Anda di halaman 1dari 17

Neuropathology and Applied Neurobiology (2011), 37, 118–134 doi: 10.1111/j.1365-2990.2010.01131.

Review: The neuropathology of drug abuse _ 118..134

A. Büttner
Institute of Forensic Medicine, University of Rostock, Rostock, Germany

A. Büttner (2011) Neuropathology and Applied Neurobiology 37, 118–134


The neuropathology of drug abuse

Drug abuse represents a significant health issue. The methamphetamine and MDMA has been related to neuro-
major substances abused include cannabis, opiates, toxicity in human long-term abusers and to the risk of
cocaine, amphetamine, methamphetamine and ‘ecstasy’. developing Parkinson’s disease. However, whether such
Alterations of intracellular messenger pathways, tran- neurotoxicity occurs remain to be established. Systematic
scription factors and immediate early genes within the histological, immunohistochemical and morphometric
brain reward system seem to be fundamentally important investigations have shown profound morphological
for the development of addiction and chronic drug abuse. alterations in the brains of polydrug abusers. The major
Genetic risk factors and changes in gene expression asso- findings comprise neuronal loss, neurodegenerative
ciated with drug abuse are still poorly understood. Besides alterations, a reduction of glial fibrillary acidic protein-
cardiovascular complications, psychiatric and neurologic immunopositive astrocytes, widespread axonal damage
symptoms are the most common manifestations of drug with concomitant microglial activation as well as reactive
toxicity. A broad spectrum of changes affecting the central and degenerative changes of the cerebral microvascula-
nervous system is seen in drug abusers. The major find- ture. These observations demonstrate that drugs of abuse
ings result from the consequences of ischaemia and cere- initiate a cascade of interacting toxic, vascular and
brovascular diseases. Except for a few observations of hypoxic factors, which finally result in widespread distur-
vasculitis, the aetiology of these cerebrovascular acci- bances within the complex network of central nervous
dents is not fully understood. The abuse of amphetamine, system cell-to-cell interactions.

Keywords: astrocytes, cerebral microvasculature, drug abuse, microglia, neurodegeneration

part on what is available, most perform polysubstance


Introduction
abuse [4–8].
Drug abuse represents a serious health issue worldwide. The methodological problems in investigating the
Drugs of abuse can be grouped as stimulants, analgesics effects of drugs of abuse on the central nervous system
and narcotics, hypnotics as well as antidepressants, nico- (CNS) consist of distinguishing between substance-
tine and alcohol [1–3]. Besides the latter two substances, specific effects related to the properties of the drug itself,
the predominant substances abused include cannabis, the influence of adulterants, and of secondary effects
opiates, cocaine, amphetamine, methamphetamine and related to the lifestyle of drug abusers, for example, mal-
designer drugs [1,2]. Although individuals with a drug nutrition, infections and systemic diseases. Because
habit may favour one or other class of drug, depending in polysubstance abuse is seen in the majority of cases, it is
difficult to relate the observed findings to a single sub-
stance. Therefore, in most cases the exact aetiology of the
Correspondence: Andreas Büttner, Institute of Forensic Medicine, various CNS alterations remains unclear.
University of Rostock, St.-Georg-Strasse 108, 18055 Rostock,
Germany. Tel: +49 381 494 9900; Fax: +49 381 494 9902; E-mail: Injecting drug abuse is also a risk factor for the
andreas.buettner@med.uni-rostock.de acquisition of HIV-1 infection and there is evidence that
118 © 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society
The neuropathology of drug abuse 119

HIV-1-associated CNS disorders are accentuated in drug tions and hyperintense lesions have been demonstrated
abusers [9,10]. in the white matter, which were attributed to ischaemic
Infection of the brain with hepatitis viruses, especially lesion [34,38–40].
with hepatitis C virus (HCV) is another potential con- Single photon emission computed tomography showed
founding factor in the neuropathological appearances of focal perfusions deficits in various brain regions [41–45].
the observed brain changes. Injection drug abusers are On positron emission tomography a reduction of the
the largest group of persons infected with HCV, with a global glucose metabolism has been demonstrated
prevalence of 50–90% [11]. The transmission of HCV is [46,47].
not the effect of the drug injected but of sharing con- Studies using proton magnetic resonance spectroscopy
taminated equipment [11]. In contrast to HIV, HCV is have identified several biochemical changes in the brain
detectable in high concentrations in other utensils for that may underlie the neuropathology that subsequently
heroin injection, such as filters, spoons and rinsing gives rise to the cognitive and behavioural impairments
liquids. Besides hepatic encephalopathy, there is also associated with drug addiction [48]. Some neurochemical
emerging evidence of neurocognitive impairment in HCV abnormalities have been attributed to alterations in non-
infection independently of drug abuse or HIV-1 infection neuronal brain tissue [48].
[12–15]. Diffusion tensor imaging revealed microstructural
Research on the neurobiology of addiction has shown abnormalities in the white matter and the corpus callo-
that the reinforcing properties of most drugs of abuse are sum, suggestive of axonal injury [49–54].
mediated by activation of the mesolimbic dopaminergic Despite these numerous neuroimaging studies, the
system, the orbitofrontal cortex and the extended cause or possible morphological correlates of these alter-
amygdala [16–19]. Alterations of the intracellular mes- ations are still not fully resolved.
senger pathways, transcription factors and immediate
early gene expression in these reward circuits are believed
CNS effects of the major drugs of abuse
to be important for the development of addiction and
chronic drug abuse [16–20]. Depending of the frequency and types of drugs abused,
Possible genetic risk factors for drug addiction and the predominant neurologic-psychiatric complications
changes in gene expression, associated with drug abuse, of drug abuse include a high prevalence of depression,
are still poorly understood [21,22]. memory loss and cognitive decline, and the possible pre-
Although a broad spectrum of alterations affecting the disposition to schizophrenia.
CNS has been described in drug abusers [1,2,23], there is
no specific change that is characteristic and little is known
Cannabis
about the fundamental neuropathological alterations of
the cellular elements of the human brain. Therefore, Cannabis is the most commonly used illicit drug world-
despite a large body of literature on animal models, the wide [2,55–59].
following review will focus on the relevant human CNS Cannabis preparations (marihuana, hashish) are
findings. The pharmacology and CNS alterations in usually mixed with tobacco and smoked. Oral ingestion as
alcohol and nicotine abuse are reviewed elsewhere tea or addition to pastries is also widespread. Cannabis is
[3,24–27]. unsuitable for intravenous use as it is relatively water-
insoluble. The concentration of the major psychoactive
component of cannabis, D9-tetrahydrocannabinol (THC),
Neuroimaging findings
varies according to the preparation and the origin of the
Neuroimaging studies revealed widespread alterations in plant from 1% to 7% in marihuana, 2% to 10% in hashish
the brains of drug abusers [28]: and 10% to 60% in hash oil [55,57].
Computed tomography studies have shown diffuse Cannabis combines many of the properties of alcohol,
brain atrophy [29–31]. tranquillizers, opiates and hallucinogens; it is anxiolytic,
On magnetic resonance imaging decreased grey and sedative, analgesic and psychedelic [55]. The psychotropic
white matter volume has been demonstrated in particular action of cannabis starts after several minutes reaches a
areas of the brain [32–37]. Moreover, focal demyelina- maximum by 20–30 min and persists for 2–4 h. However,
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
120 A. Büttner

the action of cannabis is of highly individual variability. cannabis-induced hypotension [74–78]. However, due to
Furthermore, there is a dose-related impairment of cogni- the widespread use of this drug, it is difficult and often
tive and psychomotor performance. Acute effects include impossible to establish whether these strokes are truly
euphoria and relaxation with perceptual changes. associated with cannabis, other ingested drugs, or purely
However, dysphoric reactions, including anxiety and coincidental.
panic, depression, paranoia and psychosis can also The important question whether cannabis can cause
occur. Systemic effects include a dose-related tachycardia, irreversible brain damage, particularly to adolescents,
vasodilation and postural hypotension [55–59]. or lead to increased risk for schizophrenia, persisting
THC is a lipid-soluble substance that is distributed het- beyond transient intoxication, is still a matter of debate
erogeneously within the brain, with its highest concentra- [79,80].
tions in neocortical, limbic, sensory and motor areas
[55–58]. THC and other cannabinoids exert their effects
Opioids and derivatives
by the interaction with specific cannabinoid receptors,
which are distributed heterogeneously within the brain Opioids, especially heroin, are the leading substances that
[59–64]. cause death in drug abusers. Derivatives include mor-
Two cannabinoid receptors, CB1 and CB2, have been phine, hydrocodone, oxycodone, hydromorphine, codeine
pharmacologically characterized and anatomically and other narcotics such as fentanyl, meperidene, metha-
localized. CB1 receptors are found predominantly in the done and opium [2,3]. Heroin is made from opium and
central and peripheral nervous system, where they have crosses the blood–brain barrier (BBB) faster than
been implicated in presynaptic inhibition of neurotrans- morphine [81].
mitter release. CB2 receptors are present on immune Heroin is usually administered intravenously. Intrana-
cells, where they may be involved in cytokine release sal and subcutaneous administration is also possible.
[59–65]. Heroin alkaloid may be prepared for inhalation by heating
The highest density of CB1 receptors is found in the on metallic foil (‘chasing the dragon’) that results in a
substantia nigra, the basal ganglia, the hippocampus and higher bioavailability of heroin compared with smoking
the cerebellum [63,66,67]. Within the neocortex they are in a cigarette [3].
present with the highest density in the frontal cortex, the Intravenous heroin induces an extreme euphoria
dentate gyrus, the mesolimbic dopaminergic system and lasting for several minutes. Subsequently, there is a pleas-
the temporal lobe [63,66,67]. This specific distribution of ant dream-like state with drowsiness and analgesia [81].
CB1 receptors correlates well with the effects of cannab- However, marked tolerance develops to euphoria. Opiate
inoids on memory, perception and movement control. The overdose produces the triad of coma, respiratory depres-
very low density of CB1 receptors in the brain stem and sion and miosis [81]. Between 50% and 70% of intrave-
medulla oblongata explains the low acute toxicity and nous heroin abusers have experienced a non-fatal
lack of lethality of cannabis [55,59,66]. Nevertheless, the overdose at some time in their lives [4]. Medical complica-
CNS toxicity of cannabis seems to have been underesti- tions of long-term heroin abuse include thrombophlebitis,
mated for a long time [68], as recent findings revealed endocarditis, hepatitis, pneumonia, nephropathy and
THC-induced neuronal death in cultured rat neurones immunodepression [2,81]. Due to the practise of needle-
[69–71]. sharing, there is a high risk for infections [11].
The discovery of specific endogenous cannabinoid Risk factors for opioid-induced deaths include overdose,
receptor ligands (endocannabinoids), and the distribution concomitant use of other CNS depressants and loss of tol-
of their receptors, strongly suggests that these lipid neu- erance after a period of abstinence [4–8].
rotransmitter systems play an important role in higher At necropsy, up to 90% of all cases of persons dying of
cortical-emotional functions, memory storage, move- heroin overdose show brain oedema with prominent ton-
ment coordination and several pathological conditions sillar herniation and uncal grooving. However, rapid
[59,62–65,72,73]. death after heroin intake will not lead to any morphologi-
Despite its widespread abuse, cannabis-related cere- cal evidence of neuronal cell injury. In cases of delayed
brovascular events are infrequently reported. They are death after a survival period of 5 h or longer, hypoxic-
believed to be due to a cannabis-induced vasospasm or a ischaemic leukoencephalopathy with loss of neurones in
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 121

the hippocampal formation, the Purkinje cell layer and/or


Alterations of neurotransmitters and receptors
the olivary nucleus, as well as vascular congestion, will
become apparent [82–86]. In the globus pallidus, neu- Long-term heroin abuse seems not to be related with a
ronal loss and bilateral, symmetrical ischaemic lesions/ reduced density of CNS m- and d-opioid receptors [103–
necrosis have been described in 5–10% of cases [85,87]. 106]. Instead, the molecular mechanisms underlying
All the above mentioned alterations are assumed to be opiate addiction seem to involve the second messenger
caused rather by recurrent episodes of hypoxia during the signalling system [16,18,19,104,107–114]. Necropsy
intoxicated state than to be related to direct neurotoxic studies revealed that long-term heroin abuse causes an
effects of the opioid drugs [82–87]. increase in certain G proteins in different regions of the
Stroke in heroin abusers, occurring in the absence of brain [105,107,110]. Others have shown a decreased
endocarditis or mycotic aneurysms, has rarely been level of Ca2+-dependent protein kinase C-a [104] and an
observed [84,88–93]. increased level of a membrane-associated G protein-
Within the hippocampus of heroin deaths, enhanced coupled receptor kinase [113].
expression of glial fibrillary acidic protein (GFAP) by astro- Further findings include the downregulation of the
cytes and/or a proliferation of microglia have been found adenylyl-cyclase subtype I [109,111], a decrease in the
[84]. However, an increased GFAP expression could not be density of alpha 2-adrenoreceptors [103], a decrease in
confirmed by other authors [23]. An increase in polysialic the immunoreactivity of protein kinase C-ab [114], and
acid neural cell adhesion molecule positive hippocampal decreased levels of neurofilament proteins [115].
neurones and astrocytes is assumed to reflect an attempt Within the dopaminergic system the levels of tyrosine
to repair cell damage [94]. hydroxylase protein and those of the dopamine (DA)
Perivascular pigment-laden macrophages and pigment metabolite homovanillic acid were reduced in the
depositions are sometimes observed and are attributed to nucleus accumbens [116]. Striatal levels of serotonin
repeated intravenous injections of impure heroin [95], or (5-hydroxytryptamine or 5-HT) were either normal or
to BBB breakdown [23,96]. elevated, whereas the concentration of the 5-HT metabo-
Infections in heroin abusers result from unsterile injec- lite 5-hydroxyindoleacetic acid was decreased [116].
tion techniques and from immunosuppression, caused by According to the authors, chronic heroin abuse might
chronic opiate abuse [1,2,11,85,97]. Endocarditis might produce a modest reduction in dopaminergic and seroton-
lead to septic foci in the brain or to intracranial mycotic ergic activity that could affect motivational state and
aneurysms [1,2,85,97]. impulse control.
Transverse myelitis/myelopathy is an exceptionally rare Maintenance therapy for heroin addiction includes
lesion described in heroin abusers [1,2,85,98,99]. The codeine, dihydrocodeine, methadone and buprenorphine
aetiology is still unclear and neither the clinical picture [117–120]. In the majority of deaths related to these sub-
nor the pathological changes correspond to any particular stances, additional CNS depressant drugs, mainly alcohol
pattern. and benzodiazepines, can be detected. The neuropatho-
A distinct entity, spongiform leukoencephalopathy, has logical findings are similar to those encountered in heroin
been reported to occur almost exclusively after inhalation deaths.
of heroin pyrolysate vapours (‘chasing the dragon’)
[1,2,100–102]. The most characteristic finding is the
Cocaine
selective involvement of the posterior limb of the internal
capsule and sparing of subcortical U-fibres. Neuropatho- Cocaine is derived from the leaves of the coca plant.
logical examination reveals spongiform degeneration of Cocaine hydrochloride is a water-soluble white salt and
the deep white matter with vacuolation of the myelin the most frequent preparation of the drug that is available
sheath, loss of oligodendrocytes, axonal reduction and in form of cystals, granules or a white powder. It can be
astrogliosis. The grey matter is usually unremarkable and administered intranasally (‘snorting’) or injected [2,3].
the brainstem, spinal cord and peripheral nerves are The free alkaloid form (‘free base’) that had been extracted
spared [100–102]. A lipophilic toxin-induced process was with volatile solvents is usually smoked. ‘Crack cocaine’ is
considered to be due to contaminants, but a definite toxin produced by first dissolving the hydrochloride salt in
could not yet be identified. water, then mixing with baking soda and heating. The
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
122 A. Büttner

cracking sound made by the crystals when they are heated pressure and heart rate from the sympathomimetic effect
provides the name [2]. of the drug [92,128–130,132,141,145].
Cocaine initially induces profound subjective well-being A cocaine-induced cerebral vasculitis as a cause for
together with alertness and increased self-confidence. cerebrovascular events could only be demonstrated in rare
Subsequently, there is a mild euphoria followed by fatigue. cases [89,146,147].
Acute psychiatric disturbances include dysphoria, agita- In experimental models, cocaine enhances leucocyte
tion, aggressive behaviour, depression, paranoia, psy- migration across the cerebral vessel wall and opens the
chosis and hallucinations. Medical complications include BBB to HIV-1 invasion by a direct effect on brain endothe-
vasoconstriction, tachycardia, hypertension and sudden lial cells and by the induction of pro-inflammatory
cardiac death [3,25,31]. cytokines and chemokines [148,149].
Cocaine is a potent CNS stimulant. It crosses the BBB
rapidly due to its highly lipophilic properties [121,122].
Alterations of neurotransmitters and receptors
Throughout the brain, cocaine and its major metabolites
are widely distributed and receptors with varying In the striatum of cocaine-related deaths, reductions in
affinities are found [123,124]. Cocaine acts by binding to the levels of enkephalin mRNA, m-opioid receptor binding
specific receptors at presynaptic sites preventing the and DA uptake site binding, along with elevation in levels
reuptake of neurotransmitters [2,3,122]. The major of dynorphin mRNA and k-opioid receptor binding have
synaptic effect of cocaine is the release of dopamine been described [150]. In chronic cocaine abusers, a
(DA) from the synaptic vesicles and the blocking of DA decreased level of DA was identified in the caudate nucleus
reuptake resulting in an enhanced dopaminergic neu- and frontal cortex, but not in the putamen, nucleus
rotransmission [2,3,122]. accumbens and cerebral cortex [151–155]. This decrease
Cocaine is the most frequent drug of abuse associated was not paralleled by an increase of DA D1 and D2 gene
with fatal and non-fatal cerebrovascular events, with expression [156]. Simultaneously, there was an increase
either haemorrhagic or ischaemic strokes [89,91– of cocaine binding sites on the DA transporter with a
93,125–135]. In contrast to the non-drug-using popula- decrease of the DA D1 receptor density in the striatum
tion, cocaine-related stroke occurs primarily in young and of DA D1 and DA D3 receptor density in the nucleus
adults [1,125–128]. accumbens [152–154,157–159]. A marked reduction
Cocaine-related ischaemic infarctions can occur in in immunoreactivity of the vesicular monoamine
every brain region [2,128,130]. The underlying cause is transporter-2 [151] and of the transcription factor
attributed to cerebral vasospasm either by cocaine or its NURR1 [160] in necropsy samples of human cocaine
metabolites [92,128,131,136–138]. Other mechanisms abusers might reflect damage to the dopaminergic system.
include cocaine-induced cardiac arrhythmia with second- An overexpression of a-synuclein in midbrain DA neu-
ary cerebral ischaemia, or the direct effects of cocaine rones in chronic cocaine abusers may occur as a protec-
on haemostasis with increased platelet aggregation tive response to changes in DA turnover and increased
[89,128,131,135,139,140]. oxidative stress resulting from cocaine abuse [161]. This
In cocaine-related intracerebral (ICH) and subarach- accumulation of a-synuclein protein in long-term cocaine
noidal (SAH) haemorrhage, underlying arteriovenous abuse might increase the risk of developing Parkinsonism
malformations or aneurysms are frequently observed [161].
[93,125,127–130,132,141,142]. Cocaine abuse has Further necropsy findings include an upregulation of
been shown to predispose to aneurysmal rupture at a sig- k2-opioid receptors in the limbic system [162] and of
nificantly earlier age, and in much smaller aneurysms, cAMP response element-binding protein in the ventral
compared with non-drug-using persons [125,143]. tegmental area [163].
Cocaine-related ICH is associated more frequently with In the serotonergic system an increase of the 5-HT
subcortical locations, a higher risk of intraventricular transporter in the striatum, substantia nigra and limbic
haemorrhage, and poor prognosis compared with non- system has been demonstrated [164]. In the putamen, a
drug-abusing patients with spontaneous ICH [144]. DA-rich brain area, the activity of phospholipase A2
Rupture of an arteriovenous malformation, or aneurysm, and phosphocholine cytidylyltransferase was selectively
are most likely related to the sudden elevation of blood decreased [165].
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 123

dopaminergic system markers [181,184], the evidence is


Amphetamines and methamphetamine
inconclusive in regard to dopaminergic system degenera-
Amphetamines and methamphetamine are widely used tion. To define methamphetamine abuse as a risk factor in
psychostimulants that act on monoamine transporters Parkinson disease, it would be important to know whether
[2,3]. these alterations represent neurodegenerative changes or
These substances are available as powder, capsules, a drug-induced compensatory response to the disruption
tablets or fluids. Therefore, they can be swallowed, of neurochemical homeostasis [182,183].
‘snorted’, injected intravenously or smoked. The composi- The mechanisms of methamphetamine-induced
tion, purity and dose are highly variable [2]. neurotoxicity are thought to be mediated by multiple
Their potent sympathomimetic effects include an eleva- mechanisms including the generation of free radicals
tion of pulse rate, blood pressure and an increased level and nitric oxide, excitotoxicity, mitochondrial dysfunc-
of alertness. Adverse effects include insomnia, excitability, tion, and the induction of immediate early genes as well
seizures, panic attacks, psychosis and aggressive behav- as transcription factors [175–177,182–185]. However,
iour [2,3,25]. Throughout the brain, methamphetamine whether neurotoxicity occurs in human methamphet-
is heterogeneously distributed [166]. amine and amphetamine abusers remains to be
Amphetamines and methamphetamine are the established.
second most common cause (after cocaine) of strokes
occurring largely in persons younger than 45 years
Amphetamine and methamphetamine
[127]. Furthermore, SAH and ICH have been described
derivatives
[2,3,91,92,125,126,129,167–172]. Similar to cocaine, a
sudden elevation in blood pressure [167,169,171] is pos- Amphetamine and methamphetamine derivatives
tulated as a major mechanism. The vasoconstrictive effect (‘designer drugs’) comprise a broad spectrum of sub-
of both substances may also lead to the development of stances [2,186]. The street name ‘ecstasy’ subsumes
ischaemic infarction [167,171]. Methamphetamine has different hallucinogenic amphetamine derivatives with
been shown to induce inflammatory genes in human MDMA (3,4-methylenedioxymethamphetamine) and
brain endothelial cells [173]. MDE (3,4-methylenedioxyethylamphetamine) being the
main components [187]. These drugs are taken orally in
form of tablets that are usually embossed with a logo
Alterations of neurotransmitters and receptors
[2]. The effects of MDMA and MDEA last about 3–5 h
The neurotoxic effects of amphetamines and metham- and include relaxation, euphoria, sensual enhancement,
phetamine on the dopaminergic system have been reduction of anxiety and emotional closeness to others.
described in various animal species and in humans. These This psychological profile has been called ‘entactogenic’
effects are characterized by desensitization of DA receptor and has the connotation of ‘inducing a feeling of touch
function, marked reduction of DA transporters and of DA with the world within’. In addition, these substances also
levels as well as other dopaminergic axonal markers [174– have stimulant-like and hallucinogenic effects [2,188–
181]. Similar alterations have been observed in the sero- 191]. The effects of MDMA vary according to the dose and
tonergic system [175]. However, the irreversibility of the frequency and duration of use [190]. Adverse effects
these changes has not been established [175]. include headache, nausea, tooth grinding (bruxism),
Although these alterations have been attributed to neu- tachycardia and an increase in body temperature. Fur-
rodegeneration, direct evidence for the loss of nerve ter- thermore, hyperactivity, flight of ideas, depersonalization,
minals and/or their corresponding substantia nigra cell panic attacks, anxiety, depression, agitation, delirium, and
bodies have not been provided unequivocally [175,177]. insomnia may occur [190].
Nevertheless, based on animal studies, there is concern MDMA interacts with various neurotransmitter circuits
that the alterations in the dopaminergic system may but predominantly affects the dopaminergic and seroton-
predispose methamphetamine abusers to develop ergic system [186,188–191]. In human post mortem
Parkinsonism as they age [175,182,183]. Because tissue, a distinct immunopositive reaction for MDMA and
neuroimaging and necropsy studies of human metham- MDA was observed in the white matter in all cortical
phetamine abusers suggest changes in only some regions and in neurones of the basal ganglia, the hypo-
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
124 A. Büttner

thalamus, the hippocampus and the cerebellar vermis, but ations have been attributed to drug-induced respiratory
relatively weak staining of neurones in the brainstem was failure and were therefore considered to be non-specific
seen [192]. [84,220]. However, in most of these studies, there was no
In reports on human fatalities following MDMA con- control group and systematic data on frequency or topog-
sumption, detailed neuropathological examinations were raphy of the lesions are lacking. Subsequent systematic
mostly not performed [193–196]. neuropathological studies of polydrug abusers revealed
Cerebrovascular complications after ‘ecstasy’ consump- ischaemia-independent widespread neuronal loss, a
tion have been described only occasionally [197–203]. In reduction of GFAP-positive astrocytes, an axonal damage
the globus pallidus bilateral hyperintense lesions have with concomitant microglia activation as well as reactive
been found [204,205]. On neuropathological examina- and degenerative vascular changes [23].
tion necrosis of the globus pallidus, diffuse astrogliosis
and spongiform changes of the white matter have been
Neurones
described [205]. Other neuropathological findings in
deaths after ‘ecstasy’ abuse were mainly due to the com- The neuronal loss has been ascribed to be due to a direct
plications of hyperthermia with disseminated intravascu- impairment of neurones by drugs of abuse and, indirectly,
lar coagulopathy and consisted of cerebral oedema, focal to drug-induced damage of astrocytes, axons and the
haemorrhages and nerve cell loss [196]. cerebral vasculature [23]. In support of this finding is the
observation in animal models that drugs of abuse can
induce apoptotic neuronal cell death [69,175,221–225].
Alterations of neurotransmitters and receptors
In human studies drug-induced alterations of neurofila-
In the brains of laboratory animals, including non- ment proteins [115,226,227], of neuronal TUNEL posi-
human primates, exposure to MDMA has been associated tivity [96] or of transcription factors, for example, cAMP
with dopaminergic and especially serotonergic neurotox- response element-binding protein [112] and c-fos [20] is
icity [176,188–191,206–209]. There is strong evidence thought to be an alternative pathway for neuronal loss.
for neurodegeneration and axonal loss, although the In young opiate abusers an increase in the number and
exact mechanism is still unclear [189–191,206–209]. distribution of hyperphosphorylated tau-positive neu-
Current hypotheses include the formation of toxic MDMA rofibrillary pretangles, fully formed tangles and neuritic
metabolites with generation of free radicals as well as oxi- threads has been reported [228]. This, together with
dative stress, excitotoxicity, apoptosis and mitochondrial reports of occasional ubiquitin-positive inclusions in
dysfunction [176,209–212]. neurones of drug abusers [96], indicates drug-related
Although in recent years, the question of ‘ecstasy’- neurodegeneration. Whether these changes are poten-
induced neurotoxicity and possible functional sequelae tially reversible is still unclear.
has been addressed in several studies, the extent to which Within the substantia nigra there was a decrease of the
these animal and non-human primate data are applicable numerical density of the pigmented neurones, whereas
to humans and whether persistent neurotoxicity occurs in the density of the non-pigmented neurones was
humans is still controversial [42,207–209,213–219]. unchanged [23]. The presence of eosinophilic ubiquiti-
To date, the most consistent findings associate subtle nated cytoplasmic inclusions, which did not resemble
cognitive, particularly memory, impairments with heavy classic Lewy bodies, and which are negative for
ecstasy abuse [213,218,219]. a-synuclein, indicates abnormal cytoplasmic protein
sequestration [96]. To date it is not clear whether this
finding represents a transient neuronal abnormality
Neuropathological studies
or commitment to an irreversible neurodegenerative
The consequences of drugs of abuse on the cellular ele- pathway [96].
ments of the human brain have not been studied system-
atically. There are only few reports on histopathological
White matter
alterations in the brains of human drug abusers describ-
ing oedema, vascular congestion, ischaemic nerve cell In the white matter of polydrug abusers a widespread
damage and neuronal loss [83–85,96,220]. These alter- axonal damage has been demonstrated by using b-APP
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 125

negative drug abusers and non-drug using controls there


was no statistical difference between these groups in rela-
tion to astrocytes [231,234].
In the brains of polydrug abusers the numerical density
of GFAP-positive astrocytes has been shown to be reduced
[23]. This reduction of GFAP-positive astrocytes is believed
to be due to the interference of drugs with the GFAP gene
transcription, inducing an altered GFAP phosphorylation
[235], as well as the generation of free radicals by induc-
tion of a cytochrome P450 isoform [236]. I2-imidazoline
receptors are involved in the regulation of the GFAP
expression [226]. In the frontal cortex of heroin deaths
Figure 1. Immunohistochemistry demonstrating the density of I2-imidazoline receptors and the immu-
b-APP-immunopositive bundles and b-APP-immunopositive noreactivity of the related imidazoline receptor protein
globular deposits in the pons of polydrug abusers, counterstained
were decreased [237].
with haematoxylin, original magnification ¥100.

immunohistochemistry [23,229]. The alterations con- Vascular changes


sisted of b-APP-immunopositive bundles and globular In AIDS patients concentric small-vessel wall thickening,
depositions (Figure 1), but they never showed the exten- perivascular space dilatation, pigment deposition, vessel
sive pattern seen in traumatic brain injury [23,229]. wall mineralization and perivascular inflammatory cell
Furthermore, there is a concomitant activation of infiltrates were seen in 50% of the former drug abusers
microglia predominantly in the white matter and in [238]. Similar vascular alterations can be observed in
most subcortical regions [23,83,220,229]. This upregula- HIV-1-negative polydrug abusers (Figure 2A) [23,96].
tion of microglia has also been shown in drug abusers The presence of acute and chronic BBB breakdown in
with and without pre-symptomatic HIV-1 infection drug abusers, suggests that the brain parenchyma is
[230,231]. Activated microglial cells are a source of pro- exposed to unusual quantities of serum proteins, includ-
inflammatory cytokines, which are linked to neuronal ing immunoglobulins, as well as other blood-borne
damage and loss [10]. The axonal injury suggests a toxic- factors, including HIV [96]. It may further be the stimu-
metabolic drug effect, as there were no sufficient findings lus for the occasional perivascular lymphocytic aggre-
for a secondary phenomenon in these cases, neither due to gates that are seen in the brains of HIV-1-negative
a generalized hypoxic-ischaemic condition, nor to a brain drug abusers (Figure 2B) [10]. Furthermore, reactive
oedema and the concomitant activation of microglia endothelial cell proliferation, degenerative hyalinotic
is indicative of a long-standing progressive process thickening, marked endothelial swelling and endothelial
[23,229]. These alterations might be the morphological cell hyperplasia are present in the brains of polydrug
correlate of the observed demyelination and hyperintense abusers (Figure 2C,D) [23]. In addition, a decrease of the
areas seen on magnetic resonance imaging. collagen type IV content of the vascular basal lamina
[239] and a disruption of the BBB tight junctions [10]
have been observed. This non-inflammatory vasculopa-
Astrocytes
thy has been interpreted as the morphological substrate
Although astrocytes play an essential role, for example, in of a disturbed BBB and might be the morphological
the maintenance of BBB, regulation of glutamatergic substrate of the alterations seen on neuroimaging
neurotransmission and neurotransmitter metabolism, [23,240].
little is known about alterations of these cells in human In conclusion, drugs of abuse initiate a cascade of inter-
drug abusers [232,233]. In an older study ‘widespread acting toxic, vascular and hypoxic factors that finally
fragmentation’ and a numerical depletion of astrocytes in result in widespread disturbances within the complex
the white matter have been reported in the brains of drug network of CNS cell interactions (Figure 3). However,
deaths [85]. In HIV-1-positive opiate abusers, HIV-1- much work will need to be done to clarify the yet
© 2011 The Author
Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
126 A. Büttner

A
B

C
D

Figure 2. Photomicrographs illustrating the spectrum of cerebral vascular changes in polydrug abusers. (A) Small artery in occipital white
matter showing concentric wall thickening. The surrounding perivascular space contains occasional macrophages and pigment deposition.
H&E, original magnification ¥200. (B) Small vessel in the orbital white matter with perivascular lymphocytic aggregates. H&E, original
magnification ¥200. (C) Endothelial proliferation in the dentate nucleus, H&E, original magnification ¥200. (D) Endothelial hyperplasia in
the parietal white matter, H&E, original magnification ¥200.

unresolved questions of the disastrous role of drugs of


abuse on the CNS.

Acknowledgements
The help of Claire Delbridge in correcting the manuscript
is highly appreciated. I thank Jeanne Bell, Institute of
Neuropathology, University of Edinburgh for critical dis-
cussions and Serge Weis, Laboratory of Neuropathology,
Neuropsychiatric Hospital Wagner-Jauregg, Linz, Austria
for his long-standing support and friendship.

References

Figure 3. The consequences of drugs of abuse on the cellular 1 Büttner A, Weis S. Central nervous system alterations
elements of the CNS. in drug abuse. In Forensic Pathology Reviews, Vol. 1. Ed. M
Tsokos. Totowa, NJ: Humana Press, 2004; 79–136

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 127

2 Karch SB. Karch’s Pathology of Drug Abuse 4th edn. Boca 19 Nestler EJ. Molecular mechanisms of drug addiction.
Raton: CRC Press, 2008 Neuropharmacology 2004; 47: 24–32
3 Quinn DI, Wodak A, Day RO. Pharmacokinetic and 20 Harlan RE, Garcia MM. Drugs of abuse and immediate-
pharmacodynamic principles of illicit drug use and early genes in the forebrain. Mol Neurobiol 1998; 16:
treatment of illicit drug users. Clin Pharmacokinet 1997; 221–67
33: 344–400 21 Kuhar MJ, Joyce A, Dominguez G. Genes in drug abuse.
4 Coffin PO, Galea S, Ahern J, Leon AC, Vlahov D, Tardiff Drug Alcohol Depend 2001; 62: 157–62
K. Opiates, cocaine and alcohol combinations in acci- 22 Nestler EJ, Landsman D. Learning about addiction from
dental drug overdose deaths in New York City, 1990- the genome. Nature 2001; 409: 834–5
1998. Addiction 2003; 98: 739–47 23 Büttner A, Weis S. Neuropathological alterations in
5 Darke S. Polydrug use and overdose: overthrowing old drug abusers: the involvement of neurons, glial, and
myths. Addiction 2003; 98: 711 vascular systems. Forensic Sci Med Pathol 2006; 2:
6 Gerostamoulos J, Staikos V, Drummer OH. Heroin- 115–26
related deaths in Victoria: a review of cases for 1997 and 24 Büttner A, Weis S. Central nervous system alterations in
1998. Drug Alcohol Depend 2001; 61: 123–7 alcohol abuse. In Forensic Pathology Reviews, Vol. 5. Ed. M
7 Preti A, Miotto P, De Coppi M. Deaths by unintentional Tsokos. Totowa, NJ: Humana Press, 2008; 69–89.
illicit drug overdose in Italy, 1984-2000. Drug Alcohol 25 Trelles L, Jeri R. Central nervous system stimulants:
Depend 2002; 66: 275–82 cocaine, amphetamine, nicotine. In Handbook of Clinical
8 Steentoft A, Teige B, Holmgren P, Vuori E, Kristinsson J, Neurology, Vol. 65: Intoxications of the Nervous System,
Hansen AC, Ceder G, Wethe G, Rollmann D. Fatal poi- Part II. Eds PJ Vinkens, GW Bruyn, FA de Wolff. Amster-
soning in Nordic drug addicts in 2002. Forensic Sci Int dam: Elsevier, 1995; 251–72.
2006; 160: 148–56 26 Harper C. The neuropathology of alcohol-related brain
9 Anthony IC, Arango JC, Stephens B, Simmonds P, Bell damage. Alcohol Alcohol 2009; 44: 136–40
JE. The effects of illicit drugs on the HIV infected brain. 27 Yildiz D. Nicotine, its metabolism and an overview of its
Front Biosci 2008; 13: 1294–307 biological effects. Toxicon 2004; 43: 619–32
10 Bell JE, Arango JC, Anthony IC. Neurobiology of mul- 28 Kaufman MJ. Brain Imaging in Substance Abuse: Research,
tiple insults: HIV-1-associated brain disorders in those Clinical, and Forensic Applications. Totowa, NJ: Humana
who use illicit drugs. J NeuroImmune Pharmacol 2006; 1: Press, 2001
182–91 29 Pascual-Leone A, Dhuna A, Anderson DC. Cerebral
11 Backmund M, Reimer J, Meyer K, Gerlach JT, Zachoval atrophy in habitual cocaine users. A planimetric CT
R. Hepatitis C virus infection and injection drug users: study. Neurology 1991; 41: 34–8
prevention, risk factors, and treatment. Clin Infect Dis 30 Pezawas LM, Fischer G, Diamant K, Schneider C, Schin-
2005; 40 (Suppl. 5): S330–5 dler SD, Thurnher M, Ploechl W, Eder H, Kasper S. Cere-
12 Cherner M, Letendre SL, Heaton RK, Durelle J, Marquie- bral CT findings in male opioid-dependent patients:
Beck J, Gragg B, Grant I. Hepatitis C augments cognitive stereological, planimetric and linear measurements.
deficits associated with HIV infection and methamphet- Psychiatry Res Neuroimag 1998; 83: 139–47
amine. Neurology 2005; 64: 1343–7 31 Strang J, Gurling H. Computerized tomography and
13 Forton DM, Taylor-Robinson SD, Thomas HC. Central neuropsychological assessment in long-term high-dose
nervous system changes in hepatitis C virus infection. heroin addicts. Br J Addict 1989; 84: 1011–19
Eur J Gastroenterol Hepatol 2006; 18: 333–8 32 Bartzokis G, Beckson M, Lu PH, Edwards N, Rapoport R,
14 Martin-Thormeyer EM, Paul RH. Drug abuse and hepa- Wiseman E, Bridge P. Age-related brain volume reduc-
titis C infection as comorbid features of HIV associated tions in amphetamine and cocaine addicts and normal
neurocognitive disorder: neurocognitive and neuroim- controls: implications for addiction research. Psychiatry
aging features. Neuropsychol Rev 2009; 19: 215– Res 2000; 98: 93–102
21 33 Berman S, O’Neill J, Fears S, Bartzokis G, London ED.
15 Morgello S. The nervous system and hepatitis C virus. Abuse of amphetamines and structural abnormalities in
Semin Liver Dis 2005; 25: 118–21 the brain. Ann N Y Acad Sci 2008; 1141: 195–220
16 Cunha-Oliveira T, Rego AC, Oliveira CR. Cellular and 34 Lim KO, Wozniak JR, Mueller BA, Franc DT, Specker SM,
molecular mechanisms involved in the neurotoxicity of Rodriguez CP, Silverman AB, Rotrosen JP. Brain macro-
opioid and psychostimulant drugs. Brain Res Brain Res structural and microstructural abnormalities in cocaine
Rev 2008; 58: 192–208 dependence. Drug Alcohol Depend 2008; 92: 164–72
17 Hyman SE, Malenka RC, Nestler EJ. Neural mechanisms 35 Lyoo IK, Pollack MH, Silveri MM, Ahn KH, Diaz CI,
of addiction: the role of reward-related learning and Hwang J, Kim SJ, Yurgelun-Todd D, Kaufman MJ,
memory. Annu Rev Neurosci 2006; 29: 565–98 Renshaw PF. Prefrontal and temporal gray matter
18 Koob GF, Volkow ND. Neurocircuitry of addiction. density decreases in opiate dependence. Psychopharma-
Neuropsychopharmacology 2010; 35: 217–38 cology (Berl) 2006; 184: 139–44

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
128 A. Büttner

36 Schlaepfer TE, Lancaster E, Heidbreder R, Strain EC, imaging and their relevance to addiction. Ann N Y Acad
Kosel M, Fisch H-U, Pearlson GD. Decreased frontal Sci 2010; 1187: 148–71
white-matter volume in chronic substance abuse. Int J 49 Alicata D, Chang L, Cloak C, Abe K, Ernst T. Higher
Neuropsychopharmacol 2007; 9: 147–53 diffusion in striatum and lower fractional anisotropy in
37 Yücel M, Solowij N, Respondek C, Whittle S, Fornito A, white matter of methamphetamine users. Psychiatry
Pantelis C, Lubman DI. Regional brain abnormalities Res Neuroimag 2009; 174: 1–8
associated with long-term heavy cannabis use. Arch Gen 50 Arnone D, Barrick TR, Chengappa S, Mackay CE, Clark
Psychiatry 2008; 65: 694–701 CA, Abou-Saleh MT. Corpus callosum damage in heavy
38 Bae SC, Lyoo IK, Sung YH, Yoo J, Chung A, Yoon S-J, marijuana use: preliminary evidence from diffusion
Kim D-J, Hwang J, Kim SJ, Renshaw PF. Increased white tensor tractography and tract-based spatial statistics.
matter hyperintensities in male methamphetamine Neuroimage 2008; 41: 1067–74
abusers. Drug Alcohol Depend 2006; 81: 83–8 51 Liu H, Li L, Hao Y, Cao D, Xu L, Rohrbaugh R, Xue Z,
39 Bartzokis G, Goldstein IB, Hance DB, Beckson M, Hao W, Shan B, Liu Z. Disrupted white matter integrity
Shapiro D, Lu PH, Edwards N, Mintz J, Bridge P. The in heroin dependence: a controlled study utilizing diffu-
incidence of T2-weighted MR imaging signal abnor- sion tensor imaging. Am J Drug Alcohol Abuse 2008; 34:
malities in the brain of cocaine-dependent patients is 562–74
age-related and region-specific. AJNR Am J Neuroradiol 52 Ma L, Hasan KM, Steinberg JL, Narayana PA, Lane SD,
1999; 20: 1628–35 Zuniga EA, Kramer LA, Moeller FG. Diffusion tensor
40 Lyoo IK, Streeter CC, Ahn KH, Lee HK, Pollack MH, imaging in cocaine dependence: regional effects of
Silveri MM, Nassar LE, Levin JM, Sarid-Segal O, Ciraulo cocaine on corpus callosum and effect of cocaine
DA, Renshaw PF, Kaufman MJ. White matter hyperin- administration route. Drug Alcohol Depend 2009; 104:
tensities in subjects with cocaine and opiate dependence 262–7
and healthy comparison subjects. Psychiatry Res Neu- 53 Romero MJ, Asensio S, Palau C, Sanchez A, Romero FJ.
roimag 2004; 131: 135–45 Cocaine addiction: diffusion tensor imaging study of the
41 Botelho MF, Relvas JS, Abrantes M, Cunha MJ, Marques inferior frontal and anterior cingulate white matter.
TR, Rovira E, Fontes Ribeiro CA, Macedo T. Brain blood Psychiatry Res Neuroimag 2010; 181: 57–63
flow SPET imaging in heroin abusers. Ann N Y Acad Sci 54 Tobias MC, O’Neill J, Hudkins M, Bartzokis G, Dean AC,
2006; 1074: 466–77 London ED. White-matter abnormalities in brain during
42 Chang L, Grob CS, Ernst T, Itti L, Mishkin FS, Jose- early abstinence from methamphetamine abuse. Psy-
Melchor R, Poland RE. Effect of ecstasy [3,4- chopharmacology (Berl) 2010; 209: 13–24
methylenedioxymethamphetamine (MDMA)] on 55 Ashton CH. Pharmacology and effects of cannabis: a
cerebral blood flow: a co-registered SPECT and MRI brief review. Br J Psychiatry 2001; 178: 101–6
study. Psychiatry Res 2000; 98: 15–28 56 Iversen L. Cannabis and the brain. Brain 2003; 126:
43 Ernst T, Chang L, Oropilla G, Gustavson A, Speck O. 1252–79
Cerebral perfusion abnormalities in abstinent cocaine 57 Johns A. Psychiatric effects of cannabis. Br J Psychiatry
abusers: a perfusion MRI and SPECT study. Psychiatry 2001; 178: 116–22
Res 2000; 99: 63–74 58 Nahas GG. The pharmacokinetics of THC in fat and
44 Gottschalk PC, Kosten TR. Cerebral perfusion defects in brain: resulting functional responses to marihuana
combined cocaine and alcohol dependence. Drug Alcohol smoking. Hum Psychopharmacol Clin Exp 2001; 16:
Depend 2002; 68: 95–104 247–55
45 O’Leary DS, Block RI, Koeppel JA, Flaum M, Schultz SK, 59 Ameri A. The effects of cannabinoids on the brain. Prog
Andreasen NC, Boles Ponto LL, Watkins GL, Hurtig RR, Neurobiol 1999; 58: 315–48
Hichwa RD. Effects of smoking marijuana on brain per- 60 Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P,
fusion and cognition. Neuropsychopharmacology 2002; Devane WA, Felder CC, Herkenham M, Mackie K, Martin
26: 802–16 BR, Mechoulam R, Pertwee RG. Classification of can-
46 London ED, Cascella NG, Wong DF, Phillips RL, Dannals nabinoid receptors. Pharmacol Rev 2002; 54: 161–202
RF, Links JM, Herning R, Grayson R, Jaffe JH, Wagner 61 Pertwee RG. Pharmacology of cannabinoid CB1 and
HN Jr. Cocaine-induced reduction of glucose utilization CB2 receptors. Pharmacol Ther 1997; 74: 129–80
in human brain. Arch Gen Psychiatry 1990; 47: 567– 62 Fride E. Endocannabinoids in the central nervous
74 system - an overview. Prostaglandins Leukot Essent Fatty
47 Volkow ND, Fowler JS, Wolf AP, Hitzemann R, Dewey S, Acids 2002; 66: 221–33
Bendriem B, Alpert R, Hoff A. Changes in brain glucose 63 Glass M, Dragunow M, Faull RLM. Cannabinoid recep-
metabolism in cocaine dependence and withdrawal. Am tors in the human brain: a detailed anatomical and
J Psychiatry 1991; 148: 621–6 quantitative autoradiographic study in the fetal neona-
48 Licata SC, Renshaw PF. Neurochemistry of drug action: tal and adult human brain. Neuroscience 1997; 77:
insights from proton magnetic resonance spectroscopic 299–318

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 129

64 Childers SR, Breivogel CS. Cannabis and endogenous System, Part II. Eds PJ Vinkens, GW Bruyn, FA de Wolff.
cannabinoid systems. Drug Alcohol Depend 1998; 51: Amsterdam: Elsevier, 1995; 349–61
173–87 82 Ginsberg MD, Hedley-Whyte ET, Richardson EP Jr.
65 Wilson RI, Nicoll RA. Endocannabinoid signaling in the Hypoxic-ischemic leukoencephalopathy in man. Arch
brain. Science 2002; 296: 678–82 Neurol 1976; 33: 5–14
66 Herkenham M. Cannabinoid receptor localization in 83 Gosztonyi G, Schmidt V, Nickel R, Rothschild MA,
brain: relationship to motor and reward systems. Ann N Camacho S, Siegel G, Zill E, Pauli G, Schneider V. Neu-
Y Acad Sci 1992; 654: 19–32 ropathologic analysis of postmortal brain samples of
67 Mailleux P, Parmentier M, Vanderhaeghen J-J. Distribu- HIV-seropositive and -seronegative i.v. drug addicts.
tion of cannabinoid receptor messenger RNA in the Forensic Sci Int 1993; 62: 101–5
human brain: an in situ hybridization histochemistry 84 Oehmichen M, Meißner C, Reiter A, Birkholz M.
with oligonucleotides. Neurosci Lett 1992; 143: 200–4 Neuropathology in non-human immunodeficiency
68 Scallet AC. Neurotoxicology of cannabis and THC: a virus-infected drug addicts: hypoxic brain damage after
review of chronic exposure studies in animals. Pharma- chronic intravenous drug abuse. Acta Neuropathol 1996;
col Biochem Behav 1991; 40: 671–6 91: 642–6
69 Campbell VA. Tetrahydrocannabinol-induced apoptosis 85 Pearson J, Richter RW. Addiction to opiates: neurologic
of cultured cortical neurones is associated with cyto- aspects. In Handbook of Clinical Neurology. Intoxications
chrome c release and caspase-3 activation. Neurophar- of the Nervous System, Part II. Eds PJ Vinken, GW Bruyn.
macology 2001; 40: 702–9 Amsterdam: North-Holland Publishing Company,
70 Chan GCK, Hinds TR, Impey S, Storm DR. Hippocampal 1979; 365–400
neurotoxicity of delta9-tetrahydrocannabinol. J Neurosci 86 Protass LM. Delayed postanoxic encephalopathy after
1998; 18: 5322–32 heroin use. Ann Intern Med 1971; 74: 738–9
71 Guzmán M, Sánchez C, Galve-Roperh I. Control of the 87 Andersen SN, Skullerud K. Hypoxic/ischaemic brain
cell survival/death decision by cannabinoids. J Mol Med damage, especially pallidal lesions, in heroin addicts.
2001; 78: 613–25 Forensic Sci Int 1999; 102: 51–9
72 Di Marzo V. Regulation of endocannabinoid levels 88 Adle-Biassette H, Marc B, Benhaiem-Sigaux N, Durigon
under physiological and pathological conditions. A M, Gray F. Infarctus cérébraux chez un toxicomane
mini-review. Pharm Pharmacol Commun 2000; 6: inhalant l’héroine. Arch Anat Cytol Pathol 1996; 44:
235–41 12–17
73 Tanda G, Goldberg SR. Cannabinoids: reward, depen- 89 Brust JCM. Clinical, radiological, and pathological
dence, and underlying neurochemical mechanisms - a aspects of cerebrovascular disease associated with drug
review of recent preclinical data. Psychopharmacology abuse. Stroke 1993; 24 (Suppl. I): I-129-33
(Berl) 2003; 169: 115–34 90 Brust JCM, Richter RW. Stroke associated with addiction
74 Barnes D, Palace J, O’Brien MD. Stroke following mari- to heroin. J Neurol Neurosurg Psychiatry 1976; 39:
juana smoking. Stroke 1992; 23: 1381 194–9
75 Mateo I, Pinedo A, Gomez-Beldarrain M, Basterretxea 91 Caplan LR, Hier DB, Banks G. Current concepts of cere-
JM, Garcia-Monco JC. Recurrent stroke associated with brovascular disease - stroke: stroke and drug abuse.
cannabis use. J Neurol Neurosurg Psychiatry 2005; 76: Stroke 1982; 13: 869–72
435–7 92 Kelly MA, Gorelick PB, Mirza D. The role of drugs in
76 Mouzak A, Agathos P, Kerezoudi E, Mantas A, Vourdeli- the etiology of stroke. Clin Neuropharmacol 1992; 15:
Yiannakoura E. Transient ischemic attack in heavy can- 249–75
nabis smokers - how ‘safe’ is it? Eur Neurol 2000; 44: 93 Sloan MA, Kittner SJ, Rigamonti D, Price TR. Occur-
42–4 rence of stroke associated with use/abuse of drugs.
77 Thanvi BR, Treadwell SD. Cannabis and stroke: is there a Neurology 1991; 41: 1358–64
link? Postgrad Med J 2009; 85: 80–3 94 Weber M, Modemann S, Schipper P, Trauer H, Franke H,
78 Zachariah SB. Stroke after heavy marijuana smoking. Illes P, Geiger KD, Hengstler JG, Kleemann WJ.
Stroke 1991; 22: 406–9 Increased polysialic acid neural cell adhesion molecule
79 DeLisi LE. The effect of cannabis on the brain: can it expression in human hippocampus of heroin addicts.
cause brain anomalies that lead to increased risk for Neuroscience 2006; 138: 1215–23
schizophrenia? Curr Opin Psychiatry 2008; 21: 140–50 95 Gray F, Lescs MC, Keohane C, Paraire F, Marc B, Durigon
80 Moore THM, Zammit S, Lingford-Hughes A, Barnes M, Gherardi R. Early brain changes in HIV infection:
TRE, Jones PB, Burke M, Lewis G. Cannabis use and risk neuropathological study of 11 seropositive, non-AIDS
of psychotic or affective mental health outcomes: a sys- cases. J Neuropathol Exp Neurol 1992; 51: 177–
tematic review. Lancet 2007; 370: 319–28 85
81 Brust JCM. Opiate addiction and toxicity. In Handbook of 96 Bell JE, Arango JC, Robertson R, Brettle RP, Leen C,
Clinical Neurology, Vol. 65: Intoxications of the Nervous Simmonds P. HIV and drug misuse in the Edinburgh

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
130 A. Büttner

cohort. J Acquir Immune Defic Syndr 2002; 31 (Suppl. 2): postmortem brains of heroin addicts. Arch Gen Psychia-
S35–42 try 1994; 51: 494–501
97 Adelman LS, Aronson SM. The neuropathologic compli- 111 Shichinohe S, Ozawa H, Saito T, Hashimoto E, Lang C,
cations of narcotic drug addiction. Bull N Y Acad Med Riederer P, Takahata N. Differential alteration of adenyl
1969; 45: 225–34 cyclase subtypes I, II, and V/VI in postmortem human
98 McCreary M, Emerman C, Hanna J, Simon J. Acute brains of heroin addicts. Alcohol Clin Exp Res 1998; 22:
myelopathy following intranasal insufflation of heroin: 84S–7S
a case report. Neurology 2000; 55: 316–17 112 Lane-Ladd SB, Pineda J, Boundy VA, Pfeuffer T, Krupin-
99 Pearson J, Richter RW, Baden MM, Challenor YB, Bruun ski J, Aghajanian GK, Nestler EJ. CREB (cAMP response
B. Transverse myelopathy as an illustration of the neu- element-binding protein) in the locus coeruleus: bio-
rologic and neuropathologic features of heroin addic- chemical, physiological, and behavioral evidence for a
tion. Hum Pathol 1972; 3: 107–13 role in opiate dependence. J Neurosci 1997; 17: 7890–
100 Kriegstein AR, Armitage BA, Kim PY. Heroin inhalation 901
and progressive spongiform leukoencephalopathy. N 113 Ozaita A, Escribá PV, Ventayol P, Murga C, Mayor F Jr,
Engl J Med 1997; 336: 589–90 García-Sevilla JA. Regulation of g protein-coupled
101 Rizzuto N, Morbin M, Ferrari S, Cavallaro T, Sparaco M, receptor kinase 2 in brains of opiate-treated rats and
Boso G, Gaetti L. Delayed spongiform leukoencephal- human opiate addicts. J Neurochem 1998; 70: 1249–57
opathy after heroin abuse. Acta Neuropathol 1997; 94: 114 Busquets X, Escribá PV, Sastre M, García-Sevilla JA. Loss
87–90 of protein kinase C-ab in brain of heroin addicts and
102 Wolters EC, Stam FC, Lousberg RJ, van Wijngaarden morphine-dependent rats. J Neurochem 1995; 64:
GK, Rengelink H, Schipper MEI, Verbeeten B. Leucoen- 247–52
cephalopathy after inhalating ‘heroin’ pyrolysate. 115 García-Sevilla JA, Ventayol P, Busquets X, La Harpe R,
Lancet 1982; 320: 1233–7 Walzer C, Guimón J. Marked decrease of immunola-
103 Gabilondo AM, Meana JJ, Barturen F, Sastre M, García- belled 68 kDa neurofilament (NF-L) proteins in brains of
Sevilla JA. m-opioid receptor and a2-adrenoreceptor opiate addicts. Neuroreport 1997; 8: 1561–70
agonist binding sites in the postmortem brain of heroin 116 Kish SJ, Kalasinsky KS, Derkach P, Schmunk GA,
addicts. Psychopharmacology (Berl) 1994; 115: 135–40 Guttman M, Ang L, Adams V, Furukawa Y, Haycock
104 García-Sevilla JA, Ventayol P, Busquets X, La Harpe R, JW. Striatal dopaminergic and serotonergic markers in
Walzer C, Guimón J. Regulation of immunolabelled human heroin users. Neuropsychopharmacology 2001;
mu-opioid receptors and protein kinase C-alpha and 24: 561–7
zeta isoforms in the frontal cortex of human opiate 117 Gerostamoulos J, Burke MP, Drummer OH. Involvement
addicts. Neurosci Lett 1997; 226: 29–32 of codeine in drug-related deaths. Am J Forensic Med
105 Meana JJ, González-Maeso J, García-Sevilla JA, Guimón Pathol 1996; 17: 327–35
J. m-Opioid receptor and a2-adrenoreceptor agonist 118 Karch SB, Stephens BG. Toxicology and pathology of
stimulation of [35S]GTPgS binding to G-proteins in deaths related to methadone: retrospective review. West
postmortem brains of opioid addicts. Mol Psychiatry J Med 2000; 172: 11–14
2000; 5: 308–15 119 Kintz P. A new series of 13 buprenorphine-related
106 Schmidt P, Schmolke C, Musshoff F, Menzen M, Pro- deaths. Clin Biochem 2002; 35: 513–16
haska C, Madea B. Area-specific increased density of 120 Milroy CM, Forrest ARW. Methadone deaths: a toxico-
m-opioid receptor immunoreactive neurons in the cere- logical analysis. J Clin Pathol 2000; 53: 277–81
bral cortex of drug-related fatalities. Forensic Sci Int 121 Prakash A, Das G. Cocaine and the nervous system. Int
2003; 133: 204–11 J Clin Pharmacol Ther Toxicol 1993; 31: 575–81
107 Hashimoto E, Frölich L, Ozawa H, Saito T, Shichinohe 122 White SM, Lambe CJT. The pathophysiology of cocaine
S, Takahata N, Riederer P. Alteration of guanosine abuse. J Clin Forensic Med 2003; 10: 27–39
triphosphate binding proteins in postmortem brains 123 Biegon A, Dillon K, Volkow ND, Hitzemann RJ, Fowler
of heroin addicts. Alcohol Clin Exp Res 1996; 20: JS, Wolf AP. Quantitative autoradiography of cocaine
301A–4A binding sites in human brain postmortem. Synapse
108 Law PY, Wong YH, Loh HH. Molecular mechanisms 1992; 10: 126–30
and regulation of opioid receptor signaling. Annu Rev 124 Kalasinsky KS, Bosy TZ, Schmunk GA, Ang L, Adams V,
Pharmacol Toxicol 2000; 40: 389–430 Gore SB, Smialek J, Furukawa Y, Guttman M, Kish SJ.
109 Shichinohe S, Ozawa H, Hashimoto E, Tatschner T, Regional distribution of cocaine in postmortem brain of
Riederer P, Saito T. Changes in the cAMP-related signal chronic human cocaine users. J Forensic Sci 2000; 45:
transduction mechanism in postmortem human brains 1041–8
of heroin addicts. J Neural Transm 2001; 108: 335–47 125 McEvoy AW, Kitchen ND, Thomas DGT. Intracerebral
110 Escribá PV, Sastre M, García-Sevilla JA. Increased haemorrhage and drug abuse in young adults. Br J
density of guanine nucleotide-binding proteins in the Neurosurg 2000; 14: 449–54

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 131

126 Petitti DB, Sidney S, Quesenberry C, Bernstein A. Stroke 142 Peterson PL, Roszler M, Jacob I, Wilner HI. Neurovascu-
and cocaine or amphetamine use. Epidemiology 1998; lar complications of cocaine abuse. J Neuropsychiatry
9: 596–600 Clin Neurosci 1991; 3: 143–9
127 Kaku DA, Lowenstein DH. Emergence of recreational 143 Vannemreddy P, Caldito G, Willis B, Nanda A. Influence
drug abuse as a major risk factor for stroke in young of cocaine on ruptured intracranial aneurysms: a case
adults. Ann Intern Med 1990; 113: 821–7 control study of poor prognostic indicators. J Neurosurg
128 Levine SR, Brust JCM, Futrell N, Brass LM, Blake D, 2008; 108: 470–6
Fayad P, Schultz LR, Millikan CH, Ho K-L, Welch KMA. 144 Martin-Schild S, Albright KC, Hallevi H, Barreto AD,
A comparative study of the cerebrovascular complica- Philip M, Misra V, Grotta JC, Savitz SI. Intracerebral
tions of cocaine: alkaloidal versus hydrochloride - a hemorrhage in cocaine users. Stroke 2010; 41: 680–4
review. Neurology 1991; 41: 1173–7. 145 Oyesiku NM, Colohan ART, Barrow DL, Reisner A
129 Davis GD, Swalwell CI. The incidence of acute cocaine (1993) Cocaine-induced aneurysmal rupture: an emer-
or methamphetamine intoxication in deaths due to rup- gent negative factor in the natural history of intracra-
tured cerebral (berry) aneurysms. J Forensic Sci 1996; nial aneurysms? Neurosurgery 1993; 32: 518–26
41: 626–8 146 Merkel PA, Koroshetz WJ, Irizarry MC, Cudkowicz ME.
130 Fessler RD, Esshaki CM, Stankewitz RC, Johnson RR, Cocaine-associated cerebral vasculitis. Semin Arthritis
Diaz FG. The neurovascular complications of cocaine. Rheum 1995; 25: 172–83
Surg Neurol 1997; 47: 339–45 147 Díez-Tejedor E, Frank A, Gutiérrez M, Barreiro P.
131 Konzen JP, Levine SR, Garcia JH. Vasospasm and Encephalopathy and biopsy-proven cerebrovascular
thrombus formation as possible mechanism of stroke inflammatory changes in a cocaine abuser. Eur J Neurol
related to alkaloidal cocaine. Stroke 1995; 26: 1114– 1998; 5: 103–7
18 148 Gan X, Zhang L, Berger O, Stins MF, Way D, Taub DD,
132 Mangiardi JR, Daras M, Geller ME, Weitzner I, Tuchman Chang SL, Kim KS, House SD, Weinand M, Witte M,
AJ. Cocaine-related intracranial hemorrhage. Report of Graves MC, Fiala M. Cocaine enhances brain endothelial
nine cases and review. Acta Neurol Scand 1988; 77: adhesion molecules and leukocyte migration. Clin
177–80 Immunol 1999; 91: 68–76
133 Qureshi AI, Akbar MS, Czander E, Safdar K, Janssen RS, 149 Zhang L, Looney D, Taub D, Chang SL, Way D, Witte
Frankel MR. Crack cocaine use and stroke in young MH, Graves MC, Fiala M. Cocaine opens the blood-brain
patients. Neurology 1997; 48: 341–5 barrier to HIV-1 invasion. J Neurovirol 1998; 4: 619–26
134 Rajab R, Stearns E, Baithun S. Autopsy pathology of 150 Hurd YL, Herkenham M. Molecular alterations in the
cocaine users from the Eastern district of London: a neostriatum of human cocaine addicts. Synapse 1993;
retrospective cohort study. J Clin Pathol 2008; 61: 13: 357–69
848–50 151 Little KY, Krolewski DM, Zhang L, Cassin BJ. Loss of
135 Treadwell SD, Robinson TG. Cocaine use and stroke. striatal vesicular monoamine transporter protein
Postgrad Med J 2007; 83: 389–94 (VMAT2) in human cocaine users. Am J Psychiatry
136 Herning RI, King DE, Better WE, Cadet JL. Neurovascu- 2003; 160: 47–55
lar deficits in cocaine abusers. Neuropsychopharmacology 152 Little KY, McLaughlin DP, Zhang L, McFinton PR,
1999; 21: 110–18 Dalack GW, Cook EH Jr, Cassin BJ, Watson SJ. Brain
137 Kaufman MJ, Levin JM, Ross MH, Lange N, Rose SL, dopamine transporter messenger RNA and binding sites
Kukes TJ, Mendelson JH, Lukas SE, Cohen BM, Renshaw in cocaine users: a postmortem study. Arch Gen Psychia-
PF. Cocaine-induced cerebral vasoconstriction detected try 1998; 55: 793–9
in humans with magnetic resonance angiography. 153 Little KY, Patel UN, Clark TB, Butts JD. Alterations of
JAMA 1998; 279: 376–80 brain dopamine and serotonin levels in cocaine users:
138 Schreiber MD, Madden JA, Covert RF, Torgerson LJ. a preliminary report. Am J Psychiatry 1996; 153:
Effects of cocaine, benzoylecgonine, and cocaine 1216–18
metabolites on cannulated pressurized fetal sheep cere- 154 Little KY, Kirkman JA, Carroll FI, Clark TB, Duncan GE.
bral arteries. J Appl Physiol 1994; 77: 834–9 Cocaine use increases [3H]WIN 35428 binding sites in
139 Jennings LK, White MM, Sauer CM, Mauer AM, Robert- human striatum. Brain Res 1993; 628: 17–25
son JT. Cocaine-induced platelets defects. Stroke 1993; 155 Wilson JM, Levey AI, Bergeron C, Kalasinsky K, Ang L,
24: 1352–9 Peretti F, Adams VI, Smialek J, Anderson WR, Shannak
140 Kugelmass AD, Oda A, Monahan K, Cabral C, Ware JA. K, Deck J, Niznik HB, Kish SJ. Striatal dopamine, dopam-
Activation of human platelets by cocaine. Circulation ine transporter, and vesicular monoamine transporter
1993; 88: 876–83 in chronic cocaine users. Ann Neurol 1996; 40: 428–
141 Nolte KB, Brass LM, Fletterick CF. Intracranial hemor- 39
rhage associated with cocaine abuse: a prospective 156 Meador-Woodruff JH, Little KY, Damask SP, Mansour A,
autopsy study. Neurology 1996; 46: 1291–6. Watson SJ. Effects of cocaine on dopamine receptor gene

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
132 A. Büttner

expression: a study in the postmortem human brain. 171 Yen DJ, Wang SJ, Ju TH, Chen CC, Liao KK, Fuh JL, Hu
Biol Psychiatry 1993; 34: 348–55 HH. Stroke associated with methamphetamine inhala-
157 Mash DC, Pablo J, Ouyang Q, Hearn WL, Itzenwasser S. tion. Eur J Neurol 1994; 34: 16–22
Dopamine transport function is elevated in cocaine 172 Zhu BL, Oritani S, Shimotouge K, Ishida K, Quan L,
users. J Neurochem 2002; 81: 292–300 Fujita MQ, Ogawa M, Maeda H. Methamphetamine-
158 Segal DM, Moraes CT, Mash DC. Up-regulation of D3 related fatalities in forensic autopsy during 5 years in
dopamine receptor mRNA in the nucleus accumbens of southern half of Osaka city and surrounding areas.
human cocaine fatalities. Mol Brain Res 1997; 45: Forensic Sci Int 2000; 113: 443–7
335–9 173 Lee YW, Hennig B, Yao J, Toborek M. Methamphet-
159 Staley JK, Hearn WL, Ruttenber AJ, Wetli CV, Mash DC. amine induces AP-1 and NF-kappaB binding and trans-
High affinity cocaine recognition sites on dopamine activation in human brain endothelial cells. J Neurosci
transporter are elevated in fatal cocaine overdose Res 2001; 66: 583–91
victims. J Pharmacol Exp Ther 1994; 271: 1678–85 174 Brown JM, Hanson GR, Fleckenstein AE. Methamphet-
160 Bannon MJ, Pruetz B, Manning-Bog AB, Whitty CJ, amine rapidly decreases vesicular dopamine uptake.
Michelhaugh SK, Sacchetti P, Granneman JG, Mash DC, J Neurochem 2000; 74: 2221–3
Schmidt CJ. Decreased expression of the transcription 175 Davidson C, Gow AJ, Lee TH, Ellinwood EH. Metham-
factor NURR1 in dopamine neurons of cocaine abusers. phetamine neurotoxicity: necrotic and apoptotic
Proc Natl Acad Sci USA 2002; 99: 6382–5 mechanisms and relevance to human abuse and treat-
161 Mash DC, Ouyang Q, Pablo J, Basile M, Itzenwasser S, ment. Brain Res Brain Res Rev 2001; 36: 1–22
Lieberman A, Perrin RJ. Cocaine users have an overex- 176 Frost DO, Cadet JL. Effects of methamphetamine-
pression of a-synuclein in dopamine neurons. J Neurosci induced neurotoxicity on the development of neural cir-
2003; 23: 2564–72 cuits: a hypothesis. Brain Res Brain Res Rev 2000; 34:
162 Staley JK, Rothman RB, Rice KC, Partilla J, Mash DC. k2 103–18
opioid receptors in limbic areas of the human brain are 177 Harvey DC, Lacan G, Tanious SP, Melega WP. Recovery
upregulated by cocaine in fatal overdose victims. J Neu- from methamphetamine induced long-term nigrostri-
rosci 1997; 17: 8225–33 atal dopaminergic deficits without substantia nigra cell
163 Tang W-X, Fasulo WH, Mash DC, Hemby SE. Molecular loss. Brain Res 2000; 871: 259–70
profiling of midbrain dopamine regions in cocaine over- 178 Seiden LS, Sabol KE. Methamphetamine and methylene-
dose victims. J Neurochem 2003; 85: 911–24 dioxymethamphetamine neurotoxicity: possible mecha-
164 Mash DC, Staley JK, Itzenwasser S, Basile M, Ruttenber nisms of cell destruction. NIDA Res Monogr 1996; 163:
AJ. Serotonin transporters upregulate with chronic 251–76
cocaine use. J Chem Neuroanat 2000; 20: 271–80 179 Sekine Y, Ouchi Y, Takei N, Yoshikawa E, Nakamura K,
165 Ross BM, Moszczynska A, Peretti F, Adams V, Schmunk Futatsubashi M, Okada H, Minabe Y, Suzuki K, Iwata Y,
GA, Kalasinsky KS, Ang L, Mamalis N, Turenne SD, Tsuchiya KJ, Tsukada H, Iyo M, Mori N. Brain serotonin
Kish SJ. Decreased activity of brain phospholipid transporter density and aggression in abstinent meth-
metabolic enzymes in human users of cocaine and amphetamine abusers. Arch Gen Psychiatry 2006; 63:
methamphetamine. Drug Alcohol Depend 2002; 67: 90–100
73–9 180 Tong J, Ross BM, Schmunk GA, Peretti FJ, Kalasinsky
166 Kalasinsky KS, Bosy TZ, Schmunk GA, Reiber G, KS, Furukawa Y, Ang L-C, Aiken SS, Wickham DJ, Kish
Anthony RM, Furukawa Y, Guttman M, Kish SJ. SJ. Decreased striatal dopamine D1 receptor-stimulated
Regional distribution of methamphetamine in autop- adenylyl cyclase activity in human methamphetamine
sied brain of chronic methamphetamine users. Forensic users. Am J Psychiatry 2003; 160: 896–903
Sci Int 2001; 116: 163–9 181 Volkow ND, Chang L, Wang G-J, Fowler JS, Leonido-Yee
167 Heye N, Hankey GJ. Amphetamine-associated stroke. M, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R,
Cerebrovasc Dis 1996; 6: 149–55 Ding Y-S. Association of dopamine transporter reduc-
168 Karch SB, Stephens BG, Ho C-H. Methamphetamine- tion with psychomotor impairment in methamphet-
related deaths in San Francisco: demographic, patho- amine abusers. Am J Psychiatry 2001; 158: 377–82
logic, and toxicologic profiles. J Forensic Sci 1999; 44: 182 Guilarte TR. Is methamphetamine abuse a risk factor in
359–68 parkinsonism? Neurotoxicology 2001; 22: 725–31
169 Logan BK, Fligner CL, Haddix T. Cause and manner of 183 Thrash B, Thiruchelvan K, Ahuja M, Suppiramaniam
death in fatalities involving methamphetamine. J Foren- V, Dhanasekaran M. Methamphetamine-induced neu-
sic Sci 1998; 43: 28–34 rotoxicity: the road to Parkinson’s disease. Pharmacol
170 Moriya F, Hashimoto Y. A case of fatal hemorrhage in Rep 2009; 61: 966–77
the cerebral ventricles following intravenous use of 184 Wilson JM, Kalasinsky KS, Levey AI, Bergeron C, Reiber
methamphetamine. Forensic Sci Int 2002; 129: 104– G, Anthony RM, Schmunk GA, Shannak K, Haycock
9 JW, Kish SJ. Striatal dopamine nerve terminal markers

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
The neuropathology of drug abuse 133

in human, chronic methamphetamine users. Nat Med 200 Hanyu S, Ikeguchi K, Imai H, Imai N, Yoshida M.
1996; 2: 699–703 Cerebral infarction associated with 3,4-
185 Cadet JL, Krasnova IN. Molecular bases of methylenedioxymethamphetamine (‘ecstasy’) abuse.
methamphetamine-induced neurodegeneration. Int Rev Eur Neurol 1995; 35: 173
Neurobiol 2009; 88: 101–19 201 Harries DP, De Silva R. ‘Ecstasy’ and intracerebral
186 Christophersen AS. Amphetamine designer drugs – an haemorrhage. Scott Med J 1992; 37: 150–2
overview and epidemiology. Toxicol Lett 2000; 112: 202 Hughes JC, McCabe M, Evans RJ. Intracranial haemor-
127–31 rhage associated with ingestion of ‘ecstasy’. Arch Emerg
187 Parrott AC. Is ecstasy MDMA? A review of the propor- Med 1993; 10: 372–4
tion of ecstasy tablets containing MDMA, their dosage 203 Manchanda S, Connolly MJ. Cerebral infarction in asso-
levels, and the changing perceptions of purity. Psychop- ciation with ecstasy abuse. Postgrad Med J 1993; 69:
harmacology (Berl) 2004; 173: 234–41 874–5
188 De la Torre R, Farre M, Roset PN, Pizarro N, Abanades S, 204 Spatt J, Glawar B, Mamoli B. A pure amnestic syndrome
Segura M, Segura J, Cami J. Human pharmacology of after MDMA (‘ecstasy’) ingestion. J Neurol Neurosurg
MDMA: pharmacokinetics, metabolism and disposition. Psychiatry 1997; 62: 418–19
Ther Drug Monit 2004; 26: 137–447 205 Squier MV, Jalloh S, Hilton-Jones D, Series H. Death after
189 Huether G, Zhou D, Rüther E. Causes and con- ecstasy ingestion: neuropathological findings. J Neurol
sequences of the loss of serotonergic presynapses Neurosurg Psychiatry 1995; 58: 756
elicited by the consumption of 3,4-methylenedioxy 206 Commins DL, Vosmer G, Virus RM, Woolverton WL,
methamphetamine (MDMA, ‘ecstasy’) and its conge- Schuster CR, Seiden LS. Biochemical and histologi-
ners. J Neural Transm 1997; 104: 771–94 cal evidence that methylenedioxymethamphetamine
190 Kalant H. The pharmacology and toxicology of ‘ecstasy’ (MDMA) is toxic to neurons in the rat brain. J Pharmacol
(MDMA) and related drugs. CMAJ 2001; 165: 917–28 Exp Ther 1987; 241: 338–45
191 McKenna DJ, Peroutka SJ. Neurochemistry and 207 Ricaurte GA, McCann UD, Szabo Z, Scheffel U. Toxico-
neurotoxicity of 3,4-methylenedioxymethamphetamine dynamics and long-term toxicity of the recreational
(MDMA, ‘ecstasy’). J Neurochem 1990; 54: 14–22 drug, 3,4-methylenedioxymethamphetamine (MDMA,
192 De Letter EA, Espeel M, Craeymeersch M, Lambert WE, ‘ecstasy’). Toxicol Lett 2000; 112: 143–6
Clauwaert K, Dams R, Mortier KA, Piette M. Immuno- 208 Curran HV. Is MDMA (‘ecstasy’) neurotoxic in humans?
histochemical demonstration of the amphetamine An overview of evidence and of methodological prob-
derivatives 3,4-methylenedioxymethamphetamine lems in research. Neuropsychobiology 2000; 42: 34–
(MDMA) and 3,4-methylenedioxyamphetamine (MDA) 41
in human post-mortem brain tissues and the pituitary 209 Lyles J, Cadet JL. Methylenedioxymethamphetamine
gland. Int J Legal Med 2003; 117: 2–9 (MDMA, ecstasy) neurotoxicity: cellular and molecular
193 Fineschi V, Centini F, Mazzeo E, Turillazzi E. Adam mechanisms. Brain Res Brain Res Rev 2003; 42: 155–68
(MDMA) and Eve (MDEA) misuse: an immunohis- 210 Sprague JE, Everman SL, Nichols DE. An integrated
tochemical study on three fatal cases. Forensic Sci Int hypothesis for the serotonergic axonal loss induced by
1999; 104: 65–74 3,4-methylenedioxymethamphetamine. Neurotoxicol-
194 Gill JR, Hayes JA, deSouza IS, Marker E, Stajic M. Ecstasy ogy 1998; 19: 427–42
(MDMA) deaths in New York City: a case series and 211 Simantov R, Tauber M. The abused drug MDMA
review of the literature. J Forensic Sci 2002; 47: 121– (ecstasy) induces programmed cell death of human
6 serotonergic cells. FASEB J 1997; 11: 141–6
195 Henry JA, Jeffreys KJ, Dawling S. Toxicity and 212 Zhou JF, Chen P, Zhou YH, Zhang L, Chen HH. 3,4-
deaths from 3,4-methylenedioxymethamphetamine Methylenedioxymethamphetamine (MDMA) abuse may
(‘ecstasy’). Lancet 1992; 340: 384–7 cause oxidative stress and potential free radical damage.
196 Milroy CM, Clark JC, Forrest ARW. Pathology of deaths Free Radic Res 2003; 37: 491–7
associated with ‘ecstasy’ and ‘eve’ misuse. J Clin Pathol 213 Gouzoulis-Mayfrank E, Daumann J. Neurotoxicity of
1996; 49: 149–53 methylenedioxyamphetamines (MDMA; ecstasy) in
197 Bertram M, Egelhoff T, Schwarz S, Schwab S. Toxic leu- humans: how strong is the evidence for persistent brain
koencephalopathy following ‘ecstasy’ ingestion. J Neurol damage? Addiction 2006; 101: 348–61
1999; 246: 617–18 214 Kish SJ. How strong is the evidence that brain serotonin
198 Bitsch A, Thiel A, Rieckmann P, Prange H. Acute neurons are damaged in human users of ecstasy? Phar-
inflammatory CNS disease after MDMA (‘ecstasy’). Eur macol Biochem Behav 2002; 71: 845–55
Neurol 1996; 36: 328–9 215 McGuire P. Long term psychiatric and cognitive effects
199 Gledhill JA, Moore DF, Bell D, Henry JA. Subarachnoid of MDMA use. Toxicol Lett 2000; 112–113: 153–6
haemorrhage associated with MDMA abuse. J Neurol 216 Parrott AC, Marsden CA. MDMA (3,4-
Neurosurg Psychiatry 1993; 56: 1036–7 methylenedioxymethamphetamine) or ecstasy: the

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134
134 A. Büttner

contemporary human and animal research perspective. young drug abusers. Neuropathol Appl Neurobiol 2005;
J Psychopharmacol 2006; 20: 143–6 31: 439–48
217 Parrott AC. Human psychopharmacology of ecstasy 229 Büttner A, Rohrmoser K, Mall G, Penning R, Weis S.
(MDMA): a review of 15 years of empirical research. Widespread axonal damage in the brain of drug abusers
Hum Psychopharmacol Clin Exp 2001; 16: 557–77 as evidenced by accumulation of b-amyloid precursor
218 Reneman L, Booij J, Majoie CBL, van den Brink W, den protein (b-APP): an immunohistochemical investiga-
Heeten GJ. Investigating the potential neurotoxicity of tion. Addiction 2006; 101: 1339–46
ecstasy (MDMA): an imaging approach. Hum Psychop- 230 Anthony IC, Ramage SN, Carnie FW, Simmonds P, Bell
harmacol Clin Exp 2001; 16: 579–88 JE. Does drug abuse alter microglial phenotype and cell
219 Verbaten MN. Specific memory deficits in ecstasy users? turnover in the context of advancing HIV infection?
The results of a meta-analysis. Hum Psychopharmacol Neuropathol Appl Neurobiol 2005; 31: 325–38.
Clin Exp 2003; 18: 281–90 231 Tomlinson GS, Simmonds P, Busuttil A, Chiswick A, Bell
220 Makrigeorgi-Butera M, Hagel C, Laas R, Püschel K, JE. Upregulation of microglia in drug users with and
Stavrou D. Comparative brain pathology of HIV- without pre-symptomatic HIV infection. Neuropathol
seronegative and HIV-infected drug addicts. Clin Neuro- Appl Neurobiol 1999; 25: 369–79
pathol 1996; 15: 324–9 232 Miguel-Hidalgo JJ. The role of glial cells in drug abuse.
221 Boronat MA, García-Fuster MJ, García-Sevilla JA. Curr Drug Abuse Rev 2009; 2: 76–82
Chronic morphine induces up-regulation of the pro- 233 Sorensen RG, Lawrence DMP. Glial cells and drugs of
apoptotic Fas receptor and down-regulation of the anti- abuse in the nervous system. Drug Alcohol Depend 2009;
apoptotic Bcl-2 oncoprotein in rat brain. Br J Pharmacol 102: 166–9
2001; 134: 1263–70 234 Anderson CE, Tomlinson GS, Pauly B, Brannan FW,
222 Hu S, Sheng WS, Lokensgard JR, Peterson PK. Morphine Chiswick A, Brack-Werner R, Simmonds P, Bell JE. Rela-
induces apoptosis of human microglia and neurons. tionship of Nef-positive and GFAP-reactive astrocytes to
Neuropharmacology 2002; 42: 829–36 drug use in early and late HIV infection. Neuropathol
223 Jiang Y, Yang W, Zhou Y, Ma L. Up-regulation of Appl Neurobiol 2003; 29: 378–88
murine double minute clone 2 (MDM2) gene expression 235 Stadlin A, Lau JWS, Szeto YK. A selective regional
in rat brain after morphine, heroin, and cocaine admin- response of cultured astrocytes to methamphetamine.
istrations. Neurosci Lett 2003; 352: 216–20 Ann N Y Acad Sci 1998; 844: 108–21
224 Mao J, Sung M, Ji R-R, Lim G. Neuronal apoptosis asso- 236 Castagnoli N Jr, Castagnoli KP. Metabolic bioactivation
ciated with morphine tolerance: evidence for an opioid- reactions potentially related to drug toxicities. NIDA Res
induced neurotoxic mechanism. J Neurosci 2002; 22: Monogr 1997; 173: 85–105
7650–61 237 Sastre M, Ventayol P, García-Sevilla JA. Decreased
225 Stumm G, Schlegel J, Schäfer T, Würz C, Mennel HD, density of I2-imidazoline receptors in the postmortem
Krieg J-C, Vedder H. Amphetamines induce apoptosis brains of heroin addicts. Neuroreport 1996; 7: 509–12
and regulation of bcl-x splice variants in neocortical 238 Connor MD, Lammie GA, Bell JE, Warlow CP, Simmonds
neurons. FASEB J 1999; 13: 1065–72 P, Brettle RP. Cerebral infarction in adult AIDS patients:
226 Boronat MA, Olmos G, García-Sevilla JA. Attenuation observations from the Edinburgh HIV autopsy cohort.
of tolerance to opioid-induced antinociception and Stroke 2000; 31: 2117–26
protection against morphine-induced decrease of 239 Büttner A, Kroehling C, Mall G, Penning R, Weis S.
neurofilament proteins by idazoxan and other Alterations of the vascular basal lamina in the cerebral
I2-imidazoline ligands. Br J Pharmacol 1998; 125: cortex in drug abuse: a combined morphometric and
175–85 immunohistochemical investigation. Drug Alcohol
227 Ferrer-Alcón M, García-Sevilla JA, Jaquet PE, La Harpe Depend 2005; 69: 63–79
R, Riederer BM, Walzer C, Guimón J. Regulation of non- 240 Geibprasert S, Gallucci M, Krings T. Addictive illegal
phosphorylated and phosphorylated forms of neurofila- drugs: structural neuroimaging. AJNR Am J Neuroradiol
ment proteins in the prefrontal cortex of human opioid 2010; 31: 803–8
addicts. J Neurosci Res 2000; 61: 338–49
228 Ramage SN, Anthony IC, Carnie FW, Busuttil A, Rob- Received 26 July 2010
ertson R, Bell JE. Hyperphosphorylated tau and amyloid Accepted after revision 30 September 2010
precursor protein deposition is increased in the brains of Published online Article Accepted on 6 October 2010

© 2011 The Author


Neuropathology and Applied Neurobiology © 2011 British Neuropathological Society, 37, 118–134

Anda mungkin juga menyukai