You are on page 1of 16

The Role of

Norepinephrine in the
Behavioral and
Psychological Symptoms
of Dementia
Nathan Herrmann, M.D., F.R.C.P.C.
Krista L. Lanctôt, Ph.D.
Lyla R. Khan, Hon.B.Sc.

The behavioral and psychological symptoms of de-


mentia (BPSD) are common serious problems that A notable source of morbidity and mortality, de-
mentia accounts for an excess of health care costs.1
The total economic cost of the disorder, including direct
are a major contributor to caregiver burden. De-
and indirect medical costs, has been estimated to be $100
spite their significance, the underlying neurobiol-
billion per year (in U.S. dollars).2 Alzheimer’s disease
ogy of these disturbances is still unclear. This re-
(AD) is a progressive and incurable disease character-
view examines the role of norepinephrine (NE) on ized by cognitive and behavioral abnormalities and is
BPSD, including depression, aggression, agitation the most common cause of dementia in North America.1
and psychosis. A number of lines of evidence sug- The estimated prevalence of AD among individuals
gest that NE dysfunction leading to BPSD may above 65 years of age in the United States is 10.3%.3
result from increased NE activity and/or hyper- The behavioral abnormalities associated with AD fre-
sensitive adrenoreceptors compensating for loss of quently disrupt patient care and present a management
NE neurons with progression of Alzheimer’s dis- problem to the caregiver. Behavioral and psychological
ease (AD). With greater appreciation of the un- symptoms of dementia (BPSD) that complicate AD in-
derlying neurobiology of behavioral and psycho- clude behaviors with psychotic features (e.g., delusions
logical symptoms of dementia (BPSD) more and hallucinations) as well as those without (e.g., de-
effective, rational, targeted pharmacotherapy will pression, anxiety, agitation, wandering, hostility, and
uncooperativeness).4 Disruptive agitated behaviors are
hopefully emerge.
common and occur in 40% to 90% of patients with AD
(The Journal of Neuropsychiatry and Clinical
at some point during the course of their illnesses.5,6 Evi-
Neurosciences 2004; 16:261–276)
dence suggests that BPSD have a detrimental impact on

Received August 1, 2002; revised October 29, 2002; accepted Novem-


ber 18, 2002. From the Division of Geriatric Psychiatry, Sunnybrook
and Women’s College Health Sciences Centre, Geriatric Psychiatry,
Department of Psychiatry, University of Toronto; HOPE Research Cen-
tre, Sunnybrook and Women’s College Health Sciences Centre, De-
partments of Psychiatry and Pharmacology, University of Toronto.
Address correspondence to Dr. Herrmann, Division of Geriatric Psy-
chiatry, Sunnybrook and Women’s College Health Sciences Centre,
2075 Bayview Avenue, Room FG05, Toronto, ON M4N 3M5, Canada;
Nathan.Herrmann@sw.ca (E-mail).
Copyright 䉷 2004 American Psychiatric Publishing, Inc.

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 261


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

both patients7 and caregivers.8 Aggression occurs in 18% erences from bibliographies of relevant literature. The
to 65% of patients9–11 and is the behavioral disturbance following keywords were used: norepinephrine, nor-
that causes the greatest impact. Behaviors such as agi- adrenergic, Alzheimer’s disease, dementia, behavior,
tation and aggression complicate management12,13 and behavioral disorders, aggression, agitation, psychosis,
contribute to caregiver burden14 and the institutionali- depression, and noncognitive.
zation of demented patients.15–18
The treatment of behavioral disorders in AD has em-
phasized the use of psychotropic medications.19–21 NOREPINEPHRINE SYSTEM
Studies in nursing homes,22–24 hospitals,25 and the com-
munity18 have shown that psychotropic medications The central noradrenergic system is known to have two
are frequently used in elderly patients. Guidelines out- different projections: 1) neurons originating from the
lining the appropriate use of psychotropics in elderly ventrolateral tegmental noradrenergic cells—involved
institutionalized patients have been created in re- in sexual and feeding behaviors35—that flow to the fore-
sponse to persistent psychotropic use.26 The Omnibus brain, an area commonly involved in traumatic brain
Budget Reconciliation Act of 1987 was enacted to cur- injury and associated with violent behavior and reduc-
tail inappropriate and overuse of psychoactive medi- tion in rage control,36 and 2) neurons originating from
cations in long-term care facilities, which often resulted the locus coeruleus (LC), which line the bottom of the
in high incidence of adverse events.27,28 For example, fourth ventricle in the rostral pons37 and are associated
antipsychotic medications, the most commonly used with cognitive functions.38 Noradrenergic neurons give
psychotropic for BPSD, can produce serious extrapyra- rise to diffuse axonal projections that innervate large ar-
midal side effects. Extrapyramidal symptoms such as eas of the brain, including the cerebellum, thalamus, hy-
rigidity, tremors, and parkinsonian gait29are common pothalamus, and midbrain,35 and thus can be expected
with antipsychotic drug administration and may limit to influence many different functions.
treatment in the elderly demented population and lead Norepinephrine (NE) and epinephrine are catechol-
to discontinuation of medication. Psychotropics have amines and are formed from the amino acid L-tyrosine
also been found to induce agitation, confusion, and cog- which is actively transported from the blood. L-Tyrosine
nitive impairment in elderly demented patients,30 fur- is converted to L-dihydroxyphenylacetic acid (L-dopa)
ther contributing to BPSD. Novel therapies targeting through tyrosine hydroxylase, which is the rate limiting
specific neurotransmitters are believed to underlie be- enzyme of the metabolic pathway. L-dihydroxypheny-
havioral dysfunction in AD are being studied as alter- lacetic acid is then transformed to dopamine (DA), the
natives to the modestly efficacious antipsychotics.31,32 immediate precursor of NE, by dopa-decarboxylase and
The idea here is that we can rationalize treatment by transported into storage vesicles. Storage vesicles con-
studying the neurotransmitters involved. This has been taining DA beta hydroxylase (DBH), found in NE-spe-
demonstrated with serotonin, one of the best examined cific neurons, convert the stored DA to NE through b-
neurotransmitters.33,34 hydroxylation. Dopamine producing neurons have
storage vesicles without DBH.
Norepinephrine is the primary neurotransmitter of
METHOD the sympathetic nervous system in the periphery but is
also prevalent in the brain. Epinephrine is an analog of
Neuropathological and neurochemical disruptions in NE and is formed by N-methylation. It is released prin-
the central noradrenergic system have been established cipally from the adrenal medulla; however, its function
as important factors in the etiology of AD and BPSD. in the CNS has not been fully elucidated. Norepineph-
Evidence supporting these neurobiological alterations rine is released from the vesicles into the synaptic cleft
as well as the putative links between the noradrenergic at the occurrence of an action potential. Norepinephrine
system and behavior, including BPSD, are reviewed. transporters on the synaptic terminals take up NE into
Noradrenergic pharmacotherapies are also examined to storage vesicles. Monoamine oxidase (MAO) deami-
assess the clinical significance they may have in AD and nates and, hence, deactivates free NE in the cytosol that
BPSD. Sources used to obtain articles included electronic has not been taken up into the vesicles, leading to the
databases (MEDLINE, EMBASE) and manual cross-ref- formation of the metabolite, 3,4-dihydroxyphenylglycol

262 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


HERRMANN et al.

(DHPG). Dihydroxyphenylglycol is then converted to 3- neuronal system dysfunction is the cholinergic system.
methoxy-4-hydroxyphenylglycol (MHPG) by catechol However, evidence supporting noradrenergic, seroto-
O-methyltransferase (COMT). Residual NE in the syn- nergic, dopaminergic, corticotropin-releasing factor, and
apse is first metabolized by COMT and then MAO to somatostatin system dysfunction has also accumu-
form MHPG and other metabolites. lated,56–58 and mounting evidence supports the presence
Noradrenergic receptors are divided into ␣-adrener- of significant abnormalities in the noradrenergic system
gic and b-adrenergic subtypes, each of which has further in AD.58–78
subtypes: ␣1, ␣2, b1, b2 and b3. The postsynaptic b-ad-
renergic receptors activate Gs type G proteins to stimu-
late adenylyl cyclase, leading to a physiological re- Norepinephrine Concentrations
sponse. The ␣2 adrenergic receptors decrease adenylyl Postmortem studies have consistently shown noradren-
cyclase activity or activate specific KⳭ channels through ergic system involvement in the AD disease process
coupling to Gi and/or Go,39 and are located both pre- with decreased NE levels being recognized in many
and postsynaptically. The ␣1 adrenergic receptor stim- brain areas.58,59,61,62,64–67,71,72,74–78 The majority of studies
ulates the action of phospholipase C through Gx. and it indicate significantly decreased cortical and subcortical
is located on the postsynaptic terminals. Only ␣2 and b2
NE levels in the frontal medial gyrus, temporal superior
adrenoreceptors are located on both presynaptic and
gyrus, cingulate gyrus, hippocampus, amygdala, thala-
postsynaptic terminals. Norepinephrine and epineph-
mus, hypothalamus, caudate, putamen as well as the
rine stimulate both the ␣- and b-adrenergic receptors in
LC.58,59,62,64,66,67,71,75,76 However, some studies did not
the periphery and CNS with the effects, depending on
replicate these results due to possible confounders such
the balance between the ␣- and b-receptors that are pres-
as postmortem delay.
ent and have been activated. Presynaptic ␣2 adrenore-
ceptors modulate the negative feedback of NE release
and are pharmacologically different from postsynaptic Loss of Noradrenergic Neurons
␣2 adrenergic receptors. Norepinephrine release can be A loss of noradrenergic neurons in the LC, the major
enhanced by the stimulation of b2 adrenoreceptors. Epi- noradrenergic source in the brain, has been well estab-
nephrine is more potent than norepinephrine on the b2 lished in patients with AD,59,68–70,73–75,79,80 and has not
adrenoreceptor. been found in other types of dementia such as vascular
Norepinephrine mediates and is modulated by other dementia.59 These changes in the LC may account for a
neurochemicals in the brain, many known to affect be- deficiency in the noradrenergic system in the pathogen-
havior. A loss or alteration in any one neurotransmitter esis of AD.
is likely to affect other neurotransmitters and potentially However, NE concentrations in tissue may not di-
lead to deviations in behavior. The noradrenergic sys- rectly correlate with functional noradrenergic transmis-
tem interacts with acetylcholine,40–43 serotonin,44–46 sion. Rather, noradrenergic turnover and neuronal ac-
DA47–49 and c-aminobutyric acid.50 Other neurotrophic tivity deduced through measurements of relative
factors that interact with and modulate the noradren- concentrations of NE and its predominant intraneuronal
ergic system include a corticotropin releasing hor- metabolite, MHPG, provide a more accurate represen-
mone,51 ,52 somatostatin,53 and neuropeptide Y.54 Many tation of actual noradrenergic function. In postmortem
of these neurotrophic factors have been associated with studies, when both NE and MHPG were quantified to-
behaviors such as aggression, anxiety, and depression in gether, although brain NE was often decreased, MHPG
nondemented populations55 and thus alterations in par- was found to be unchanged or high in AD patients com-
ticular neuropeptides may play a role in aggravating pared to control subjects.58,62,65,67,71,72,81 These findings
already disrupted neurotransmitter systems. Since mul- suggest that neuronal loss may lead to subsequent in-
tiple neurochemical systems are known to be compro- creased NE metabolism and hence increased noradren-
mised in AD,56 they may magnify behavioral distur- ergic activity, possibly as a compensatory mechanism
bances. for LC NE loss. Hoogendijk et al.62 demonstrated a sig-
nificant inverse correlation between the MHPG/NE ra-
NOREPINEPHRINE AND ALZHEIMER’S DISEASE tio and the number of remaining pigmented LC neurons
Alzheimer’s disease is associated with multiple neuro- in AD patients, inferring an overactivity or activation of
transmitter system dysfunction. The most well studied the remaining LC neurons to counterbalance cerebral

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 263


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

NE losses. It remains unclear whether this compensa- vious suppositions that increased MHPG from height-
tory activity occurs in the noradrenergic neuron termi- ened NE turnover, as a result of losses of NE, is
nals in the projection areas, in the LC cell bodies or in correlated with increased cognitive dysfunction.
both.62
Galanin (GAL), a major inhibitory modulator of cho-
Dopamine Beta Hydroxylase (DBH)
linergic and noradrenergic transmission, has been Studies of DBH may further substantiate evidence of
shown to be hyperinnervated in the basal forebrain and noradrenergic dysfunction in AD. Both postmortem and
cortex of AD patients. Galanin fibers are thought to antemortem studies have shown DBH, a noradrenergic
originate in the LC. Miller et al.73 explored the possibil- biological marker, to be reduced in the hippocampus
ity that GAL expression is increased in the LC of patients and frontal and temporal cortices of AD patients when
with AD. Although the authors observed a significant compared to controls74,83,84 and patients with vascular
reduction in the amount of neuromelanin-pigmented, dementia.83,84 However, these results must still be inter-
which identifies NE presence, neurons in the LC of AD preted with caution as the extent to which DBH activity
patients, when investigated alongside age- and sex- is associated with LC neuronal degeneration is still un-
matched control subjects, there was no significant dif- clear.84
ference found between the number of both the pig-
mented and nonpigmented GAL mRNA expressing
neurons in either demented patients or control subjects. Cerebral Spinal Fluid (CSF)
This resulted in a significant increase in the percentage Resting cerebral spinal fluid NE is thought to be gen-
of neuromelanin-pigmented GAL co-expressing cells in erally increased with age, particularly in patients with
patients with AD compared to control subjects, hence, advanced AD, and has been shown to be higher in AD
suggesting preservation or sparing of noradrenergic patients when compared to control subjects.63,85 This is
neurons co-expressing GAL mRNA in a brain area oth- consistent with the proposed compensatory action of
erwise diminished of noradrenergic neurons. These re- noradrenergic neurons. It is essential to note that the
sults indicate a possible neuroprotective feature of GAL, major method of NE clearance from the CSF is through
safeguarding the GAL co-localized group of noradren- NE reuptake by presynaptic noradrenergic terminals.86
ergic neurons from damage resulting from the disease Therefore, it is possible that AD related noradrenergic
process.73 The possibility that augmented GAL fiber in- circuit degeneration may lead to impairment of NE
nervation may be due to the enhanced expression of reuptake resulting in increased CSF NE levels.87 Hence,
GAL within the remaining neurons in the LC region re- measuring CSF NE concentrations may not allow a di-
mains to be investigated further. rect evaluation of CNS noradrenergic dynamics. It is
more important to measure NE precursors and metab-
Link With Severity olites as well CSF NE levels for a comprehensive anal-
There is a well-established link between AD severity ysis of NE activity.88 Moreover, MHPG, unlike NE,
and loss of noradrenergic neurons.69 Bondareff et al.69 crosses the blood-brain barrier freely, hence CSF MHPG
compared total neuronal counts of 19 AD patients com- can provide an indication of neuronal NE metabolism
pared to 10 nondemented subjects and found the group and dispersion of circulating MHPG into CSF. Therefore,
of AD patients could be further divided into two distinct it is imperative that plasma MHPG levels are accounted
groups according to LC neuronal cell counts, which also for in order to accurately interpret CNS noradrenergic
correlated with severity of dementia. The authors re- activity.63 Raskind et al.87 compared levels of NE, its me-
ported an 81% decrease in nucleus LC neurons in the tabolite DHPG, and precursor dopa in both CSF and
group of demented patients with more severe dementia, plasma following administration of yohimbine (a selec-
when compared to control subjects. The group of pa- tive ␣2 adrenergic receptor blocker), clonidine (a selec-
tients with less severe dementia showed a 20% decrease tive ␣2 agonist), and placebo in older subjects with AD,
in LC neuronal loss. NE levels in the brain have also older subjects without AD and younger subjects. The
been found to have an inverse relationship with cogni- results suggested that the increase in CSF NE subse-
tive impairment.61,78 Lawlor et al.82 noted a significant quent to yohimbine administration in both older sub-
positive relationship between basal plasma MHPG lev- jects and AD subjects is not likely due to a reduction in
els and cognitive impairment, further corroborating pre- NE clearance but rather mechanisms that compensate

264 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


HERRMANN et al.

for NE neuron loss through augmented NE biosynthe- which can activate not only the central noradrenergic
sis. system and sympathoadrenal system but may result in
Cerebral spinal fluid NE response to manipulation of behavioral manifestations as well. The LC is involved in
the ␣2 adrenergic receptor by the administration of yo- regulation of levels of arousal, flight-and- fight re-
himbine and clonidine has been studied to evaluate nor- sponses,89,90 agitation, anxiety,89 sleep-wake cycle, and
adrenergic tone, yet a very limited amount of evidence levels of vigilance and emotion70,91,92 as well as aggres-
is available regarding noradrenergic dysfunction. A sive behaviors.93–95 Noradrenergic projections from the
general increase in CSF NE has been seen in response to LC also modulate sympathetic nervous system re-
yohimbine in both AD and older control subjects (with sponse, including blood pressure, pulse rate and danger
a greater increase in the AD subjects), when compared signals. Hence, the noradrenergic system, directly or in-
to younger control subjects.87,89 Results of studies of the directly, has the potential to modify a variety of behav-
CSF NE response to clonidine have been inconsistent, iors through actions in both the cortical and subcortical
with insignificant results predominating. Further re- regions. A disruption of such an integrated system
search into this area is needed to elucidate the value and could potentially lead to abnormal behavioral responses
capacity of CSF catechols as an index of neuronal neu-
to otherwise ordinary stimuli. LC damage has been de-
rotransmission function.
termined in several neurologic disorders that are char-
Compensating for lost neurons results in the tonic ac-
acterized by altered behavior including Parkinson’s dis-
tivation of the noradrenergic system leading to an in-
ease, schizophrenia and depression.70,92
creased noradrenergic response. Mechanisms that may
The link between NE disruption and behavior has
account for this include the following: 1) Neuronal plas-
been studied in a variety of psychiatric disturbances in-
ticity38,89—resulting in the hyperinnervation of certain
cluding depression, anxiety, agitation and aggression. In
brain regions. This may be a result of regenerating neu-
rons and has been observed in rats.38,89 2) Increased NE depression there is evidence of ␣2 receptor dysfunction
synthetic capacity and firing rate in damaged LC neu- including postsynaptic ␣2 receptor downregulation,96–98
rons.38,89 3) Decreased ability of presynaptic ␣2 receptors increased presynaptic ␣2 receptor sensitivity,99 and in-
to inhibit noradrenergic activity.38,89 Studies have indi- creased ␣2 receptor density in the LC.100,101 Decreased
cated a loss of ␣2 receptor inhibition in AD, leading to noradrenergic receptor sensitivity and increased nor-
the tonically activated state of the noradrenergic system adrenergic turnover have been noted in patients with
and increased CSF NE levels.87,89 This increase in com- anxiety,102 generalized anxiety disorder,103 and posttrau-
pensatory activity is usually accompanied by compen- matic stress disorder.104 High levels of NE release have
satory activity in other neurotransmitter systems as been associated with aggression in animals, healthy
well. adults93,105,106 and patients with depression and ma-
Both postmortem and antemortem evidence supports nia.107,108 In animal models, NE enhances aggressive be-
a consistent link between noradrenergic dysfunction haviors106,109 and b-adrenoreceptor blockers have been
and AD (Tables 1 and 2). Postmortem studies provide shown to decrease aggression.106 Finally NE and NE me-
the strongest evidence of noradrenergic degeneration in tabolite levels are positively correlated with aggressive
AD. Although postmortem evidence may provide a de- behaviors in patients with personality disorders.110–112
scriptive representation of the loss of noradrenergic neu- In summary, there is significant evidence of noradren-
rons, postmortem results may not directly reflect the an- ergic system involvement in a variety of psychiatric and
temortem state. Nonetheless, much of the evidence behavioral disorders from animals and nondemented
reviewed reveals decreased levels of both NE and nor- human studies.
adrenergic neurons, while MHPG levels have been
found to be increased, suggesting increased NE metab-
olism and, hence, activity. The fact that this correlates NOREPINEPHRINE AND BEHAVIOR IN
positively with increased AD severity, further validates
DEMENTIA (BPSD)
these findings. As previously mentioned, behavioral problems, particu-
larly agitation and aggression are prevalent in AD. This
NOREPINEPHRINE AND BEHAVIOR has been consistently shown in various studies.113–116 A
The LC is sensitive to both variations in the body’s in- prospective, 10-year, longitudinal study on the change
ternal homeostasis and external environmental stimuli, in aggressive behavior throughout the course of demen-

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 265


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

tia by Keene et al.117 measured physical aggression, ag- adrenergic system has been implicated in the disorder
gressive resistance, physical threats, verbal aggression, of behavior in animals as well as other psychiatric ail-
refusing to speak, destructive behavior and general ir- ments, it has been suggested that NE may likely play a
ritability. The authors reported 96% of the 99 subjects role in the noncognitive behavioral disturbances asso-
with AD/vascular dementia showed profound or con- ciated with AD.118 Agitation, particularly aggression
tinuing aggressive behavior of at least one type during and akathisia, has been linked with increased norepi-
the dementia disease process, with verbal aggression as nephrine activity.119 Noradrenergic system components
the most common behavioral disturbance. As the nor- have been associated with behaviors associated with de-

TABLE 1. Summary of Literature Concerning Changes in NE Neurons and Levels in Patients with AD
Measures and References Finding Comments
NE neurons postmortem
Mann et al. 198259 f neurons (vagus nucleus, LC) 19 patients with AD, eight with MID, versus 21 control
subjects
79
Förstl et al. 1992 f neurons (LC), depressed AD ⬍ nondepressed AD 52 patients with AD; depressed AD (n⳱14); no control
subjects
Bondareff et al. 198269 f neurons (LC) 20 patients with AD versus 10 control subjects
Mann et al. 198468 f neurons (LC, nucleus basalis) 22 patients with AD versus control subjects; AD severity
varied from moderate to severe
Hoogendijk et al. 199570 f neurons (in large neurons), f neurons (in small Five patients with AD, five with Parkinson’s disease, five
neurons) with amyotrophic lateral sclerosis, versus five control
subjects; control subjects concomitant with viral
encephalitis and AIDS, cerebral infarctions without
dementia; diabetes II and liver cirrhosis from chronic
non-A non-B hepatitis
Miller et al. 199973 f neurons (rostral, caudal LC) Six patients with AD versus control subjects
Perry et al. 198180 f neurons (LC) Patients with AD, unipolar depressed, versus control
subjects
Iversen et al. 198374 f neurons (LC) Six patients with AD versus six control subjects
Moll et al. 199075 f neurons (LC) Seven patients with AD versus seven control subjects
Matthews et al. 200278 f neurons (rostral LC, mid LC) 36 patients with AD, 10 with mixed/other dementia, 33
control subjects; concurrently on antidepressant (n⳱1),
sedative hypnotics (n⳱10), neuroleptics (n⳱13) in AD
group; concurrently on antidepressant (n⳱2), sedative
hypnotics (n⳱5), neuroleptics (n⳱3) in mixed/other
dementia group
NE levels postmortem
Mann et al. 198259 f NE (hypothalamus, caudate) 19 patients with AD, eight patients with MID versus 21
control subjects
Adolfsson et al. 1979 f NE (putamen, cortex gyrus frontalis)
61
19 patients with AD versus control subjects
Hoogendijk et al. 199962 f NE (frontal medial gyrus, temporal superior gyrus, 16 patients with AD (depressed n⳱6, transiently
amygdala, thalamus, LC) depressed n⳱5, nondepressed n⳱5) versus control
subjects; comorbid hallucinations (n⳱3), anxiety
(n⳱2), agitation (n⳱4), compulsive eating (n⳱1),
delirium (n⳱2), apathy (n⳱1), chronic psychosis
(n⳱1); clinical assessment by NINCDS-ADRDA, DSM-
III-R, Cornell scale
Arai 198464 f NE (cingulate gyrus, substantia innominata, Four patients with AD versus nine control subjects;
hypothalamus, putamen, medial nucleus of thalamus, concurrent sulpiride (n⳱1), estazolam (n⳱1)
raphe)
Francis et al. 198565 f NE (early and late onset) 48 patients with AD (early onset n⳱19, older onset
n⳱29) versus 34 control subjects
Nazarali et al. 199266 f NE (temporal cortex, amygdala) 13 patients with AD versus 22 control subjects (non-AD
dementia n⳱12, normal n⳱10)
Storga et al. 199667 f NE (amygdala, caudate, substantia nigra, cingulate Eight patients with AD versus six control subjects;
gyri) concurrent neuroleptic use (n⳱6)
Gottfries et al. 198371 f NE (hypothalamus, caudate, hippocampus, cortex of 14 patients with AD versus 16 control subjects;
the cingulate gyrus) comorbidity in AD with mild Parkinson’s disease and
treated with l dopa (n⳱1)
a
NE⳱norepinephrine; LC⳱locus caeruleus; AD⳱Alzheimer’s disease; MID⳱multi-infarct dementia; NINCDS-ADRDA⳱National Institute
of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders Association criteria.

266 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


TABLE 2. Summary of Literature Concerning Changes in NE Metabolites in Patients with ADa
Measures and References Finding Comments
MHPG
Postmortem
Bierer et al. 199558 F MHPG 47 patients with AD versus 10 control subjects, clinical assessment by
CERAD, Khachaturian criteria, control group included patients with
MID (n⳱5); concurrent antibiotic treatment, diuretic, antiarrhythmics
or antianginal treatment, and neuroleptic use
Hoogendijk et al. 199962 } MHPG (frontal medial gyrus, superior temporal gyrus, 16 patients with AD (depressed n⳱6, transiently depressed n⳱5,
LC, hippocampus, amygdala, thalamus) nondepressed n⳱5) versus control subjects; comorbid hallucinations
(n⳱3), anxiety (n⳱2), agitation (n⳱4), compulsive eating (n⳱1),
delirium (n⳱2), apathy (n⳱1), chronic psychosis (n⳱1); clinical
assessment by NINCDS-ADRDA, DSM-III-R, Cornell Scale
Francis et al. 198565 F MHPG (early onset), F MHPG (late onset) 48 patients with AD (early onset n⳱19, older onset n⳱29) versus 34
control subjects
67
Storga et al. 1996 F MHPG (globus pallidus, raphe putamen, amygdala, Eight patients with AD versus six control subjects; concurrent neuroleptic
caudate, substantia nigra, cingulate gyrus) use (n⳱6)
Gottfries et al. 198371 F MHPG (caudate, cortex of the cingulate gyrus, 14 patients with AD versus 16 control subjects; comorbidity in patients
hippocampus) with AD with mild Parkinson’s disease and treated with l-dopa (n⳱1)
Herregodts et al. 198972 F MHPG (cortical, subcortical) Eight patients with AD (early-onset n⳱4, late-onset n⳱4) versus six

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


control subjects
Antemortem
Palmer et al. 198760 } MHPG (temporal lobe) Patients with AD versus control subjects; clinical assessment by WAIS,
the Token Test, a continuous Visual Reaction Time task; duration of
onset not reported.
CSF
Raskind et al. 198463 F MHPG (in advanced AD) 16 patients with AD (advanced n⳱9, moderate n⳱7) versus control
subjects; concurrent sympathetic nervous system medication (n⳱4),
low dose antipsychotic use (n⳱2), low dose tricyclic antidepressant use
(n⳱2); tricyclic antidepressant effect on MHPG level
Tohgi et al. 1992(81 F MHPG 11 patients with AD/SDAT, 17 patients with VDBT, and 15 control
subjects
Plasma
Raskind et al. 198463 F MHPG (in advanced AD) 16 patients with AD (advanced n⳱9, moderate n⳱7) versus control
subjects. See above for further information
Lawlor et al. 199582 F MHPG (in AD with respect to decline of cognition) 23 patients with AD; results controlled for age, age at onset, sex, and
interval between plasma MHPG determination and cognitive testing
MHPG/NE ratio
Postmortem
Hoogendijk et al. 199962 F MHPG/NE ratio (LC, frontal medial gyrus, superior 16 patients with AD (depressed n⳱6, transiently depressed n⳱5,
temporal gyrus, hippocampus, thalamus) nondepressed n⳱5) versus control subjects. See above for further
information.
Antemortem
Palmer et al. 198760 F MHPG/NE ratio Patients with AD versus control subjects. See above for further
information.
DHPG
CSF (in response to yohimbine challenge)
Raskind et al. 199987 } DBH 10 patients with AD versus 21 control subjects (young n⳱11, old n⳱10);
behavior rating nurses were not blinded; behavior assessment with
BPRS
Plasma (in response to yohimbine
challenge)
Raskind et al. 199987 } DBH 10 patients with AD versus 21 control subjects (young n⳱11, old n⳱10).
See preceding page for further information.
HERRMANN et al.

267
continued
NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

12 patients with AD, five patients with MID, nine with depression versus
mentia (Table 3). The role of norepinephrine in the fa-

control subjects; clinical assessment with Hachinski’s ischemic score,


31 patients with AD and 31 patients with MID versus 46 age-matched

31 patients with AD and 31 patients with MID versus 46 age-matched

MHPG⳱3-methoxy-4-hydroxyphenylglycol; CERAD⳱Consortium to Establish a Registry for Alzheimer’s Disease; AD⳱Alzheimer’s disease; MID⳱multi-infarct dementia;
computerized tomography, Hasegawa’s Dementia Rating Scale
cilitation of BPSD in AD is reviewed below.

LC⳱locus caeruleus; NINCDS-ADRDA⳱National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders
16 control subjects; subject background not reported

control subjects. See above for further information.

Association criteria; SDAT⳱senile dementia of the Alzheimer type; PD⳱Parkinson’s disease; VDBT⳱vascular dementia of the Binswanger type; NE⳱norepinephrine;
Depression
Six patients with AD versus six control subjects

Depression may complicate AD and has been found to


be most common among people with mild severity of
dementia.120 Both AD and depression share similar
signs and symptoms, including loss of interest and ap-
petite, apathy, and sleep disturbances.4 Studies have
found that AD patients suffering from comorbid
depression have significantly more LC neuron degen-
eration when compared to nondepressed AD pa-
tients.79,121,122 However, contradictory evidence exists.
Hoogendijk et al.123 also compared LC neuronal cell
counts between depressed and nondepressed AD pa-
tients but found no significant differences in LC degen-
DHPG⳱3,4-dihydroxyphenylglycol; DBH⳱dopamine-beta-hydroxylase enzyme; BPRS⳱Brief Psychiatric Rating Scale.

eration between depressed and nondepressed AD pa-


tients. This may be due to methodological differences
Summary of Literature Concerning Changes in NE Metabolites in Patients with ADa (continued)

between the studies such as confounding by level of se-


verity of both dementia121 and depression,121,122 LC sam-
ple section size and location of analysis.79,122 A reduction
in NE has also been observed in cortex of depressed AD
patients124 further supporting the role altered noradren-
ergic function may play in influencing depressive symp-
f DBH activity (frontal, temporal cortex, hippocampus)

f DBH (especially with severe dementia and/or severe

toms. Thus, the literature currently supports an associ-


ation between NE and depression in AD.

Aggression and Agitation


Russo-Neustadt and Cotman125 observed a small but
brain atrophy)

significant increase in total b-adrenoreceptor concentra-


f DBH (LC)

tion in the cerebella of aggressive AD patients when


f DBH

compared to nonaggressive AD patients and healthy


control subjects. This finding suggests a possible link
between aggression/agitation and b-adrenergic receptor
binding.
Since presynaptic ␣2 receptors regulate the negative
feedback of NE and postsynaptic receptors are thought
to regulate postsynaptic neuronal function, the differ-
ential location of the ␣2 adrenoreceptor may play a role
in modulating aggression.106 Activation of the ␣2 adre-
noreceptor by an agonist may lead to differing results
depending on whether the post- or presynaptic recep-
Iversen et al. 198374

tors were stimulated or, possibly, depending on the ratio


Miyata et al. 198483

Miyata et al. 198483


Cross et al. 198184

of the activated receptors. A postmortem study125 of the


frontal cortex, hypothalamus and cerebellum (areas in-
Postmortem

nervated by LC neurons) of agitated AD patients, non-


TABLE 2.

agitated AD subjects and healthy elderly control sub-


Serum

jects revealed a significantly elevated level of ␣2


CSF
DBH

receptors in the cerebellum of aggressive subjects, 70%

268 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


HERRMANN et al.

higher than the nonagitated AD subjects. The levels of Psychosis


␣2 receptors found in the nonagitated AD patients were Psychosis is a cluster of clinically disruptive behaviors
slightly but not significantly higher than the levels often observed in AD patients. Psychotic behaviors in-
found in the healthy elderly control subjects. Measure- volve delusions and hallucinations and can aggravate
ments of the ␣2 receptors in the frontal cortex and hy- aggression.129 Both evidence supporting the association
pothalamus were not found to vary significantly. between psychotic behaviors and noradrenergic pres-
There is some evidence that supports an association ervation,130 and evidence showing no association be-
between motor restlessness and increased MHPG levels. tween psychosis and NE121,131 have been published. Zu-
MHPG levels have been found to be correlated with benko and associates130 found higher NE concentrations
restlessness in AD patients.126 in the substantia nigra of psychotic AD patients when
In vivo studies have also been performed to address compared to nonpsychotic AD patients. Förstl et al.131
the link between BPSD and NE. Yohimbine has been found that AD patients with auditory hallucinations or
reported to lead to increased release of NE and succes- delusions had significantly higher neuron counts in the
sive activation of the postsynaptic ␣1 adrenoreceptors.127 parahippocampal gyrus and a trend towards lower dor-
It has been shown that AD patients are more sensitive sal raphe nucleus neuron counts compared to AD pa-
to administration of yohimbine, compared to other el- tients without psychosis; however, this was not ob-
derly patients and younger subjects,87,128 and exhibit served in the LC. The link between the noradrenergic
more agitated behaviors. This may be due to increased system and psychotic behaviors is still unclear and fur-
postsynaptic noradrenergic sensitivity in AD.87,89 This is ther investigation is necessary.
plausible, as postsynaptic b-receptors have been re- In summary, evidence from both animal and human
ported to be up-regulated in the prefrontal cortex and studies supports a link between noradrenergic dysfunc-
hippocampus of AD patients,89 consistent with the pos- tion and behavior, particularly aggressive behavior. De-
tulated increase in sensitivity to noradrenergic stimu- pressed patients, patients suffering from anxiety disor-
lation.87 As a result of noradrenergic overactivation, AD ders and AD patients all show blunted growth hormone
patients would no longer be able to focus attention and (GH) responses to clonidine, strongly suggesting a dis-
their mechanisms of coping with stressful stimuli would ruption in noradrenergic control. Intensified noradren-
be compromised. Even in the absence of stressful stim- ergic activity and/or hypersensitive adrenoreceptors are
uli, the noradrenergic system would be active. This may thought to mediate this NE dysfunction in aggression
account for the aggression displayed in many AD pa- associated both with and without AD as well as anxiety
tients.38,62 These findings further consolidate the prob- and depression. It is believed that compensation for the
ability of an adrenergic dysfunction contributing to the loss of the noradrenergic neurons that occurs during the
pathophysiology of agitated behaviors in AD. progression of AD leads to an increase in the number of

TABLE 3. Summary of Studies Linking Noradrenergic System Components With BPSDa


Noradrenergic System Finding Behavior Reference
Component
␣1—post — — —
␣2 (not specified if pre or post) F cerebellum, } frontal cortex, Aggression Russo-Neustadt et al. 1997125
hypothalamus
b1—post F cerebellum Aggression Russo-Neustadt et al. 1997125
b2—post F cerebellum Aggression Russo-Neustadt et al. 1997125
3-Methoxy-4- F CSF MHPG Restlessness Brane et al. 1989126
hydroxyphenylglycol (MHPG)
levels
LC cell counts F degeneration Aggression Matthews et al. 200278
F degeneration Depression Förstl et al. 199279, Zubenko et al.
1988122, Zweig et al. 1988121
} degeneration Depression Hoogendijk et al. 1999
} degeneration, f degeneration Psychosis Zweig et al. 1988121, Förstl et al. 1994131,
Zubenko et al. 1991130
a
F⳱increased; }⳱no change.

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 269


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

noradrenergic receptors and/or increased activity of re- Pindolol wields a partial agonist effect thereby exerting
maining noradrenergic neurons, the potential source of similar therapeutic benefits as propanolol but with less
aggressive behavior. This is credible as b1, b2 and ␣2 re- cardiovascular side effects such as hypotension, brady-
ceptor upregulation has been shown in aggressive AD cardia and decreased cardiac output. Patients were
patients when compared to nonaggressive AD patients. maintained on an individualized optimum dosage be-
The clinical expression of aggression likely is also de- tween 40–60 mg/day for a minimum of 10 days. Signifi-
pendent on other neurotransmitters. In summary, it has cant reductions in assaultiveness, hostility, lack of com-
been shown that both ␣ and b-adrenoreceptors are in- munication, uncooperativeness and repetitive behaviors
volved in the modulation of aggression. However, the were noted. Shankle et al.135 observed a 71% response
exact role each receptor plays is still uncertain and fur- rate in patients with either AD or vascular dementia
ther research is needed to clearly assess the link with NE (VaD) with both agitated and aggressive behaviors who
and behavior, particularly BPSD. were treated with propanolol. The 12 patients received
20 mg propanolol per day which was increased to an
efficacious maximum that ranged between 30 and 80 mg
NORADRENERGIC PHARMACOTHERAPY
per day. One patient suffered an adverse event (brady-
The probability that the noradrenergic system contrib- cardia), which was resolved by lowering the dose. A
utes to the pathophysiology of BPSD associated with case series by Weiler et al.137 also demonstrated success
AD has led to potential therapeutic interventions. Phar- in relieving treatment refractory disruptive behavior in
macological manipulation of the CNS noradrenergic 4 patients with AD and 2 patients with VaD. The pro-
system through the blockade of the beta adrenergic re- panolol doses ranged from 80 mg/day to 520 mg/day
ceptors has been studied as a treatment for a variety of and no adverse events were noted. These studies indi-
neuropsychiatric disorders.132 As it is believed norepi- cate that the modulation of the b-adrenoreceptors
nephrine is involved in the modulation of behavior, nor- through the use of b-blockers has been useful in the re-
adrenergic agents have much pharmacological potential duction of aggressive behaviors. However, many of
for the amelioration of behavioral disturbances. Study- these studies involved patients with varying dementia
ing the effects of noradrenergic agents on the various etiologies. A randomized controlled trial with AD pa-
behavioral disturbances associated with dementia can tients treated for BPSD using b-blockers is necessary be-
also lead to a better understanding of their neurochem- fore one can accurately judge effectiveness. Important
ical etiology in the way neuroleptic response led to an clinical considerations must be recognized when using
understanding of the role of DA in schizophrenia. b-blockers in elderly patients. b- Blockers may exacer-
bate various concomitant illnesses such as chronic ob-
Beta Blockers structive pulmonary disease, diabetes mellitus and pe-
Previous studies have indicated propanolol, a long-act- ripheral vascular disease. Additionally, beta-blockers
ing b-blocker, to be an effective treatment for aggression have been shown to increase plasma concentrations of
and agitation in patients with dementia who have been some antipsychotics, including chlorpromazine and
unsuccessfully treated with conventional therapies.133–137 thioridazine.139
In a randomized double-blind crossover placebo-con-
trolled study, Greendyke et al.133 evaluated the efficacy Antidepressants
of propanolol for an 11-week period at a maintenance Imipramine, a tricyclic antidepressant, inhibits the reup-
dosage of 520 mg/day, in assaultive patients with or- take of noradrenaline and has been shown to prevent
ganic brain impairment. The authors reported signifi- upregulation of b-adrenoreceptors induced by stress140
cantly fewer assaults and attempted assaults by the nine and may hinder the induction of tyrosine hydroxylase
patients who completed the study, during the active in the LC following cold stress,51 suggesting a lessening
treatment phase when compared to the placebo phase. of stress-induced norepinephrine release. Reifler et al.141
Five patients showed marked improvement, two compared the use of imipramine with placebo in 61 AD
showed moderate improvement and two showed little patients, of those, 28 had a diagnosis of depression and
or no improvement of violent behavior. A similar study 33 did not. The authors found that the depressed pa-
was repeated by Greendyke and Kanter138 using pin- tients scored significantly higher than the nondepressed
dolol, another b1 blocking agent, rather than propanolol. patients on the Hamilton Depression Rating Scale and

270 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


HERRMANN et al.

though all patients improved significantly during the Doses were increased up to the maintenance dose of 10
study, the depressed patients improved significantly mg bid. Appetite was found to increase within 3–7 days
more than the patients without depression. Although of initiating therapy, and complete resolution of an-
clinical recommendations have been made about avoid- orexia, leading to weight gain, was observed within a
ing the use of tertiary amines in the elderly demented few weeks. Improvement in social interaction and hence
population,142 imipramine was found to be extremely a decrease in apathy was also noted by the nurses. The
well tolerated in Reifler’s study, as no significant differ- patients were maintained on methylphenidate for 9, 16
ences in adverse events were noted in the imipramine- and 24 months, respectively, and were eventually ta-
treated group compared to the placebo-treated group. pered off successfully. No adverse effects were noted
Mirtazapine is a noradrenergic and specific seroto- and no other psychotropic was used during the period
nergic antidepressant that directly blocks the pre- and of treatment. Kaufmann et al.146 also found favorable
postsynaptic ␣2 receptors and antagonizes the 5-hydrox- results with one dementia patient who was treated suc-
ytryptamine (5-HT)2 and 5-HT3 receptors with high af- cessfully for depressive symptoms including appetite
finity and 5-HT1 receptors with low affinity.143 Mirta- loss and apathy, using methylphenidate.
zapine has been used with favorable results in a case Branconnier and Cole147 also conducted a placebo-
series by Raji and Brady144 in AD patients suffering from controlled study in demented, apathetic patients using
depression as well as anxiety, weight loss and insomnia. 20–30 mg MPD and found an improvement in apathy
Three patients were given a trial of mirtazapine. Two that correlated with a lessening of depressive symp-
patients were started at 7.5 mg and titrated up over 2 toms. Galynker et al.148 treated 12 AD patients and 15
weeks to 15 mg. One of those patients was further ti- VaD patients using methylphenidate and found a sig-
trated up over 2 more weeks to 30 mg. The third patient nificant improvement in negative symptoms.
was started at 15 mg and was increased to 30 mg after Although the benefits of methylphenidate therapy in
2 weeks. No changes were noted in cognition; however, the elderly have been reported for the last 50 years, the
improvement in appetite and sleep was documented results are limited by poor designs and lack of stan-
within 2–4 weeks and cessation of anxiety and depres- dardized assessments for behavior and even diagnoses.
sion was noted at 2 months. None of the patients suf- Methylphenidate appears to be useful in alleviating de-
fered any adverse effects. This case series suggests that pressive symptoms including appetite loss and apathy;
mirtazapine may be an effective therapy for depressed however, more research is needed to explore the value
AD patients with comorbid insomnia, anxiety and ap- of; MPD in the pharmacotherapy of other behavioral
petite loss. However, there is a lack of randomized con- disturbances associated with AD. Furthermore, as noted
trolled trials using mirtazapine, and therefore the extent previously, psychostimulants have effects on other neu-
of the usefulness of mirtazapine pharmacotherapy is rotransmitters, and it is not clear whether their beneficial
still unknown and remains to be explored. effects for BPSD are due to their actions on NE or other
Other antidepressants with putative effects on the NE systems.
system include venlafaxine, bupropion, and reboxetine, It is difficult to treat BPSD pharmacologically as some
though none of these agents have been tested for BPSD. patients may be very sensitive to first-line therapy with
antipsychotic medications. Treatment emergent side ef-
Psychostimulants fects may be intolerable resulting in the discontinuation
Psychostimulants, such as methylphenidate (MPD), of the drug. Hence, prescribing medication for BPSD of-
have been used in the past to treat affective disorders, ten involves trying a series of medications on each pa-
particularly depression. MPD is believed to block both tient. The use of b adrenergic antagonists for BPSD such
norepinephrine and DA uptake and promote catechol- as agitation and aggression may be clinically advanta-
amine release.145 Few controlled studies have evaluated geous due to the absence of the extrapyramidal symp-
the efficacy of psychostimulants in the treatment BPSD. toms and sedation commonly seen with antipsychotics.
A case series by Maletta and Winegarden145 examined Other adrenergic agents have been studied for the alle-
three severely demented patients, suffering from apathy viation of other behavioral symptoms frequently asso-
and anorexia resulting in weight loss, using methyl- ciated with dementia, however, the vast majority of nor-
phenidate therapy. Two patients were started on 5 mg adrenergic agents prescribed are for depression.
bid and one patient was started on 5 mg once daily. Although the pharmacological management of depres-

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 271


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

sion with noradrenergic drugs has been shown to be ceptors and their subtypes in controlling behavior in AD
successful, there is a great need for further research to is necessary to elucidate the extent behavior dysregu-
elucidate the role of noradrenergic pharmacotherapy in lation may be accounted for by noradrenergic dysfunc-
the treatment of depression associated with AD. Given tion. A limitation of the current knowledge of the role
the modest efficacy of antipsychotics and the variable of NE in BPSD is lack of information on other behaviors
responses to antidepressants, randomized controlled tri- such as apathy, wandering, and sexually disinhibited
als of noradrenergic agents for BPSD are urgently behaviors. The current literature search revealed studies
needed. that addressed depression, agitation, aggression and
psychosis only.
Drugs that target NE have been successfully used in
CONCLUSIONS
treatment of depression, which is commonly found in AD
The bulk of neuropathological studies in AD show nor- as well. However, there is little research, especially ran-
adrenergic neuron degeneration, decreased NE levels domized controlled trials, into the use of noradrenergic
and increased NE metabolites. This evidence strongly treatments for BPSD. Studies on beta blocker treatment
and consistently suggests enhanced NE turnover. The for aggression and psychostimulant use for affective dis-
vast majority of studies; however, have been done post- orders appear to be the better studied pharmacologic in-
mortem, and consequently caution is necessary in their terventions in AD; however, they too suffer from lack of
interpretation. As altered behavior may be a result of standardization in behavioral assessments, heterogeneity
“state” rather than “trait” phenomenon, it is important in dementia diagnoses and small sample sizes. Further
that the behavior being examined is manifested at the comprehensive research is necessary to explore the po-
time of death, so as to accurately measure the neurolog- tential clinical utility of noradrenergic agents for the
ical changes believed to be responsible. Postmortem de- pharmacological management of disruptive behaviors
lay is a confounding variable in neurophysiological associated with dementia.
analysis as postmortem results may not correctly rep- The study of the neurobiology of BPSD in AD is
resent the antemortem state. The consequences of delay fraught with methodological difficulties. While much
in freezing brain tissue after death and prolonged pres- has been learned over the past two decades, the goal of
ervation of brain tissue prior to catecholamine analysis a unitary etiological hypothesis may never be reached
have not yet been fully accounted for and may limit the given the heterogeneity of AD and its variable effects on
usefulness of postmortem studies. behavior. Providing putative links between discrete
Norepinephrine is involved in the sympathetic neurotransmitter dysfunction and behavior; however,
“flight-or-fight” response and thus is sensitive to envi- will allow for more targeted pharmacological interven-
ronmental challenges and can modulate behavior ac- tions. Only then will treatment of BPSD be truly “ra-
cordingly. The noradrenergic system has been shown to tional.”
mediate behavior, particularly aggression, in animals as
well as in psychiatric illnesses. Although there is sub- This study was funded in part by a grant from the Physi-
stantial evidence correlating NE with aggression, a di- cians’ Services Incorporated Foundation, grant number 98-
rect link between NE function and aggression associated 39.
with dementia needs to be examined further. More re- The authors thank Luis Lewis, Jr. and Florance Chan for
search into identifying the role of the noradrenergic re- their assistance.

References

1. Fratiglioni L, De Ronchi D, Aguero-Torres H: Worldwide prev- 4. Raskind MA: Geriatric psychopharmacology: management of
alence and incidence of dementia. Drugs Aging 1999; 15:365–375 late-life depression and the noncognitive behavioral distur-
2. Fillit HM: The pharmacoeconomics of Alzheimer’s disease. Am bances of Alzheimer’s disease. Psychiatr Clin North Am 1993;
J Manag Care 2000; 6(22 suppl):S1139–S1144; discussion S1145– 16:815–827
S1138 5. Teri L, Rabins P, Whitehouse P, et al: Management of behavior
3. Evans DA, Funkenstein HH, Albert MS, et al: Prevalence of Alz- disturbance in Alzheimer disease: current knowledge and future
heimer’s disease in a community population of older persons: directions. Alzheimer Dis Assoc Disord 1992; 6:77–88
higher than previously reported. JAMA 1989; 262:2551–2556 6. Reisberg B, Borenstein J, Salob SP, et al: Behavioral symptoms in

272 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


HERRMANN et al.

Alzheimer’s disease: phenomenology and treatment. J Clin Psy- onciliation Act 1987 on antipsychotic prescribing in nursing
chiatry 1987; 48(May suppl):9–15 home residents. J Am Geriatr Soc 1994; 42:648–652
7. Gilley DW, Whalen ME, Wilson RS, et al: Hallucinations and 28. Dodds MJ: Pre- and post-OBRA ’87 psychoactive drug use in
associated factors in Alzheimer’s disease. J Neuropsychiatry long-term care facilities. J Long Term Care Adm 1996; 24:17–22
Clin Neurosci 1991; 3:371–376 29. Zayas EM, Grossberg GT: Treating the agitated Alzheimer pa-
8. Rabins PV, Mace NL, Lucas MJ: The impact of dementia on the tient. J Clin Psychiatry 1996; 57(suppl 7):46–51; discussion 52–44
family. JAMA 1982; 248:333–335 30. Lanctôt KL, Bowles SK, Herrmann N, et al: Drugs mimicking
9. Nagaratnam N, Lewis-Jones M, Scott D, et al: Behavioral and dementia: dementia symptoms associated with psychotropic
psychiatric manifestations in dementia patients in a community: drugs in institutionalised cognitively impaired patients. CNS
caregiver burden and outcome. Alzheimer Dis Assoc Disord Drugs 2000; 14:381–390
1998; 12:330–334 31. Lanctôt KL, Best TS, Mittmann N, et al: Efficacy and safety of
10. Stoppe G, Brandt CA, Staedt JH: Behavioural problems associ- neuroleptics in behavioral disorders associated with dementia.
ated with dementia: the role of newer antipsychotics. Drugs Ag- J Clin Psychiatry 1998; 59:550–561; quiz 562–553
ing 1999; 14:41–54 32. Schneider LS, Pollock VE, Lyness SA: A metaanalysis of con-
11. Mega MS, Cummings JL, Fiorello T, et al: The spectrum of be- trolled trials of neuroleptic treatment in dementia. J Am Geriatr
havioral changes in Alzheimer’s disease. Neurology 1996; Soc 1990; 38:553–563
46:130–135 33. Herrmann N, Lanctôt KL: The management of behavioral dis-
12. Petrie W, Lawson E, Hollender M: Violence in geriatric patients. turbances in dementia: the role of serotonergic therapies. IDrugs
JAMA 1982; 248:443–444 1998; 1:214–220
13. Gaspar D: Hollymoor Hospital Dementia Service: analysis of 34. Lanctôt KL, Herrmann N, Mazzotta P: Role of serotonin in the
outcome of 230 consecutive referrals to a psychiatric-hospital behavioral and psychological symptoms of dementia. J Neuro-
dementia service. Lancet 1980; 1:1401–1405 psychiatry Clin Neurosci 2001; 13:5–21
14. Hebert R, Dubois MF, Wolfson C, et al: Factors associated with 35. Coull JT: Pharmacological manipulations of the alpha 2-norad-
long-term institutionalization of older people with dementia: renergic system: effects on cognition. Drugs Aging 1994; 5:116–
data from the Canadian Study of Health and Aging. J Gerontol 126
A Biol Sci Med Sci 2001; 56:M693–M699 36. Yudofsky SC, Silver JM, Hales RE: Treatment of agitation and
15. Kunik ME, Yudofsky SC, Silver JM, et al: Pharmacologic ap- aggression, in Textbook of Psychopharmacology. Edited by
proach to management of agitation associated with dementia. J Schatzberg AF, Nemeroff CB. Washington, DC, American Psy-
Clin Psychiatry 1994; 55(Feb suppl):13–17 chiatric Press, 1998, pp 881–900
16. Cohen-Mansfield J, Billig N: Agitated behaviors in the elderly, I: 37. Redmond D: Studies of the nucleus locus ceruleus in monkeys
a conceptual review. J Am Geriatr Soc 1986; 34:711–721 and hypothesis for neuropsychopharmacology, in Psychophar-
17. Gleason RP, Schneider LS: Carbamazepine treatment of agitation macology: The Third Generation of Progress. Edited by Meltzer
in Alzheimer’s outpatients refractory to neuroleptics. J Clin Psy- H. New York, Raven Press, 1987, pp 967–975
chiatry 1990; 51:115–118 38. Friedman JI, Adler DN, Davis KL: The role of norepinephrine in
18. Ryden MB: Aggressive behavior in persons with dementia who the pathophysiology of cognitive disorders: potential applica-
live in the community. Alzheimer Dis Assoc Disord 1988; 2:342– tions to the treatment of cognitive dysfunction in schizophrenia
355 and Alzheimer’s disease. Biol Psychiatry 1999; 46:1243–1252
19. Herrmann N: Recommendations for the management of behav- 39. Nestler EJ, Alreja M, Aghajanian GK: Molecular control of locus
ioral and psychological symptoms of dementia. Can J Neurol Sci coeruleus neurotransmission. Biol Psychiatry 1999; 46:1131–1139
2001; 28(suppl 1):S96–S107 40. Beani L, Bianchi C, Giacomelli A, et al: Noradrenaline inhibition
20. Yeager BF, Farnett LE, Ruzicka SA: Management of the behavioral of acetylcholine release from guinea-pig brain. Eur J Pharmacol
manifestations of dementia. Arch Intern Med 1995; 155:250–260 1978; 48:179–193
21. Treatment of agitation in older persons with dementia: the ex- 41. Vizi ES: Presynaptic modulation of neurochemical transmission.
pert consensus panel for agitation in dementia. Postgrad Med Prog Neurobiol 1979; 12:181–290
Special Issue 1998, pp 1–88 42. Vizi ES, Pasztor E: Release of acetylcholine from isolated human
22. Lantz MS, Louis A, Lowenstein G, et al: A longitudinal study of cortical slices: inhibitory effect of norepinephrine and phenytoin.
psychotropic prescriptions in a teaching nursing home. Am J Exp Neurol 1981; 73:144–153
Psychiatry 1990; 147:1637–1639 43. Bianchi C, Spidalieri G, Guandalini P, et al: Inhibition of acetyl-
23. Avorn J, Dreyer P, Connelly K, et al: Use of psychoactive medi- choline outflow from guinea-pig cerebral cortex following locus
cation and the quality of care in rest homes: findings and policy coeruleus stimulation. Neurosci Lett 1979; 14:97–100
implications of a statewide study. N Engl J Med 1989; 320:227– 44. Jaim-Etcheverry G, Zieher L: Coexistence of monoamines in pe-
232 ripheral adrenergic neurones, in Co-Transmission. Edited by
24. Gurwitz JH, Soumerai SB, Avorn J: Improving medication pre- Cuello A. London, Macmillan, 1982, pp 189–207
scribing and utilization in the nursing home. J Am Geriatr Soc 45. Murphy DL, Andrews AM, Wichems CH, et al: Brain serotonin
1990; 38:542–552 neurotransmission: an overview and update with an emphasis
25. Hesse KA, Driscoll A, Jacobson S: Neuroleptic prescriptions for on serotonin subsystem heterogeneity, multiple receptors, inter-
acutely ill geriatric patients. Arch Intern Med 1993; 153:2581– actions with other neurotransmitter systems, and consequent
2587 implications for understanding the actions of serotonergic
26. Beers MH, Ouslander JG, Rollingher I, et al: Explicit criteria for drugs. J Clin Psychiatry 1998; 59(suppl 15):4–12
determining inappropriate medication use in nursing home res- 46. Gillespie DD, Manier DH, Sanders-Bush E, et al: The serotonin/
idents: UCLA Division of Geriatric Medicine. Arch Intern Med norepinephrine-link in brain, II: role of serotonin in the regula-
1991; 151:1825–1832 tion of beta adrenoceptors in the low agonist affinity confor-
27. Semla TP, Palla K, Poddig B, et al: Effect of the Omnibus Rec- mation. J Pharmacol Exp Ther 1988; 244:154–159

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 273


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

47. Soderpalm B, Andersson L, Carlsson M, et al: Serotonergic in- 67. Storga D, Vrecko K, Birkmayer JG, et al: Monoaminergic neu-
fluence on the growth hormone response to clonidine in rat. J rotransmitters, their precursors and metabolites in brains of Alz-
Neural Transm 1987; 69:105–114 heimer patients. Neurosci Lett 1996; 203:29–32
48. Yamamoto BK, Novotney S: Regulation of extracellular dopa- 68. Mann DM, Yates PO, Marcyniuk B: A comparison of changes in
mine by the norepinephrine transporter. J Neurochem 1998; the nucleus basalis and locus caeruleus in Alzheimer’s disease.
71:274–280 J Neurol Neurosurg Psychiatry 1984; 47:201–203
49. Gresch PJ, Sved AF, Zigmond MJ, et al: Local influence of en- 69. Bondareff W, Mountjoy CQ, Roth M: Loss of neurons of origin
dogenous norepinephrine on extracellular dopamine in rat me- of the adrenergic projection to cerebral cortex (nucleus locus cer-
dial prefrontal cortex. J Neurochem 1995; 65:111–116 uleus) in senile dementia. Neurology 1982; 32:164–168
50. Ferraro L, Tanganelli S, Calo G, et al: Noradrenergic modulation 70. Hoogendijk WJ, Pool CW, Troost D, et al: Image analyser-as-
of gamma-aminobutyric acid outflow from the human cerebral sisted morphometry of the locus coeruleus in Alzheimer’s dis-
cortex. Brain Res 1993; 629:103–108 ease, Parkinson’s disease and amyotrophic lateral sclerosis.
51. Melia KR, Duman RS: Involvement of corticotropin-releasing Brain 1995; 118(part 1):131–143
factor in chronic stress regulation of the brain noradrenergic sys- 71. Gottfries CG, Adolfsson R, Aquilonius SM, et al: Biochemical
tem. Proc Natl Acad Sci USA 1991; 88:8382–8386 changes in dementia disorders of Alzheimer type (AD/SDAT).
52. Veith RC, Lewis N, Langohr JI, et al: Effect of desipramine on Neurobiol Aging 1983; 4:261–271
cerebrospinal fluid concentrations of corticotropin-releasing fac- 72. Herregodts P, Bruyland M, De Keyser J, et al: Monoaminergic
tor in human subjects. Psychiatry Res 1993; 46:1–8 neurotransmitters in Alzheimer’s disease: an HPLC study com-
53. Gerner RH, Yamada T: Altered neuropeptide concentrations in paring presenile familial and sporadic senile cases. J Neurol Sci
cerebrospinal fluid of psychiatric patients. Brain Res 1982; 1989; 92:101–116
238:298–302 73. Miller MA, Kolb PE, Leverenz JB, et al: Preservation of norad-
54. Minthon L, Edvinsson L, Gustafson L: Correlation between clini- renergic neurons in the locus ceruleus that coexpress galanin
cal characteristics and cerebrospinal fluid neuropeptide Y levels mRNA in Alzheimer’s disease. J Neurochem 1999; 73:2028–2036
in dementia of the Alzheimer type and frontotemporal demen- 74. Iversen LL, Rossor MN, Reynolds GP, et al: Loss of pigmented
tia. Alzheimer Dis Assoc Disord 1996; 10:197–203 dopamine-beta-hydroxylase positive cells from locus coeruleus
55. de Wied D, van Ree JM: Neuropeptides: animal behaviour and in senile dementia of Alzheimer’s type. Neurosci Lett 1983;
human psychopathology. Eur Arch Psychiatry Neurol Sci 1989; 39:95–100
238:323–331 75. Moll G, Gsell W, Wichart I, et al: Cholinergic and monoaminergic
56. Herrmann N, Lanctôt KL: From transmitters to treatment: the neuromediator systems in DAT: neuropathological and neuro-
pharmacotherapy of behavioural disturbances in dementia. Can chemical findings, in Alzheimer’s Disease: Epidemiology, Neu-
J Psychiatry 1997; 42(suppl 1):51S–64S ropathy, Neurochemistry, and Clinics. Edited by Maurer K, Ried-
57. Lawlor BA, Sunderland T, Mellow AM, et al: Hyperresponsivity erer P, Beckmann H. New York, Springer Verlag, 1990, pp 235–
to the serotonin agonist m-chlorophenylpiperazine in Alzhei- 243
mer’s disease: a controlled study. Arch Gen Psychiatry 1989; 76. Nyberg P, Adolfsson R, Hardy JA, et al: Catecholamine topo-
46:542–549 chemistry in human basal ganglia: comparison between normal
58. Bierer LM, Haroutunian V, Gabriel S, et al: Neurochemical cor- and Alzheimer brains. Brain Res 1985; 333:139–142
relates of dementia severity in Alzheimer’s disease: relative im- 77. Yates CM, Simpson J, Gordon A, et al: Catecholamines and cho-
portance of the cholinergic deficits. J Neurochem 1995; 64:749– linergic enzymes in pre-senile and senile Alzheimer-type de-
760 mentia and Down’s syndrome. Brain Res 1983; 280:119–126
59. Mann DM, Yates PO, Hawkes J: The noradrenergic system in 78. Matthews KL, Chen CP, Esiri MM, et al: Noradrenergic changes,
Alzheimer and multi-infarct dementias. J Neurol Neurosurg aggressive behavior, and cognition in patients with dementia.
Psychiatry 1982; 45:113–119 Biol Psychiatry 2002; 51:407–416
60. Palmer AM, Francis PT, Bowen DM, et al: Catecholaminergic 79. Förstl H, Burns A, Luthert P, et al: Clinical and neuropatholog-
neurones assessed ante-mortem in Alzheimer’s disease. Brain ical correlates of depression in Alzheimer’s disease. Psychol
Res 1987; 414:365–375 Med 1992; 22:877–884
61. Adolfsson R, Gottfries CG, Roos BE, et al: Changes in the brain 80. Perry EK, Tomlinson BE, Blessed G, et al: Neuropathological and
catecholamines in patients with dementia of Alzheimer type. Br biochemical observations on the noradrenergic system in Alz-
J Psychiatry 1979; 135:216–223 heimer’s disease. J Neurol Sci 1981; 51:279–287
62. Hoogendijk WJ, Feenstra MG, Botterblom MH, et al: Increased 81. Tohgi H, Ueno M, Abe T, et al: Concentrations of monoamines
activity of surviving locus ceruleus neurons in Alzheimer’s dis- and their metabolites in the cerebrospinal fluid from patients
ease. Ann Neurol 1999; 45:82–91 with senile dementia of the Alzheimer type and vascular de-
63. Raskind MA, Peskind ER, Halter JB, et al: Norepinephrine and mentia of the Binswanger type. J Neural Transm Park Dis De-
MHPG levels in CSF and plasma in Alzheimer’s disease. Arch ment Sect 1992; 4:69–77
Gen Psychiatry 1984; 41:343–346 82. Lawlor BA, Bierer LM, Ryan TM, et al: Plasma 3-methoxy-4-
64. Arai H, Kosaka K, Iizuka R: Changes of biogenic amines and hydroxyphenylglycol (MHPG) and clinical symptoms in Alz-
their metabolites in postmortem brains from patients with Alz- heimer’s disease. Biol Psychiatry 1995; 38:185–188
heimer-type dementia. J Neurochem 1984; 43:388–393 83. Miyata S, Nagata H, Yamao S, et al: Dopamine-beta-hydroxylase
65. Francis PT, Palmer AM, Sims NR, et al: Neurochemical studies activities in serum and cerebrospinal fluid of aged and de-
of early-onset Alzheimer’s disease: possible influence on treat- mented patients. J Neurol Sci 1984; 63:403–409
ment. N Engl J Med 1985; 313:7–11 84. Cross AJ, Crow TJ, Perry EK, et al: Reduced dopamine-beta-
66. Nazarali AJ, Reynolds GP: Monoamine neurotransmitters and hydroxylase activity in Alzheimer’s disease. Br Med J (Clin Res
their metabolites in brain regions in Alzheimer’s disease: a post- Ed) 1981; 282:93–94
mortem study. Cell Mol Neurobiol 1992; 12:581–587 85. Elrod R, Peskind ER, DiGiacomo L, et al: Effects of Alzheimer’s

274 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004


HERRMANN et al.

disease severity on cerebrospinal fluid norepinephrine concen- 105. Gerra G, Zaimovic A, Avanzini P, et al: Neurotransmitter-neu-
tration. Am J Psychiatry 1997; 154:25–30 roendocrine responses to experimentally induced aggression in
86. Li PP, Warsh JJ, Godse DD: Rat brain norepinephrine metabo- humans: influence of personality variable. Psychiatry Res 1997;
lism: substantial clearance through 3,4-dihydroxyphenylethyle- 66:33–43
neglycol formation. J Neurochem 1983; 41:1065–1071 106. Haller J, Makara GB, Kruk MR: Catecholaminergic involvement
87. Raskind MA, Peskind ER, Holmes C, et al: Patterns of cerebro- in the control of aggression: hormones, the peripheral sympa-
spinal fluid catechols support increased central noradrenergic thetic, and central noradrenergic systems. Neurosci Biobehav
responsiveness in aging and Alzheimer’s disease. Biol Psychia- Rev 1998; 22:85–97
try 1999; 46:756–765 107. Redmond DE Jr, Katz MM, Maas JW, et al: Cerebrospinal fluid
88. Holmes C, Eisenhofer G, Goldstein DS: Improved assay for amine metabolites: relationships with behavioral measurements
plasma dihydroxyphenylacetic acid and other catechols using in depressed, manic, and healthy control subjects. Arch Gen Psy-
high-performance liquid chromatography with electrochemical chiatry 1986; 43:938–947
detection. J Chromatogr B Biomed Appl 1994; 653:131–138 108. Swann AC, Secunda SK, Koslow SH, et al: Mania: sympathoad-
89. Peskind ER, Wingerson D, Murray S, et al: Effects of Alzheimer’s renal function and clinical state. Psychiatry Res 1991; 37:195–
disease and normal aging on cerebrospinal fluid norepinephrine 205
responses to yohimbine and clonidine. Arch Gen Psychiatry 109. Eichelman B: Neurochemical and psychopharmacologic aspects
1995; 52:774–782 of aggressive behavior, in Psychopharmacology: The Third Gen-
90. Anand A, Charney DS: Norepinephrine dysfunction in depres- eration of Progress. Edited by Meltzer H. New York, Raven
sion. J Clin Psychiatry 2000; 61(suppl 10):16–24 Press, 1987, pp 697–704
91. Arnsten AF, Steere JC, Hunt RD: The contribution of alpha 2- 110. Brown GL, Goodwin FK, Ballenger JC, et al: Aggression in hu-
noradrenergic mechanisms of prefrontal cortical cognitive func- mans correlates with cerebrospinal fluid amine metabolites. Psy-
tion: potential significance for attention-deficit hyperactivity dis- chiatry Res 1979; 1:131–139
order. Arch Gen Psychiatry 1996; 53:448–455 111. Gerra G, Avanzini P, Zaimovic A, et al: Neurotransmitter and
92. Chan-Palay V, Asan E: Alterations in catecholamine neurons of endocrine modulation of aggressive behavior and its compo-
the locus coeruleus in senile dementia of the Alzheimer type and nents in normal humans. Behav Brain Res 1996; 81:19–24
in Parkinson’s disease with and without dementia and depres- 112. Coccaro EF, Lawrence T, Trestman R, et al: Growth hormone
sion. J Comp Neurol 1989; 287:373–392 responses to intravenous clonidine challenge correlate with be-
93. Torda C: Effects of catecholamines on behavior. J Neurosci Res havioral irritability in psychiatric patients and healthy volun-
1976; 2:193–202 teers. Psychiatry Res 1991; 39:129–139
94. Thoa NB, Eichelman B, Richardson JS, et al: 6-Hydroxydopa de- 113. Tsai SJ, Hwang JP, Yang CH, et al: Physical aggression and as-
pletion of brain norepinephrine and the function of aggressive sociated factors in probable Alzheimer disease. Alzheimer Dis
behavior. Science 1972; 178:75–77 Assoc Disord 1996; 10:82–85
95. Geyer MA, Segal DS: Shock-induced aggression: opposite effects 114. Ware CJG, Fairburn CG, Hope RA: A community-based study
of intraventricularly infused dopamine and norepinephrine. Be- of aggressive behaviour in dementia. Int J Geriatr Psychiatry
hav Biol 1974; 10:99–104 1990; 5:337–342
96. Charney DS, Heninger GR, Sternberg DE, et al: Adrenergic re- 115. Lyketsos CG, Steinberg M, Tschanz JT, et al: Mental and behav-
ceptor sensitivity in depression: effects of clonidine in depressed ioral disturbances in dementia: findings from the Cache County
patients and healthy subjects. Arch Gen Psychiatry 1982; 39:290– Study on Memory in Aging. Am J Psychiatry 2000; 157:708–714
294 116. Lee MM, Strauss ME, Dawson DV: Changes in emotional and
97. Gilles C, Ryckaert P, De Mol J, et al: Clonidine-induced growth behavioral symptoms of Alzheimer’s disease. Am J Alzheimer
hormone secretion in elderly patients with senile dementia of Dis Other Demen 2000; 15:176–179
the Alzheimer type and major depressive disorder. Psychiatry 117. Keene J, Hope T, Fairburn CG, et al: Natural history of aggres-
Res 1989; 27:277–286 sive behaviour in dementia. Int J Geriatr Psychiatry 1999; 14:541–
98. Checkly S, Corn T, Glass I, et al: Responsiveness of central alpha- 548
2 adrenoceptors in depression, in The Biology of Depression. 118. Raskind MA, Peskind ER: Neurobiologic bases of noncognitive
Edited by Deakin J. London, Gaskell, 1986, pp 100–120 behavioral problems in Alzheimer disease. Alzheimer Dis Assoc
99. Leckman JF, Maas JW, Redmond DE Jr, et al: Effects of oral clo- Disord 1994; 8(suppl 3):54–60
nidine on plasma 3-methoxy-4-hydroxyphenethyleneglycol 119. Lindenmayer JP: The pathophysiology of agitation. J Clin Psy-
(MHPG) in man: preliminary report. Life Sci 1980; 26:2179–2185 chiatry 2000; 61(suppl 14):5–10
100. Ordway GA, Widdowson PS, Smith KS, et al: Agonist binding 120. Margallo-Lana M, Swann A, O’Brien J, et al: Prevalence and
to alpha 2-adrenoceptors is elevated in the locus coeruleus from pharmacological management of behavioural and psychological
victims of suicide. J Neurochem 1994; 63:617–624 symptoms amongst dementia sufferers living in care environ-
101. Ressler KJ, Nemeroff CB: Role of norepinephrine in the patho- ments. Int J Geriatr Psychiatry 2001; 16:39–44
physiology and treatment of mood disorders. Biol Psychiatry 121. Zweig RM, Ross CA, Hedreen JC, et al: The neuropathology of
1999; 46:1219–1233 aminergic nuclei in Alzheimer’s disease. Ann Neurol 1988; 24:
102. Abelson JL, Glitz D, Cameron OG, et al: Blunted growth hor- 233–242
mone response to clonidine in patients with generalized anxiety 122. Zubenko GS, Moossy J: Major depression in primary dementia:
disorder. Arch Gen Psychiatry 1991; 48:157–162 clinical and neuropathologic correlates. Arch Neurol 1988; 45:
103. Nutt DJ: Neurobiological mechanisms in generalized anxiety dis- 1182–1186
order. J Clin Psychiatry 2001; 62(suppl 11):22–27; discussion 28 123. Hoogendijk WJ, Sommer IE, Pool CW, et al: Lack of association
104. Geracioti TD Jr, Baker DG, Ekhator NN, et al: CSF norepineph- between depression and loss of neurons in the locus coeruleus
rine concentrations in posttraumatic stress disorder. Am J Psy- in Alzheimer disease. Arch Gen Psychiatry 1999; 56:45–51
chiatry 2001; 158:1227–1230 124. Zubenko GS, Moossy J, Kopp U: Neurochemical correlates of

J Neuropsychiatry Clin Neurosci 16:3, Summer 2004 275


NOREPINEPHRINE IN SYMPTOMS OF DEMENTIA

major depression in primary dementia. Arch Neurol 1990; 47: of rage and violent behavior in patients with chronic brain syn-
209–214 dromes. Am J Psychiatry 1981; 138:218–220
125. Russo-Neustadt A, Cotman CW: Adrenergic receptors in Alz- 137. Weiler PG, Mungas D, Bernick C: Propranolol for the control of
heimer’s disease brain: selective increases in the cerebella of ag- disruptive behavior in senile dementia. J Geriatr Psychiatry
gressive patients. J Neurosci 1997; 17:5573–5580 Neurol 1988; 1:226–230
126. Brane G, Gottfries CG, Blennow K, et al: Monoamine metabolites 138. Greendyke RM, Kanter DR: Therapeutic effects of pindolol on
in cerebrospinal fluid and behavioral ratings in patients with behavioral disturbances associated with organic brain disease: a
early and late onset of Alzheimer dementia. Alzheimer Dis As- double-blind study. J Clin Psychiatry 1986; 47:423–426
soc Disord 1989; 3:148–156 139. Silver JM, Yudofsky SC, Kogan M, et al: Elevation of thioridazine
127. Anden NE, Pauksens K, Svensson K: Selective blockade of brain plasma levels by propranolol. Am J Psychiatry 1986; 143:1290–
alpha 2-autoreceptors by yohimbine: effects on motor activity 1292
and on turnover of noradrenaline and dopamine. J Neural 140. Stanford SC: Central noradrenergic neurones and stress. Phar-
Transm 1982; 55:111–120 macol Ther 1995; 68:297–242
128. Peskind ER, Elrod R, Dobie DJ, et al: Cerebrospinal fluid epi- 141. Reifler BV, Teri L, Raskind M, et al: Double-blind trial of imip-
nephrine in Alzheimer’s disease and normal aging. Neuropsy- ramine in Alzheimer’s disease patients with and without de-
chopharmacology 1998; 19:465–471 pression. Am J Psychiatry 1989; 146:45–49
129. Aarsland D, Cummings JL, Yenner G, et al: Relationship of ag- 142. Peisah C, Brodaty H: Practical guidelines for the treatment of
gressive behavior to other neuropsychiatric symptoms in pa- behavioural complications of dementia. Med J Aust 1994;
tients with Alzheimer’s disease. Am J Psychiatry 1996; 153:243– 161:558–563
247 143. de Boer T: The pharmacologic profile of mirtazapine. J Clin Psy-
130. Zubenko GS, Moossy J, Martinez AJ, et al: Neuropathologic and chiatry 1996; 57(suppl 4):19–25
neurochemical correlates of psychosis in primary dementia. 144. Raji MA, Brady SR: Mirtazapine for treatment of depression and
Arch Neurol 1991; 48:619–624 comorbidities in Alzheimer disease. Ann Pharmacother 2001;
131. Förstl H, Burns A, Levy R, et al: Neuropathological correlates of 35:1024–1027
psychotic phenomena in confirmed Alzheimer’s disease. Br J 145. Maletta GJ, Winegarden T: Reversal of anorexia by methylphen-
Psychiatry 1994; 165:53–59 idate in apathetic, severely demented nursing home patients.
132. Yudofsky S, Silver J, Schneider S: Pharmacologic treatment of Am J Geriatr Psychiatry 1993; 1:234–243
aggression. Psychiatr Annals 1987; 27:397–407 146. Kaufmann MW, Cassem NH, Murray GB, et al: Use of psycho-
133. Greendyke RM, Kanter DR, Schuster DB, et al: Propranolol treat- stimulants in medically ill patients with neurological disease
ment of assaultive patients with organic brain disease: a double- and major depression. Can J Psychiatry 1984; 29:46–49
blind crossover, placebo-controlled study. J Nerv Ment Dis 1986; 147. Branconnier RJ, Cole JO: The therapeutic role of methylpheni-
174:290–294 date in senile organic brain syndrome. Proc Annu Meet Am Psy-
134. Petrie WM, Ban TA: Propranolol in organic agitation (letter). chopathol Assoc 1980; 69:183–196
Lancet 1981; 1:324 148. Galynker I, Ieronimo C, Miner C, et al: Methylphenidate treat-
135. Shankle WR, Nielson KA, Cotman CW: Low-dose propranolol ment of negative symptoms in patients with dementia. J Neu-
reduces aggression and agitation resembling that associated ropsychiatry Clin Neurosci 1997; 9:231–239
with orbitofrontal dysfunction in elderly demented patients. 149. D’Amato RJ, Zweig RM, Whitehouse PJ, et al: Aminergic sys-
Alzheimer Dis Assoc Disord 1995; 9:233–237 tems in Alzheimer’s disease and Parkinson’s disease. Ann Neu-
136. Yudofsky S, Williams D, Gorman J: Propranolol in the treatment rol 1987; 22:229–236

276 J Neuropsychiatry Clin Neurosci 16:3, Summer 2004

You might also like