognitive decline in HD. in basal ganglia volume in patients with Huntington’s
Treatment of psychiatric symptoms is extremely disease. Neurology 48: 394–399.
important, and can greatly improve quality of life in Aylward EH, Anderson NB, Bylsma FW et al. (1998). Fron-
tal lobe volume in patients with Huntington’s disease.
these patients and reduce the burden on their caregivers.
Neurology 50: 252–258.
Prior reviews and treatment guidelines have summarized
Aylward EH, Codori AM, Rosenblatt A et al. (2000). Rate of
the limited research on treatment of behavioral symp- caudate atrophy in presymptomatic and sympto-
toms in HD (Leroi and Michalon, 1998, Rosenblatt matic stages of Huntington’s disease. Mov Disord 15:
et al., 1999; Anderson and Marder, 2002). There have 552–560.
been few clinical trials of agents to treat psychiatric Bäckman L, Robins-Wahlin TB, Lundin A et al. (1997). Cog-
symptoms, and no medication is approved specifically nitive deficits in Huntington’s disease are predicted by
for treatment of behavioral symptoms in HD. Clinical dopaminergic PET markers and brain volumes. Brain
experience suggests using standard psychiatric medica- 120 (Pt 12): 2207–2217.
tions, sometimes in lower or higher doses than used in Bartenstein P, Weindl A, Spiegel S et al. (1997). Central
the general population. Anderson and Marder (2002) motor processing in Huntington’s disease. A PET study.
Brain 120: 1553–1567.
outlined suggestions for a stepwise approach to evalua-
Baxter LR, Mazziotta JC, Pahl JJ et al. (1992). Psychiatric,
tion and treatment of various problematic behaviors.
genetic, and positron emission tomographic evaluation
Many behavioral interventions are extremely useful, of persons at risk for Huntington’s disease. Arch Gen
especially in later life, such as adherence to a set sched- Psychiatry 49: 148–152.
ule, offering limited choices to avoid frustrating the Beal MF (1992). Does impairment of energy metabolism
patient, and walking away rather than engaging in an result in excitotoxic neuronal death in neurodegenerative
argument with a patient (Moskowitz and Marder, 2001). illnesses? Ann Neurol 31: 119–130.
Other measures to improve quality of life and main- Beglinger LJ, Langbehn DR, Duff K et al. (2007). Probability
tain function in HD patients include physical therapy, of obsessive and compulsive symptoms in Huntington’s
speech and swallowing assessment and therapy, dietary disease. Biol Psychiatry 61: 415–418.
interventions to maintain weight, and modification of Beglinger LJ, Paulsen JS, Watson DB et al. (2008). Obsessive
and compulsive symptoms in prediagnosed Huntington’s
diet to prevent choking. As in all neurodegenerative
disease. J Clin Psychiatry 69: 1758–1765.
illness, maintenance of physical function and indepen-
Berent S, Giordani B, Lehtinen S et al. (1988). Positron
dence at the highest possible level may impact greatly emission tomographic scan investigations of Hunting-
on overall quality of life and mental outlook. Support ton’s disease: cerebral metabolic correlates of cognitive
for families and others involved in a patient’s care is an function. Ann Neurol 23: 541–546.
important component of treatment. This includes assis- Berrios GE, Wagle AC, Markova IS et al. (2001). Psychiatric
tance in finding respite care, and providing referrals for symptoms and CAG repeats in neurologically asymptom-
counseling if mental health issues arise in caregivers. atic Huntington’s disease gene carriers. Psychiatry Res
The challenge of treating a hereditary condition means 102: 217–225.
clinicians must be interested in the well-being of the entire Berrios GE, Wagle AC, Markova IS et al. (2002). Psychiatric
family, rather than the usual focus on a single patient. symptoms in neurologically asymptomatic Huntington’s
disease gene carriers: a comparison with gene negative at
risk subjects. Acta Psychiatr Scand 105: 224–230.
REFERENCES Biglan KM, Ross CA, Langbehn DR et al. (2009). Motor
abnormalities in premanifest persons with Huntington’s
Almqvist EW, Bloch M, Brinkman R et al. (1999). A world- disease: The PREDICT-HD study. Mov Disord 24 (12):
wide assessment of the frequency of suicide, suicide 1763–1772.
attempts or psychiatric hospitalization after predictive test- Boll TJ, Heaton R, Reitan R (1974). Neuropsychological and
ing for Huntington disease. Am J Hum Genet 64: 1293–1304. emotional correlates of Huntington’s disease. Journal of
American Psychiatric Association (1994). Diagnostic and Mental and Nervous Disorders 158: 61–69.
statistical manual of mental disorders (4th ed., text rev.). Bombard Y, Veenstra G, Friedman JM et al. (2009). Canadian
Washington, DC. Respond-HD Collaborative Group. Perceptions of genetic
Anderson KE, Marder KM (2002). Huntington’s Disease. In: discrimination among people at risk for Huntington’s dis-
RT Johnson, JW Griffin, JC McArthur (Eds.), Current Ther- ease: a cross sectional survey. BMJ 338: b2175.
apy in Neurologic Disease. 6th Edn. Mosby, Philadelphia, Bonelli RM, Hofmann P (2004). A review of the treatment
USA, pp. 282–287. options for Huntington’s disease. Expert Opin Pharmac-
Anderson KE, Louis ED, Stern Y et al. (2001). Cognitive other 5 (4): 767–776.
correlates of obsessive and compulsive symptoms in Brandt J (1991). Cognitive impairments in Huntington’s
Huntington’s disease. Am J Psychiatry 158: 799–801. disease: insights into the neuropsychology of the striatum.
22 K.E. ANDERSON
In: S Corkin, J Grafman, F Boller (Eds.), Handbook of Folstein SE, Chase GA, Wahl WE et al. (1987). Huntington’s
Neuropsychology, Vol. 5. Elsevier, Amsterdam. disease in Maryland: clinical aspects of racial variation.
Brandt J, Strauss ME, Larus J et al. (1984). Clinical corre- Am J Hum Genet 41: 168–179.
lates of dementia and disability in Huntington’s disease. Gilstad J, Reich SG (2003). Chorea in an octogenarian.
J Clin Neuropsychol 6: 401–412. Neurologist 9: 165–166.
Brothers CRD, Meadows AW (1955). An investigation Gontkovsky ST (1998). Huntington’s disease: a neuropsy-
of Huntington’s chorea in Victoria. J Ment Sci 101: chological overview. J Cogn Rehab 16: 6–9.
548–563. Hamilton JM (1998). Cognitive, motor, and behavioral corre-
Butters N (1984). The clinical aspects of memory disorders: lates of functional decline in Huntington’s disease. Clini-
contributions from experimental studies of amnesia and cal Psychology. University of California, San Diego.
dementia. J Clin Neuropsychol 6: 17–36. p. 58.
Butters N, Albert MS (1982). Processes underlying failures to Hanes KR, Andrewes DG, Pantelis C (1995). Cognitive
recall remote events. In: LS Cermak (Ed.), Human, Mem- flexibility and complex integration in Parkinson’s
ory, and Amnesia. Erlbaum, Hillsdale, NJ, pp. 257–274. disease, Huntington’s disease, and schizophrenia. J Int
Butters N, Wolfe J, Granholm E et al. (1986). An assessment Neuropsychol Soc 1: 545–553.
of verbal recall, recognition, and fluency abilities in Harris GJ, Pearlson GD, Peyser CE et al. (1992). Putamen
patients with Huntington’s disease. Cortex 22: 11–32. volume reduction on magnetic resonance imaging
Butters N, Salmon DP, Cullum CM et al. (1988). Differentia- exceeds caudate changes in mild Huntington’s disease.
tion of amnesic and demented patients with the Wechsler Ann Neurol 31: 69–75.
Memory Scale-Revised. Clin Neuropsychol 2: 133–148. Hayden MR, MacGregor JM, Beighton PH (1980). The prev-
Caine ED, Shoulson I (1983). Psychiatric syndromes in Hun- alence of Huntington’s chorea in South Africa. S Afr Med
tington’s disease. Am J Psychiatry 140: 728–733. J 2: 193–196.
Caserta MT, Sullivan E (2009). Late-onset Huntington’s Heathfield KW (1967). Huntington’s chorea. Investigation
disease masquerading as normal pressure hydrocephalus. into the prevalence of this disease in the area covered
J Neuropsychiatry Clin Neurosci 21: 97–98. by the North East Metropolitan Regional Hospital Board.
Chatterjee A, Anderson KE, Moskowitz CB et al. Brain 90: 203–232.
(2005). A comparison of self-report and caregiver assess- Hedreen JC, Peyser CE, Folstein SE, Ross CA (1991). Neuro-
ment of depression, apathy, and irritability in Huntington’s nal loss in layers V and VI of cerebral cortex in Hunting-
disease. J Neuropsychiatry Clin Neurosci 17: 378–383. ton’s disease. Neurosci Lett. 133 (2): 257–261.
Codori AM, Slavney PR, Rosenblatt A et al. (2004). Preva- Hersch SM, Rosas HD (2008). Neuroprotection for Hunting-
lence of major depression one year after predictive testing ton’s disease: ready, set, slow. Neurotherapeutics 5:
for Huntington’s disease. Genet Test 8: 114–119. 226–236.
Corrêa BB, Xavier M, Guimarães J (2006). Association of Hodges JR, Salmon DP, Butters N (1990). Differential
Huntington’s disease and schizophrenia-like psychosis impairment of semantic and episodic memory in
in a Huntington’s disease pedigree. Clin Pract Epidemol Alzheimer’s and Huntington’s diseases: A controlled pro-
Ment Health 2: 1. spective study. J Neurol Neurosurg Psychiatry 53: 1089–1095.
Craufurd D, Thompson JC, Snowden JS (2001). Behavioural Huntington Study Group (2006). Tetrabenazine as antichorea
changes in Huntington’s disease. Neuropsychiatry therapy in Huntington disease: a randomized controlled
Neuropsychol Behav Neurol 14: 219–226. trial. Neurology 66: 366–372.
Delis DC, Massman PJ, Butters N et al. (1991). Profiles of Huntington Study Group TREND-HD Investigators (2008).
demented and amnesic patients on the California Verbal Randomized controlled trial of ethyl-eicosapentaenoic
Learning Test: implications for the assessment of mem- acid in Huntington disease: the TREND-HD study. Arch
ory disorders. Psychological Assessment: A Journal of Neurol 65: 1582–1589.
Consulting and Clinical Psychology 3: 19–26. Jason GW, Suchowersky O, Pajurkova EM et al. (1997).
Dewhurst K, Oliver J, Trick KLK et al. (1969). Neuro- Cognitive manifestations of Huntington’s disease in rela-
psychiatric aspects of Huntington’s disease. Confin Neurol tion to genetic structure and clinical onset. Arch Neurol
31: 258–268. 54: 1081–1088.
Difiglia M (1990). Excitotoxic injury of the neostriatum is a Josiassen RC, Curry LM, Mancall EL (1983). Development
model for Huntington’s disease. Trends Neurosci 13: of neuropsychological deficits in Huntington’s disease.
286–289. Arch Neurol 40: 791–796.
Duff K, Paulsen JS, Beglinger LJ et al. (2007). Psychiatric Kagel MC, Leopold NA (1992). Dysphagia in Huntington’s
symptoms in Huntington’s disease before diagnosis: the disease: a 16-year retrospective. Dysphagia 7 (2): 106–114.
predict-HD study. Biol Psychiatry 62: 1341–1346. Kirkwood SC, Siemers E, Viken R et al. (2002). Longitudinal
Farrer LA (1986). Suicide and attempted suicide in Hunting- personality changes among presymptomatic Huntington
ton disease: implications for preclinical testing of persons disease gene carriers. Neuropsychiatry Neuropsychol
at risk. Am J Med Genet 24: 305–311. Behav Neurol 15: 192–197.
Folstein SE (1989). Huntington Disease: A Disorder of Klempı́r J, Klempı́rová O, Stochl J, Spacková N et al.
Families. The Johns Hopkins Press, Baltimore. (2009). The relationship between impairment of
HUNTINGTON’S DISEASE 23
voluntary movements and cognitive impairment in Hun- Murgod UA, Saleem Q, Anand A et al. (2001). A clinical
tington’s disease. J Neurol. 256 (10): 1629–1633. study of patients with genetically confirmed Huntington’s
Kramer JH, Delis DC, Blusewicz MJ et al. (1988). Verbal disease from India. J Neurol Sci 190: 73–78.
memory errors in Alzheimer’s and Huntington’s demen- Nahhas F, Garbern J, Feely S et al. (2009). An intergenera-
tias. Dev Neuropsychol 4: 1–15. tional contraction of a fully penetrant Huntington disease
Kuhl DE, Phelps ME, Markham CH et al. (1982). Cerebral allele to a reduced penetrance allele: interpretation of
metabolism and atrophy in Huntington’s disease deter- results and significance for risk assessment and genetic
mined by 18FDG and computed tomographic scan. Ann counseling. Am J Med Genet A 149A: 732–736.
Neurol 12: 425–434. Nance MA, Sanders G (1996). Characteristics of individuals
Langbehn DR, Paulsen JS (2007). Huntington Study Group. with Huntington disease in long-term care. Mov Disord
Predictors of diagnosis in Huntington disease. Neurology 11: 542–548.
68: 1710–1717. Nasreddine ZS, Phillips NA, Bédirian V et al. (2005). The
Lang CJG, Majer M, Balan P (2000). Recall and recognition Montreal Cognitive Assessment, moca: a brief screening
in Huntington’s disease. Arch Clin Neuropsychology 15: tool for mild cognitive impairment. J Am Geriatr Soc
361–371. 53: 695–699.
Lange KW, Sahakian BJ, Quinn NP et al. (1995). Compari- Nucifora FC Jr., Sasaki M, Peters MF et al. (2001). Interference
son of executive and visuospatial memory function in by huntingtin and atrophin-1 with cbp-mediated transcrip-
Huntington’s disease and dementia of Alzheimer type tion leading to cellular toxicity. Science 291: 2423–2428.
matched for degree of demetia. J Neurol Neurosurg Oliver JE (1970). Huntington’s chorea in Northamptonshire.
Psychiatry 58: 598–606. Br J Psychiatry 116: 241–253.
Lanska DJ, Lanska MJ, Lavine L, Schoenberg BS (1988). Con- Paulsen JS, Butters N, Salmon DP et al. (1993). Prism adap-
ditions associated with Huntington’s disease at death. A tation in Alzheimer’s and Huntington’s disease. Neuro-
case-control study. Arch Neurol. 45 (8): 878–880. psychology 7: 73–81.
Lawrence AD, Sahakian BJ, Hodges JR et al. (1996). Execu- Paulsen JS, Butters N, Sadek JR et al. (1995). Distinct cogni-
tive and mnemonic functions in early Huntington’s tive profiles of cortical and subcortical dementia in
disease. Brain 119: 1633–1645. advanced illness. Neurology 45: 951–956.
Leroi I, Michalon M (1998). Treatment of the psychiatric Paulsen JS, Stout JC, Delapena J et al. (1996a). Frontal
manifestations of Huntington’s disease: a review of the behavioral syndromes in cortical and subcortical demen-
literature. Can J Psychiatry 43: 933–940. tia. Assessment 3: 327–337.
Levy ML, Cummings JL, Fairbanks LA et al. (1998). Apathy Paulsen JS, Como P, Rey G et al. (1996b). The clinical util-
is not depression. J Neuropsychiatry Clin Neurosci 10: ity of the Stroop test in a multicenter study of Hunting-
314–319. ton’s disease. J Int Neuropsychol Soc 2: 35.
Lishman AW (1998). Senile dementias, presenile demen- Paulsen JS, Ready RE, Hamilton JM et al. (2001). Neuropsy-
tias, and pseudodementias. In: AW Lishman (Ed.), chiatric aspects of Huntington’s disease. J Neurol Neuro-
Organic Psychiatry: The Psychological Consequences surg Psych 71: 310–314.
of Cerebral Disorder. Blackwell Science, Oxford, Paulsen JS, Nehl C, Hoth KF et al. (2005a). Depression and
pp. 468–469. stages of Huntington’s disease. J Neuropsychiatry Clin
Marder K, Zhao H, Myers RH et al. (2000). Rate of func- Neurosci 17: 496–502.
tional decline in Huntington’s disease. Huntington Study Paulsen JS, Hoth KF, Nehl C et al. (2005b). Critical periods
Group. Neurology 54: 452–458. of suicide risk in Huntington’s disease. Am J Psychiatry
Massman PJ, Delis DC, Butters N et al. (1990). Are all sub- 162: 725–731.
cortical dementias alike? Verbal learning and memory in Pavese N, Andrews TC, Brooks DJ et al. (2003). Progres-
Parkinson’s and Huntington’s disease patients. J Clin Exp sive striatal and cortical dopamine receptor dysfunction
Neuropsychol 12: 729–744. in Huntington’s disease: a PET study. Brain 126:
Mestre T, Ferreira J, Coelho MM (2009). Therapeutic inter- 1127–1135.
ventions for disease progression in Huntington’s disease. Pflanz S, Besson JAO, Ebmeier KP et al. (1991). The clinical
Cochrane Database Syst Rev, 8 (3): CD006455. manifestation of mental disorder in Huntington’s disease:
Mickes L, Kane A, Jacobson M et al. (2008). A comparison a retrospective case record study of disease progression.
of two brief cognitive screening instruments in Acta Psychiatr Scand 83: 53–60.
Huntington Disease (HD). Neurotherapeutics 5: 37–373. Pillon B, Dubois B, Ploska A et al. (1991). Severity and
Middleton FA, Strick PL (2000). Basal ganglia and cerebel- specificity of cognitive impairment in Alzheimer’s, Hun-
lar loops: motor and cognitive circuits. Brain Res Rev 31: tington’s and Parkinson’s diseases and progressive supra-
236–250. nuclear palsy. Neurology 41: 634–643.
Mohr E, Brouwers P, Claus JJ et al. (1991). Visuospatial cogni- Potegal M (1971). A note on spatial motor deficits in patients
tion in Huntington’s disease. Mov Disord 6: 127–132. with Huntington’s disease: a test of a hypothesis. Neurop-
Moskowitz CB, Marder K (2001). Palliative care for people sychologia 9: 233–235.
with late-stage Huntington’s disease. Neurol Clin 19: Reuter I, Hu MT, Andrews TC et al. (2000). Late onset levo-
849–865. dopa responsive Huntington’s disease with minimal chorea
24 K.E. ANDERSON
masquerading as Parkinson plus syndrome. J Neurol Neu- Thompson JC, Snowden JS, Craufurd D et al. (2002). Behav-
rosurg Psychiatry. 68 (2): 238–241. ior in Huntington’s disease: Dissociating cognition-based
Ribaı̈ P, Nguyen K, Hahn-Barma V et al. (2007). Psychiatric and mood-based changes. J Neuropsychiatry Clin Neu-
and cognitive difficulties as indicators of juvenile Hun- rosci 14: 37–43.
tington disease onset in 29 patients. Arch Neurol Thompson JC, Poliakoff E, Sollom AC et al. (2010). Automatic-
64: 813–819. ity and attention in Huntington’s disease: when two hands
Robins Wahlin TB, Bäckman L, Lundin A et al. (2000). High are not better than one. Neuropsychologia 48 (1): 171–178.
suicidal ideation in persons testing for Huntington’s dis- Trejo A, Tarrats RM, Alonso ME et al. (2004). Nutrition
ease. Acta Neurol Scand 102: 150–161. 20 (2): 192–196.
Rosas HD, Goodman J, Chen YI et al. (2001). Striatal Tsuang D, DiGiacomo L, Lipe H et al. (1998). Familial
volume loss in HD as measured by MRI and the influence aggregation of schizophrenia-like symptoms in Hunting-
of CAG repeat. Neurology 25: 1025–1028. ton’s disease. Am J Med Genet 81: 323–327.
Rosas HD, Liu AK, Hersch S et al. (2002). Regional and van Duijn E, Kingma EM, van der Mast RC (2007). Psycho-
progressive thinning of the cortical ribbon in Hunting- pathology in verified Huntington’s disease gene carriers.
ton’s disease. Neurology 58: 695–701. J Neuropsychiatry Clin Neurosci 19: 441–448.
Rosas HD, Koroshetz WJ, Chen YI et al. (2003). Evidence Vassos E, Panas M, Kladi A et al. (2007). Higher levels of
for more widespread cerebral pathology in early HD: an extroverted hostility detected in gene carriers at risk for
MRI-based morphometric analysis. Neurology 27: Huntington’s disease. Biol Psychiatry 62: 1347–1352.
1615–1620. Vassos E, Panas M, Kladi A et al. (2008). Effect of CAG
Rosas HD, Salat DH, Lee SY et al. (2008). Complexity and repeat length on psychiatric disorders in Huntington’s
heterogeneity: what drives the ever-changing brain in disease. J Psychiatr Res 42: 544–549.
Huntington’s disease? Ann N Y Acad Sci 1147: 196–205. Vonsattel JP, Difiglia M (1998). Huntington’s disease.
Rosenblatt A, Ranen NG, Nance MA et al. (1999). A physi- J Neuropathol Exp Neurol 57: 369–384.
cian’s guide to the management of Huntington’s disease: Vonsattel JP, Myers RH, Stevens TJ et al. (1985). Neuropath-
Huntington’s Disease Society of America. New York. ological classification of Huntington’s disease. J Neuro-
Rothlind JC, Brandt J (1993). A brief assessment of frontal pathol Exp Neurol 44 (6): 559–577.
and subcortical functions in dementia. J Neuropsychiatry Watkins LH, Rogers RD, Lawrence AD et al. (2000). Impaired
Clin Neurosci 5: 73–77. planning but intact decision making in early Huntington’s
Savage CR (1997). Neuropsychology of subcortical demen- disease: implications for specific fronto-striatal pathology.
tias. Psychiatr Clin North Am 20: 911–931. Neuropsychologia 38: 1112–1125.
Schoenfeld M, Myers RH, Cupples LA et al. (1984). Increased Weaver KE, Richards TL, Liang O et al. (2009). Longitudinal
rate of suicide among patients with Huntington’s disease. diffusion tensor imaging in Huntington’s disease. Exp
J Neuro Neurosurg Psychiatry 47: 1283–1287. Neurol 216 (2): 525–529.
Scourfield J, Soldan J, Gray J et al. (1997). Huntington’s dis- Weingartner H, Caine ED, Ebert MH et al. (1979). Imagery,
ease: psychiatric practice in molecular genetic prediction encoding, and retrieval of information from memory:
and diagnosis. Br J Psychiatry 170: 146–149. some specific encoding-retrieval changes in Huntington’s
Snowden JS, Craufurd D, Thompson J et al. (2002). Psycho- disease. J Abnorm Psychol 88: 52–58.
motor, executive, and memory function in preclinical Wheeler VC, Persichetti F, McNeil SM et al. (2007). US-
Huntington’s disease. J Clin Exp Neuropsychol 24: Venezuela Collaborative Research Group. Factors asso-
133–145. ciated with HD CAG repeat instability in Huntington dis-
Snowden JS, Gibbons ZC, Blackshaw A et al. (2003). Social ease. J Med Genet 44 (11): 695–701.
cognition in frontotemporal dementia and Huntington’s Wheelock VL, Tempkin T, Marder K et al. (2003). Predictors
disease. Neuropsychologia 41: 688–701. of nursing home placement in Huntington disease.
Sprengelmeyer R, Lange H, Homberg V (1995). The pattern Neurology 60: 998–1001.
of attentional deficits in Huntington’s disease. Brain 118: Willingham DB, Koroshetz WJ, Peterson EW (1996). Motor
145–152. skills have diverse neural bases: spared and impaired skill
Stout JC, Rodawalt WC, Siemers ER (2001). Risky decision acquisition in Huntington’s disease. Neuropsychology 10:
making in Huntington’s disease. J Int Neuropsychol Soc 315–321.
7: 92–101. Young AB, Penney JB, Starosta-Rubinstein S et al.
Strauss ME, Brandt J (1986). An attempt at presymptomatic (1986). PET scan investigations of Huntington’s disease:
identification of Huntington’s disease with the WAIS. cerebral metabolic correlates of neurological features
J Clin Exp Neuropsychol 8: 210–228. and functional decline. Ann Neurol 20: 296–303.
Strong TV, Tagle DA, Valdes JM et al. (1993). Wide- Zappacosta B, Monza D, Meoni C et al. (1996). Psychiatric
spread expression of the human and rat Huntington’s dis- symptoms do not correlate with cognitive decline, motor
ease gene in brain and nonneural tissues. Nat Genet 5: symptoms, or CAG repeat length in Huntington’s disease.
259–265. Arch Neurol 53: 493–497.

You might also like