[Review] the Neuropathology of Drug Abuse_2011

Embed Size (px)

Citation preview

Neuropathology and Applied Neurobiology (2011), 37, 118134

doi: 10.1111/j.1365-2990.2010.01131.x

Review: The neuropathology of drug abuseA. Bttner Institute of Forensic Medicine, University of Rostock, Rostock, Germany

_

118..134

A. Bttner (2011) Neuropathology and Applied Neurobiology 37, 118134 The neuropathology of drug abuseDrug abuse represents a signicant health issue. The major substances abused include cannabis, opiates, cocaine, amphetamine, methamphetamine and ecstasy. Alterations of intracellular messenger pathways, transcription factors and immediate early genes within the brain reward system seem to be fundamentally important for the development of addiction and chronic drug abuse. Genetic risk factors and changes in gene expression associated with drug abuse are still poorly understood. Besides cardiovascular complications, psychiatric and neurologic symptoms are the most common manifestations of drug toxicity. A broad spectrum of changes affecting the central nervous system is seen in drug abusers. The major ndings result from the consequences of ischaemia and cerebrovascular diseases. Except for a few observations of vasculitis, the aetiology of these cerebrovascular accidents is not fully understood. The abuse of amphetamine, methamphetamine and MDMA has been related to neurotoxicity in human long-term abusers and to the risk of developing Parkinsons disease. However, whether such neurotoxicity occurs remain to be established. Systematic histological, immunohistochemical and morphometric investigations have shown profound morphological alterations in the brains of polydrug abusers. The major ndings comprise neuronal loss, neurodegenerative alterations, a reduction of glial brillary acidic proteinimmunopositive astrocytes, widespread axonal damage with concomitant microglial activation as well as reactive and degenerative changes of the cerebral microvasculature. These observations demonstrate that drugs of abuse initiate a cascade of interacting toxic, vascular and hypoxic factors, which nally result in widespread disturbances within the complex network of central nervous system cell-to-cell interactions.

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

IntroductionDrug abuse represents a serious health issue worldwide. Drugs of abuse can be grouped as stimulants, analgesics and narcotics, hypnotics as well as antidepressants, nicotine and alcohol [13]. Besides the latter two substances, the predominant substances abused include cannabis, opiates, cocaine, amphetamine, methamphetamine and designer drugs [1,2]. Although individuals with a drug habit may favour one or other class of drug, depending in

Correspondence: Andreas Bttner, Institute of Forensic Medicine, University of Rostock, St.-Georg-Strasse 108, 18055 Rostock, Germany. Tel: +49 381 494 9900; Fax: +49 381 494 9902; E-mail: [email protected]

part on what is available, most perform polysubstance abuse [48]. The methodological problems in investigating the effects of drugs of abuse on the central nervous system (CNS) consist of distinguishing between substancespecic effects related to the properties of the drug itself, the inuence of adulterants, and of secondary effects related to the lifestyle of drug abusers, for example, malnutrition, infections and systemic diseases. Because polysubstance abuse is seen in the majority of cases, it is difcult to relate the observed ndings to a single substance. Therefore, in most cases the exact aetiology of the various CNS alterations remains unclear. Injecting drug abuse is also a risk factor for the 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 abusers [9,10]. Infection of the brain with hepatitis viruses, especially with hepatitis C virus (HCV) is another potential confounding factor in the neuropathological appearances of the observed brain changes. Injection drug abusers are the largest group of persons infected with HCV, with a prevalence of 5090% [11]. The transmission of HCV is not the effect of the drug injected but of sharing contaminated equipment [11]. In contrast to HIV, HCV is detectable in high concentrations in other utensils for heroin injection, such as lters, spoons and rinsing liquids. Besides hepatic encephalopathy, there is also emerging evidence of neurocognitive impairment in HCV infection independently of drug abuse or HIV-1 infection [1215]. Research on the neurobiology of addiction has shown that the reinforcing properties of most drugs of abuse are mediated by activation of the mesolimbic dopaminergic system, the orbitofrontal cortex and the extended amygdala [1619]. Alterations of the intracellular messenger pathways, transcription factors and immediate early gene expression in these reward circuits are believed to be important for the development of addiction and chronic drug abuse [1620]. Possible genetic risk factors for drug addiction and changes in gene expression, associated with drug abuse, are still poorly understood [21,22]. Although a broad spectrum of alterations affecting the CNS has been described in drug abusers [1,2,23], there is no specic change that is characteristic and little is known about the fundamental neuropathological alterations of the cellular elements of the human brain. Therefore, despite a large body of literature on animal models, the following review will focus on the relevant human CNS ndings. The pharmacology and CNS alterations in alcohol and nicotine abuse are reviewed elsewhere [3,2427].

tions and hyperintense lesions have been demonstrated in the white matter, which were attributed to ischaemic lesion [34,3840]. Single photon emission computed tomography showed focal perfusions decits in various brain regions [4145]. On positron emission tomography a reduction of the global glucose metabolism has been demonstrated [46,47]. Studies using proton magnetic resonance spectroscopy have identied several biochemical changes in the brain that may underlie the neuropathology that subsequently gives rise to the cognitive and behavioural impairments associated with drug addiction [48]. Some neurochemical abnormalities have been attributed to alterations in nonneuronal brain tissue [48]. Diffusion tensor imaging revealed microstructural abnormalities in the white matter and the corpus callosum, suggestive of axonal injury [4954]. Despite these numerous neuroimaging studies, the cause or possible morphological correlates of these alterations are still not fully resolved.

CNS effects of the major drugs of abuseDepending of the frequency and types of drugs abused, the predominant neurologic-psychiatric complications of drug abuse include a high prevalence of depression, memory loss and cognitive decline, and the possible predisposition to schizophrenia.

CannabisCannabis is the most commonly used illicit drug worldwide [2,5559]. Cannabis preparations (marihuana, hashish) are usually mixed with tobacco and smoked. Oral ingestion as tea or addition to pastries is also widespread. Cannabis is unsuitable for intravenous use as it is relatively waterinsoluble. The concentration of the major psychoactive component of cannabis, D9-tetrahydrocannabinol (THC), varies according to the preparation and the origin of the plant from 1% to 7% in marihuana, 2% to 10% in hashish and 10% to 60% in hash oil [55,57]. Cannabis combines many of the properties of alcohol, tranquillizers, opiates and hallucinogens; it is anxiolytic, sedative, analgesic and psychedelic [55]. The psychotropic action of cannabis starts after several minutes reaches a maximum by 2030 min and persists for 24 h. However,

Neuroimaging ndingsNeuroimaging studies revealed widespread alterations in the brains of drug abusers [28]: Computed tomography studies have shown diffuse brain atrophy [2931]. On magnetic resonance imaging decreased grey and white matter volume has been demonstrated in particular areas of the brain [3237]. Moreover, focal demyelina-

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

120

A. Bttner

the action of cannabis is of highly individual variability. Furthermore, there is a dose-related impairment of cognitive and psychomotor performance. Acute effects include euphoria and relaxation with perceptual changes. However, dysphoric reactions, including anxiety and panic, depression, paranoia and psychosis can also occur. Systemic effects include a dose-related tachycardia, vasodilation and postural hypotension [5559]. THC is a lipid-soluble substance that is distributed heterogeneously within the brain, with its highest concentrations in neocortical, limbic, sensory and motor areas [5558]. THC and other cannabinoids exert their effects by the interaction with specic cannabinoid receptors, which are distributed heterogeneously within the brain [5964]. Two cannabinoid receptors, CB1 and CB2, have been pharmacologically characterized and anatomically localized. CB1 receptors are found predominantly in the central and peripheral nervous system, where they have been implicated in presynaptic inhibition of neurotransmitter release. CB2 receptors are present on immune cells, where they may be involved in cytokine release [5965]. The highest density of CB1 receptors is found in the substantia nigra, the basal ganglia, the hippocampus and the cerebellum [63,66,67]. Within the neocortex they are present with the highest density in the frontal cortex, the dentate gyrus, the mesolimbic dopaminergic system and the temporal lobe [63,66,67]. This specic distribution of CB1 receptors correlates well with the effects of cannabinoids on memory, perception and movement control. The very low density of CB1 receptors in the brain stem and medulla oblongata explains the low acute toxicity and lack of lethality of cannabis [55,59,66]. Nevertheless, the CNS toxicity of cannabis seems to have been underestimated for a long time [68], as recent ndings revealed THC-induced neuronal death in cultured rat neurones [6971]. The discovery of specic endogenous cannabinoid receptor ligands (endocannabinoids), and the distribution of their receptors, strongly suggests that these lipid neurotransmitter systems play an important role in higher cortical-emotional functions, memory storage, movement coordination and several pathological conditions [59,6265,72,73]. Despite its widespread abuse, cannabis-related cerebrovascular events are infrequently reported. They are believed to be due to a cannabis-induced vasospasm or a

cannabis-induced hypotension [7478]. However, due to the widespread use of this drug, it is difcult and often impossible to establish whether these strokes are truly associated with cannabis, other ingested drugs, or purely coincidental. The important question whether cannabis can cause irreversible brain damage, particularly to adolescents, or lead to increased risk for schizophrenia, persisting beyond transient intoxication, is still a matter of debate [79,80].

Opioids and derivativesOpioids, especially heroin, are the leading substances that cause death in drug abusers. Derivatives include morphine, hydrocodone, oxycodone, hydromorphine, codeine and other narcotics such as fentanyl, meperidene, methadone and opium [2,3]. Heroin is made from opium and crosses the bloodbrain barrier (BBB) faster than morphine [81]. Heroin is usually administered intravenously. Intranasal and subcutaneous administration is also possible. Heroin alkaloid may be prepared for inhalation by heating on metallic foil (chasing the dragon) that results in a higher bioavailability of heroin compared with smoking in a cigarette [3]. Intravenous heroin induces an extreme euphoria lasting for several minutes. Subsequently, there is a pleasant dream-like state with drowsiness and analgesia [81]. However, marked tolerance develops to euphoria. Opiate overdose produces the triad of coma, respiratory depression and miosis [81]. Between 50% and 70% of intravenous heroin abusers have experienced a non-fatal overdose at some time in their lives [4]. Medical complications of long-term heroin abuse include thrombophlebitis, endocarditis, hepatitis, pneumonia, nephropathy and immunodepression [2,81]. Due to the practise of needlesharing, there is a high risk for infections [11]. Risk factors for opioid-induced deaths include overdose, concomitant use of other CNS depressants and loss of tolerance after a period of abstinence [48]. At necropsy, up to 90% of all cases of persons dying of heroin overdose show brain oedema with prominent tonsillar herniation and uncal grooving. However, rapid death after heroin intake will not lead to any morphological evidence of neuronal cell injury. In cases of delayed death after a survival period of 5 h or longer, hypoxicischaemic leukoencephalopathy with loss of neurones in

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

121

the hippocampal formation, the Purkinje cell layer and/or the olivary nucleus, as well as vascular congestion, will become apparent [8286]. In the globus pallidus, neuronal loss and bilateral, symmetrical ischaemic lesions/ necrosis have been described in 510% of cases [85,87]. All the above mentioned alterations are assumed to be caused rather by recurrent episodes of hypoxia during the intoxicated state than to be related to direct neurotoxic effects of the opioid drugs [8287]. Stroke in heroin abusers, occurring in the absence of endocarditis or mycotic aneurysms, has rarely been observed [84,8893]. Within the hippocampus of heroin deaths, enhanced expression of glial brillary acidic protein (GFAP) by astrocytes and/or a proliferation of microglia have been found [84]. However, an increased GFAP expression could not be conrmed by other authors [23]. An increase in polysialic acid neural cell adhesion molecule positive hippocampal neurones and astrocytes is assumed to reect an attempt to repair cell damage [94]. Perivascular pigment-laden macrophages and pigment depositions are sometimes observed and are attributed to repeated intravenous injections of impure heroin [95], or to BBB breakdown [23,96]. Infections in heroin abusers result from unsterile injection techniques and from immunosuppression, caused by chronic opiate abuse [1,2,11,85,97]. Endocarditis might lead to septic foci in the brain or to intracranial mycotic aneurysms [1,2,85,97]. Transverse myelitis/myelopathy is an exceptionally rare lesion described in heroin abusers [1,2,85,98,99]. The aetiology is still unclear and neither the clinical picture nor the pathological changes correspond to any particular pattern. A distinct entity, spongiform leukoencephalopathy, has been reported to occur almost exclusively after inhalation of heroin pyrolysate vapours (chasing the dragon) [1,2,100102]. The most characteristic nding is the selective involvement of the posterior limb of the internal capsule and sparing of subcortical U-bres. Neuropathological examination reveals spongiform degeneration of the deep white matter with vacuolation of the myelin sheath, loss of oligodendrocytes, axonal reduction and astrogliosis. The grey matter is usually unremarkable and the brainstem, spinal cord and peripheral nerves are spared [100102]. A lipophilic toxin-induced process was considered to be due to contaminants, but a denite toxin could not yet be identied.

Alterations of neurotransmitters and receptorsLong-term heroin abuse seems not to be related with a reduced density of CNS m- and d-opioid receptors [103 106]. Instead, the molecular mechanisms underlying opiate addiction seem to involve the second messenger signalling system [16,18,19,104,107114]. Necropsy studies revealed that long-term heroin abuse causes an increase in certain G proteins in different regions of the brain [105,107,110]. Others have shown a decreased level of Ca2+-dependent protein kinase C-a [104] and an increased level of a membrane-associated G proteincoupled receptor kinase [113]. Further ndings include the downregulation of the adenylyl-cyclase subtype I [109,111], a decrease in the density of alpha 2-adrenoreceptors [103], a decrease in the immunoreactivity of protein kinase C-ab [114], and decreased levels of neurolament proteins [115]. Within the dopaminergic system the levels of tyrosine hydroxylase protein and those of the dopamine (DA) metabolite homovanillic acid were reduced in the nucleus accumbens [116]. Striatal levels of serotonin (5-hydroxytryptamine or 5-HT) were either normal or elevated, whereas the concentration of the 5-HT metabolite 5-hydroxyindoleacetic acid was decreased [116]. According to the authors, chronic heroin abuse might produce a modest reduction in dopaminergic and serotonergic activity that could affect motivational state and impulse control. Maintenance therapy for heroin addiction includes codeine, dihydrocodeine, methadone and buprenorphine [117120]. In the majority of deaths related to these substances, additional CNS depressant drugs, mainly alcohol and benzodiazepines, can be detected. The neuropathological ndings are similar to those encountered in heroin deaths.

CocaineCocaine is derived from the leaves of the coca plant. Cocaine hydrochloride is a water-soluble white salt and the most frequent preparation of the drug that is available in form of cystals, granules or a white powder. It can be administered intranasally (snorting) or injected [2,3]. The free alkaloid form (free base) that had been extracted with volatile solvents is usually smoked. Crack cocaine is produced by rst dissolving the hydrochloride salt in water, then mixing with baking soda and heating. The

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

122

A. Bttner

cracking sound made by the crystals when they are heated provides the name [2]. Cocaine initially induces profound subjective well-being together with alertness and increased self-condence. Subsequently, there is a mild euphoria followed by fatigue. Acute psychiatric disturbances include dysphoria, agitation, aggressive behaviour, depression, paranoia, psychosis and hallucinations. Medical complications include vasoconstriction, tachycardia, hypertension and sudden cardiac death [3,25,31]. Cocaine is a potent CNS stimulant. It crosses the BBB rapidly due to its highly lipophilic properties [121,122]. Throughout the brain, cocaine and its major metabolites are widely distributed and receptors with varying afnities are found [123,124]. Cocaine acts by binding to specic receptors at presynaptic sites preventing the reuptake of neurotransmitters [2,3,122]. The major synaptic effect of cocaine is the release of dopamine (DA) from the synaptic vesicles and the blocking of DA reuptake resulting in an enhanced dopaminergic neurotransmission [2,3,122]. Cocaine is the most frequent drug of abuse associated with fatal and non-fatal cerebrovascular events, with either haemorrhagic or ischaemic strokes [89,91 93,125135]. In contrast to the non-drug-using population, cocaine-related stroke occurs primarily in young adults [1,125128]. Cocaine-related ischaemic infarctions can occur in every brain region [2,128,130]. The underlying cause is attributed to cerebral vasospasm either by cocaine or its metabolites [92,128,131,136138]. Other mechanisms include cocaine-induced cardiac arrhythmia with secondary cerebral ischaemia, or the direct effects of cocaine on haemostasis with increased platelet aggregation [89,128,131,135,139,140]. In cocaine-related intracerebral (ICH) and subarachnoidal (SAH) haemorrhage, underlying arteriovenous malformations or aneurysms are frequently observed [93,125,127130,132,141,142]. Cocaine abuse has been shown to predispose to aneurysmal rupture at a signicantly earlier age, and in much smaller aneurysms, compared with non-drug-using persons [125,143]. Cocaine-related ICH is associated more frequently with subcortical locations, a higher risk of intraventricular haemorrhage, and poor prognosis compared with nondrug-abusing patients with spontaneous ICH [144]. Rupture of an arteriovenous malformation, or aneurysm, are most likely related to the sudden elevation of blood

pressure and heart rate from the sympathomimetic effect of the drug [92,128130,132,141,145]. A cocaine-induced cerebral vasculitis as a cause for cerebrovascular events could only be demonstrated in rare cases [89,146,147]. In experimental models, cocaine enhances leucocyte migration across the cerebral vessel wall and opens the BBB to HIV-1 invasion by a direct effect on brain endothelial cells and by the induction of pro-inammatory cytokines and chemokines [148,149].

Alterations of neurotransmitters and receptorsIn the striatum of cocaine-related deaths, reductions in the levels of enkephalin mRNA, m-opioid receptor binding and DA uptake site binding, along with elevation in levels of dynorphin mRNA and k-opioid receptor binding have been described [150]. In chronic cocaine abusers, a decreased level of DA was identied in the caudate nucleus and frontal cortex, but not in the putamen, nucleus accumbens and cerebral cortex [151155]. This decrease was not paralleled by an increase of DA D1 and D2 gene expression [156]. Simultaneously, there was an increase of cocaine binding sites on the DA transporter with a decrease of the DA D1 receptor density in the striatum and of DA D1 and DA D3 receptor density in the nucleus accumbens [152154,157159]. A marked reduction in immunoreactivity of the vesicular monoamine transporter-2 [151] and of the transcription factor NURR1 [160] in necropsy samples of human cocaine abusers might reect damage to the dopaminergic system. An overexpression of a-synuclein in midbrain DA neurones in chronic cocaine abusers may occur as a protective response to changes in DA turnover and increased oxidative stress resulting from cocaine abuse [161]. This accumulation of a-synuclein protein in long-term cocaine abuse might increase the risk of developing Parkinsonism [161]. Further necropsy ndings include an upregulation of k2-opioid receptors in the limbic system [162] and of cAMP response element-binding protein in the ventral tegmental area [163]. In the serotonergic system an increase of the 5-HT transporter in the striatum, substantia nigra and limbic system has been demonstrated [164]. In the putamen, a DA-rich brain area, the activity of phospholipase A2 and phosphocholine cytidylyltransferase was selectively decreased [165].

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

123

Amphetamines and methamphetamineAmphetamines and methamphetamine are widely used psychostimulants that act on monoamine transporters [2,3]. These substances are available as powder, capsules, tablets or uids. Therefore, they can be swallowed, snorted, injected intravenously or smoked. The composition, purity and dose are highly variable [2]. Their potent sympathomimetic effects include an elevation of pulse rate, blood pressure and an increased level of alertness. Adverse effects include insomnia, excitability, seizures, panic attacks, psychosis and aggressive behaviour [2,3,25]. Throughout the brain, methamphetamine is heterogeneously distributed [166]. Amphetamines and methamphetamine are the second most common cause (after cocaine) of strokes occurring largely in persons younger than 45 years [127]. Furthermore, SAH and ICH have been described [2,3,91,92,125,126,129,167172]. Similar to cocaine, a sudden elevation in blood pressure [167,169,171] is postulated as a major mechanism. The vasoconstrictive effect of both substances may also lead to the development of ischaemic infarction [167,171]. Methamphetamine has been shown to induce inammatory genes in human brain endothelial cells [173].

dopaminergic system markers [181,184], the evidence is inconclusive in regard to dopaminergic system degeneration. To dene methamphetamine abuse as a risk factor in Parkinson disease, it would be important to know whether these alterations represent neurodegenerative changes or a drug-induced compensatory response to the disruption of neurochemical homeostasis [182,183]. The mechanisms of methamphetamine-induced neurotoxicity are thought to be mediated by multiple mechanisms including the generation of free radicals and nitric oxide, excitotoxicity, mitochondrial dysfunction, and the induction of immediate early genes as well as transcription factors [175177,182185]. However, whether neurotoxicity occurs in human methamphetamine and amphetamine abusers remains to be established.

Amphetamine and methamphetamine derivativesAmphetamine and methamphetamine derivatives (designer drugs) comprise a broad spectrum of substances [2,186]. The street name ecstasy subsumes different hallucinogenic amphetamine derivatives with MDMA (3,4-methylenedioxymethamphetamine) and 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 [2]. The effects of MDMA and MDEA last about 35 h and include relaxation, euphoria, sensual enhancement, reduction of anxiety and emotional closeness to others. This psychological prole has been called entactogenic and has the connotation of inducing a feeling of touch with the world within. In addition, these substances also have stimulant-like and hallucinogenic effects [2,188 191]. The effects of MDMA vary according to the dose and the frequency and duration of use [190]. Adverse effects include headache, nausea, tooth grinding (bruxism), tachycardia and an increase in body temperature. Furthermore, hyperactivity, ight of ideas, depersonalization, panic attacks, anxiety, depression, agitation, delirium, and insomnia may occur [190]. MDMA interacts with various neurotransmitter circuits but predominantly affects the dopaminergic and serotonergic system [186,188191]. In human post mortem tissue, a distinct immunopositive reaction for MDMA and MDA was observed in the white matter in all cortical regions and in neurones of the basal ganglia, the hypo-

Alterations of neurotransmitters and receptorsThe neurotoxic effects of amphetamines and methamphetamine on the dopaminergic system have been described in various animal species and in humans. These effects are characterized by desensitization of DA receptor function, marked reduction of DA transporters and of DA levels as well as other dopaminergic axonal markers [174 181]. Similar alterations have been observed in the serotonergic system [175]. However, the irreversibility of these changes has not been established [175]. Although these alterations have been attributed to neurodegeneration, direct evidence for the loss of nerve terminals and/or their corresponding substantia nigra cell bodies have not been provided unequivocally [175,177]. Nevertheless, based on animal studies, there is concern that the alterations in the dopaminergic system may predispose methamphetamine abusers to develop Parkinsonism as they age [175,182,183]. Because neuroimaging and necropsy studies of human methamphetamine abusers suggest changes in only some

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

124

A. Bttner

thalamus, the hippocampus and the cerebellar vermis, but relatively weak staining of neurones in the brainstem was seen [192]. In reports on human fatalities following MDMA consumption, detailed neuropathological examinations were mostly not performed [193196]. Cerebrovascular complications after ecstasy consumption have been described only occasionally [197203]. In the globus pallidus bilateral hyperintense lesions have been found [204,205]. On neuropathological examination necrosis of the globus pallidus, diffuse astrogliosis and spongiform changes of the white matter have been described [205]. Other neuropathological ndings in deaths after ecstasy abuse were mainly due to the complications of hyperthermia with disseminated intravascular coagulopathy and consisted of cerebral oedema, focal haemorrhages and nerve cell loss [196].

ations have been attributed to drug-induced respiratory failure and were therefore considered to be non-specic [84,220]. However, in most of these studies, there was no control group and systematic data on frequency or topography of the lesions are lacking. Subsequent systematic neuropathological studies of polydrug abusers revealed ischaemia-independent widespread neuronal loss, a reduction of GFAP-positive astrocytes, an axonal damage with concomitant microglia activation as well as reactive and degenerative vascular changes [23].

NeuronesThe neuronal loss has been ascribed to be due to a direct impairment of neurones by drugs of abuse and, indirectly, to drug-induced damage of astrocytes, axons and the cerebral vasculature [23]. In support of this nding is the observation in animal models that drugs of abuse can induce apoptotic neuronal cell death [69,175,221225]. In human studies drug-induced alterations of neurolament proteins [115,226,227], of neuronal TUNEL positivity [96] or of transcription factors, for example, cAMP response element-binding protein [112] and c-fos [20] is thought to be an alternative pathway for neuronal loss. In young opiate abusers an increase in the number and distribution of hyperphosphorylated tau-positive neurobrillary pretangles, fully formed tangles and neuritic threads has been reported [228]. This, together with reports of occasional ubiquitin-positive inclusions in neurones of drug abusers [96], indicates drug-related neurodegeneration. Whether these changes are potentially reversible is still unclear. Within the substantia nigra there was a decrease of the numerical density of the pigmented neurones, whereas the density of the non-pigmented neurones was unchanged [23]. The presence of eosinophilic ubiquitinated cytoplasmic inclusions, which did not resemble classic Lewy bodies, and which are negative for a-synuclein, indicates abnormal cytoplasmic protein sequestration [96]. To date it is not clear whether this nding represents a transient neuronal abnormality or commitment to an irreversible neurodegenerative pathway [96].

Alterations of neurotransmitters and receptorsIn the brains of laboratory animals, including nonhuman primates, exposure to MDMA has been associated with dopaminergic and especially serotonergic neurotoxicity [176,188191,206209]. There is strong evidence for neurodegeneration and axonal loss, although the exact mechanism is still unclear [189191,206209]. Current hypotheses include the formation of toxic MDMA metabolites with generation of free radicals as well as oxidative stress, excitotoxicity, apoptosis and mitochondrial dysfunction [176,209212]. Although in recent years, the question of ecstasyinduced neurotoxicity and possible functional sequelae has been addressed in several studies, the extent to which these animal and non-human primate data are applicable to humans and whether persistent neurotoxicity occurs in humans is still controversial [42,207209,213219]. To date, the most consistent ndings associate subtle cognitive, particularly memory, impairments with heavy ecstasy abuse [213,218,219].

Neuropathological studiesThe consequences of drugs of abuse on the cellular elements of the human brain have not been studied systematically. There are only few reports on histopathological alterations in the brains of human drug abusers describing oedema, vascular congestion, ischaemic nerve cell damage and neuronal loss [8385,96,220]. These alter-

White matterIn the white matter of polydrug abusers a widespread axonal damage has been demonstrated by using b-APP

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

125

Figure 1. Immunohistochemistry demonstrating b-APP-immunopositive bundles and b-APP-immunopositive globular deposits in the pons of polydrug abusers, counterstained with haematoxylin, original magnication 100.

negative drug abusers and non-drug using controls there was no statistical difference between these groups in relation 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 induction 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 the density of I2-imidazoline receptors and the immunoreactivity of the related imidazoline receptor protein were decreased [237].

immunohistochemistry [23,229]. The alterations consisted of b-APP-immunopositive bundles and globular depositions (Figure 1), but they never showed the extensive pattern seen in traumatic brain injury [23,229]. Furthermore, there is a concomitant activation of microglia predominantly in the white matter and in most subcortical regions [23,83,220,229]. This upregulation of microglia has also been shown in drug abusers with and without pre-symptomatic HIV-1 infection [230,231]. Activated microglial cells are a source of proinammatory cytokines, which are linked to neuronal damage and loss [10]. The axonal injury suggests a toxicmetabolic drug effect, as there were no sufcient ndings for a secondary phenomenon in these cases, neither due to a generalized hypoxic-ischaemic condition, nor to a brain oedema and the concomitant activation of microglia is indicative of a long-standing progressive process [23,229]. These alterations might be the morphological correlate of the observed demyelination and hyperintense areas seen on magnetic resonance imaging.

Vascular changesIn AIDS patients concentric small-vessel wall thickening, perivascular space dilatation, pigment deposition, vessel wall mineralization and perivascular inammatory cell inltrates were seen in 50% of the former drug abusers [238]. Similar vascular alterations can be observed in HIV-1-negative polydrug abusers (Figure 2A) [23,96]. The presence of acute and chronic BBB breakdown in drug abusers, suggests that the brain parenchyma is exposed to unusual quantities of serum proteins, including immunoglobulins, as well as other blood-borne factors, including HIV [96]. It may further be the stimulus for the occasional perivascular lymphocytic aggregates that are seen in the brains of HIV-1-negative drug abusers (Figure 2B) [10]. Furthermore, reactive endothelial cell proliferation, degenerative hyalinotic thickening, marked endothelial swelling and endothelial cell hyperplasia are present in the brains of polydrug abusers (Figure 2C,D) [23]. In addition, a decrease of the collagen type IV content of the vascular basal lamina [239] and a disruption of the BBB tight junctions [10] have been observed. This non-inammatory vasculopathy has been interpreted as the morphological substrate of a disturbed BBB and might be the morphological substrate of the alterations seen on neuroimaging [23,240]. In conclusion, drugs of abuse initiate a cascade of interacting toxic, vascular and hypoxic factors that nally result in widespread disturbances within the complex network of CNS cell interactions (Figure 3). However, much work will need to be done to clarify the yet

AstrocytesAlthough astrocytes play an essential role, for example, in the maintenance of BBB, regulation of glutamatergic neurotransmission and neurotransmitter metabolism, little is known about alterations of these cells in human drug abusers [232,233]. In an older study widespread fragmentation and a numerical depletion of astrocytes in the white matter have been reported in the brains of drug deaths [85]. In HIV-1-positive opiate abusers, HIV-1-

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

126

A. Bttner

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 magnication 200. (B) Small vessel in the orbital white matter with perivascular lymphocytic aggregates. H&E, original magnication 200. (C) Endothelial proliferation in the dentate nucleus, H&E, original magnication 200. (D) Endothelial hyperplasia in the parietal white matter, H&E, original magnication 200.

unresolved questions of the disastrous role of drugs of abuse on the CNS.

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

ReferencesFigure 3. The consequences of drugs of abuse on the cellular elements of the CNS.

1 Bttner A, Weis S. Central nervous system alterations in drug abuse. In Forensic Pathology Reviews, Vol. 1. Ed. M Tsokos. Totowa, NJ: Humana Press, 2004; 79136

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

127

2 Karch SB. Karchs Pathology of Drug Abuse 4th edn. Boca Raton: CRC Press, 2008 3 Quinn DI, Wodak A, Day RO. Pharmacokinetic and pharmacodynamic principles of illicit drug use and treatment of illicit drug users. Clin Pharmacokinet 1997; 33: 344400 4 Cofn PO, Galea S, Ahern J, Leon AC, Vlahov D, Tardiff K. Opiates, cocaine and alcohol combinations in accidental drug overdose deaths in New York City, 19901998. Addiction 2003; 98: 73947 5 Darke S. Polydrug use and overdose: overthrowing old myths. Addiction 2003; 98: 711 6 Gerostamoulos J, Staikos V, Drummer OH. Heroinrelated deaths in Victoria: a review of cases for 1997 and 1998. Drug Alcohol Depend 2001; 61: 1237 7 Preti A, Miotto P, De Coppi M. Deaths by unintentional illicit drug overdose in Italy, 1984-2000. Drug Alcohol Depend 2002; 66: 27582 8 Steentoft A, Teige B, Holmgren P, Vuori E, Kristinsson J, Hansen AC, Ceder G, Wethe G, Rollmann D. Fatal poisoning in Nordic drug addicts in 2002. Forensic Sci Int 2006; 160: 14856 9 Anthony IC, Arango JC, Stephens B, Simmonds P, Bell JE. The effects of illicit drugs on the HIV infected brain. Front Biosci 2008; 13: 1294307 10 Bell JE, Arango JC, Anthony IC. Neurobiology of multiple insults: HIV-1-associated brain disorders in those who use illicit drugs. J NeuroImmune Pharmacol 2006; 1: 18291 11 Backmund M, Reimer J, Meyer K, Gerlach JT, Zachoval R. Hepatitis C virus infection and injection drug users: prevention, risk factors, and treatment. Clin Infect Dis 2005; 40 (Suppl. 5): S3305 12 Cherner M, Letendre SL, Heaton RK, Durelle J, MarquieBeck J, Gragg B, Grant I. Hepatitis C augments cognitive decits associated with HIV infection and methamphetamine. Neurology 2005; 64: 13437 13 Forton DM, Taylor-Robinson SD, Thomas HC. Central nervous system changes in hepatitis C virus infection. Eur J Gastroenterol Hepatol 2006; 18: 3338 14 Martin-Thormeyer EM, Paul RH. Drug abuse and hepatitis C infection as comorbid features of HIV associated neurocognitive disorder: neurocognitive and neuroimaging features. Neuropsychol Rev 2009; 19: 215 21 15 Morgello S. The nervous system and hepatitis C virus. Semin Liver Dis 2005; 25: 11821 16 Cunha-Oliveira T, Rego AC, Oliveira CR. Cellular and molecular mechanisms involved in the neurotoxicity of opioid and psychostimulant drugs. Brain Res Brain Res Rev 2008; 58: 192208 17 Hyman SE, Malenka RC, Nestler EJ. Neural mechanisms of addiction: the role of reward-related learning and memory. Annu Rev Neurosci 2006; 29: 56598 18 Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology 2010; 35: 21738

19 Nestler EJ. Molecular mechanisms of drug addiction. Neuropharmacology 2004; 47: 2432 20 Harlan RE, Garcia MM. Drugs of abuse and immediateearly genes in the forebrain. Mol Neurobiol 1998; 16: 22167 21 Kuhar MJ, Joyce A, Dominguez G. Genes in drug abuse. Drug Alcohol Depend 2001; 62: 15762 22 Nestler EJ, Landsman D. Learning about addiction from the genome. Nature 2001; 409: 8345 23 Bttner A, Weis S. Neuropathological alterations in drug abusers: the involvement of neurons, glial, and vascular systems. Forensic Sci Med Pathol 2006; 2: 11526 24 Bttner A, Weis S. Central nervous system alterations in alcohol abuse. In Forensic Pathology Reviews, Vol. 5. Ed. M Tsokos. Totowa, NJ: Humana Press, 2008; 6989. 25 Trelles L, Jeri R. Central nervous system stimulants: cocaine, amphetamine, nicotine. In Handbook of Clinical Neurology, Vol. 65: Intoxications of the Nervous System, Part II. Eds PJ Vinkens, GW Bruyn, FA de Wolff. Amsterdam: Elsevier, 1995; 25172. 26 Harper C. The neuropathology of alcohol-related brain damage. Alcohol Alcohol 2009; 44: 13640 27 Yildiz D. Nicotine, its metabolism and an overview of its biological effects. Toxicon 2004; 43: 61932 28 Kaufman MJ. Brain Imaging in Substance Abuse: Research, Clinical, and Forensic Applications. Totowa, NJ: Humana Press, 2001 29 Pascual-Leone A, Dhuna A, Anderson DC. Cerebral atrophy in habitual cocaine users. A planimetric CT study. Neurology 1991; 41: 348 30 Pezawas LM, Fischer G, Diamant K, Schneider C, Schindler SD, Thurnher M, Ploechl W, Eder H, Kasper S. Cerebral CT ndings in male opioid-dependent patients: stereological, planimetric and linear measurements. Psychiatry Res Neuroimag 1998; 83: 13947 31 Strang J, Gurling H. Computerized tomography and neuropsychological assessment in long-term high-dose heroin addicts. Br J Addict 1989; 84: 101119 32 Bartzokis G, Beckson M, Lu PH, Edwards N, Rapoport R, Wiseman E, Bridge P. Age-related brain volume reductions in amphetamine and cocaine addicts and normal controls: implications for addiction research. Psychiatry Res 2000; 98: 93102 33 Berman S, ONeill J, Fears S, Bartzokis G, London ED. Abuse of amphetamines and structural abnormalities in the brain. Ann N Y Acad Sci 2008; 1141: 195220 34 Lim KO, Wozniak JR, Mueller BA, Franc DT, Specker SM, Rodriguez CP, Silverman AB, Rotrosen JP. Brain macrostructural and microstructural abnormalities in cocaine dependence. Drug Alcohol Depend 2008; 92: 16472 35 Lyoo IK, Pollack MH, Silveri MM, Ahn KH, Diaz CI, Hwang J, Kim SJ, Yurgelun-Todd D, Kaufman MJ, Renshaw PF. Prefrontal and temporal gray matter density decreases in opiate dependence. Psychopharmacology (Berl) 2006; 184: 13944

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

128

A. Bttner

36 Schlaepfer TE, Lancaster E, Heidbreder R, Strain EC, Kosel M, Fisch H-U, Pearlson GD. Decreased frontal white-matter volume in chronic substance abuse. Int J Neuropsychopharmacol 2007; 9: 14753 37 Ycel M, Solowij N, Respondek C, Whittle S, Fornito A, Pantelis C, Lubman DI. Regional brain abnormalities associated with long-term heavy cannabis use. Arch Gen Psychiatry 2008; 65: 694701 38 Bae SC, Lyoo IK, Sung YH, Yoo J, Chung A, Yoon S-J, Kim D-J, Hwang J, Kim SJ, Renshaw PF. Increased white matter hyperintensities in male methamphetamine abusers. Drug Alcohol Depend 2006; 81: 838 39 Bartzokis G, Goldstein IB, Hance DB, Beckson M, Shapiro D, Lu PH, Edwards N, Mintz J, Bridge P. The incidence of T2-weighted MR imaging signal abnormalities in the brain of cocaine-dependent patients is age-related and region-specic. AJNR Am J Neuroradiol 1999; 20: 162835 40 Lyoo IK, Streeter CC, Ahn KH, Lee HK, Pollack MH, Silveri MM, Nassar LE, Levin JM, Sarid-Segal O, Ciraulo DA, Renshaw PF, Kaufman MJ. White matter hyperintensities in subjects with cocaine and opiate dependence and healthy comparison subjects. Psychiatry Res Neuroimag 2004; 131: 13545 41 Botelho MF, Relvas JS, Abrantes M, Cunha MJ, Marques TR, Rovira E, Fontes Ribeiro CA, Macedo T. Brain blood ow SPET imaging in heroin abusers. Ann N Y Acad Sci 2006; 1074: 46677 42 Chang L, Grob CS, Ernst T, Itti L, Mishkin FS, JoseMelchor R, Poland RE. Effect of ecstasy [3,4methylenedioxymethamphetamine (MDMA)] on cerebral blood ow: a co-registered SPECT and MRI study. Psychiatry Res 2000; 98: 1528 43 Ernst T, Chang L, Oropilla G, Gustavson A, Speck O. Cerebral perfusion abnormalities in abstinent cocaine abusers: a perfusion MRI and SPECT study. Psychiatry Res 2000; 99: 6374 44 Gottschalk PC, Kosten TR. Cerebral perfusion defects in combined cocaine and alcohol dependence. Drug Alcohol Depend 2002; 68: 95104 45 OLeary DS, Block RI, Koeppel JA, Flaum M, Schultz SK, Andreasen NC, Boles Ponto LL, Watkins GL, Hurtig RR, Hichwa RD. Effects of smoking marijuana on brain perfusion and cognition. Neuropsychopharmacology 2002; 26: 80216 46 London ED, Cascella NG, Wong DF, Phillips RL, Dannals RF, Links JM, Herning R, Grayson R, Jaffe JH, Wagner HN Jr. Cocaine-induced reduction of glucose utilization in human brain. Arch Gen Psychiatry 1990; 47: 567 74 47 Volkow ND, Fowler JS, Wolf AP, Hitzemann R, Dewey S, Bendriem B, Alpert R, Hoff A. Changes in brain glucose metabolism in cocaine dependence and withdrawal. Am J Psychiatry 1991; 148: 6216 48 Licata SC, Renshaw PF. Neurochemistry of drug action: insights from proton magnetic resonance spectroscopic

49

50

51

52

53

54

55 56 57 58

59 60

61 62

63

imaging and their relevance to addiction. Ann N Y Acad Sci 2010; 1187: 14871 Alicata D, Chang L, Cloak C, Abe K, Ernst T. Higher diffusion in striatum and lower fractional anisotropy in white matter of methamphetamine users. Psychiatry Res Neuroimag 2009; 174: 18 Arnone D, Barrick TR, Chengappa S, Mackay CE, Clark CA, Abou-Saleh MT. Corpus callosum damage in heavy marijuana use: preliminary evidence from diffusion tensor tractography and tract-based spatial statistics. Neuroimage 2008; 41: 106774 Liu H, Li L, Hao Y, Cao D, Xu L, Rohrbaugh R, Xue Z, Hao W, Shan B, Liu Z. Disrupted white matter integrity in heroin dependence: a controlled study utilizing diffusion tensor imaging. Am J Drug Alcohol Abuse 2008; 34: 56274 Ma L, Hasan KM, Steinberg JL, Narayana PA, Lane SD, Zuniga EA, Kramer LA, Moeller FG. Diffusion tensor imaging in cocaine dependence: regional effects of cocaine on corpus callosum and effect of cocaine administration route. Drug Alcohol Depend 2009; 104: 2627 Romero MJ, Asensio S, Palau C, Sanchez A, Romero FJ. Cocaine addiction: diffusion tensor imaging study of the inferior frontal and anterior cingulate white matter. Psychiatry Res Neuroimag 2010; 181: 5763 Tobias MC, ONeill J, Hudkins M, Bartzokis G, Dean AC, London ED. White-matter abnormalities in brain during early abstinence from methamphetamine abuse. Psychopharmacology (Berl) 2010; 209: 1324 Ashton CH. Pharmacology and effects of cannabis: a brief review. Br J Psychiatry 2001; 178: 1016 Iversen L. Cannabis and the brain. Brain 2003; 126: 125279 Johns A. Psychiatric effects of cannabis. Br J Psychiatry 2001; 178: 11622 Nahas GG. The pharmacokinetics of THC in fat and brain: resulting functional responses to marihuana smoking. Hum Psychopharmacol Clin Exp 2001; 16: 24755 Ameri A. The effects of cannabinoids on the brain. Prog Neurobiol 1999; 58: 31548 Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham M, Mackie K, Martin BR, Mechoulam R, Pertwee RG. Classication of cannabinoid receptors. Pharmacol Rev 2002; 54: 161202 Pertwee RG. Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther 1997; 74: 12980 Fride E. Endocannabinoids in the central nervous system - an overview. Prostaglandins Leukot Essent Fatty Acids 2002; 66: 22133 Glass M, Dragunow M, Faull RLM. Cannabinoid receptors in the human brain: a detailed anatomical and quantitative autoradiographic study in the fetal neonatal and adult human brain. Neuroscience 1997; 77: 299318

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

129

64 Childers SR, Breivogel CS. Cannabis and endogenous cannabinoid systems. Drug Alcohol Depend 1998; 51: 17387 65 Wilson RI, Nicoll RA. Endocannabinoid signaling in the brain. Science 2002; 296: 67882 66 Herkenham M. Cannabinoid receptor localization in brain: relationship to motor and reward systems. Ann N Y Acad Sci 1992; 654: 1932 67 Mailleux P, Parmentier M, Vanderhaeghen J-J. Distribution of cannabinoid receptor messenger RNA in the human brain: an in situ hybridization histochemistry with oligonucleotides. Neurosci Lett 1992; 143: 2004 68 Scallet AC. Neurotoxicology of cannabis and THC: a review of chronic exposure studies in animals. Pharmacol Biochem Behav 1991; 40: 6716 69 Campbell VA. Tetrahydrocannabinol-induced apoptosis of cultured cortical neurones is associated with cytochrome c release and caspase-3 activation. Neuropharmacology 2001; 40: 7029 70 Chan GCK, Hinds TR, Impey S, Storm DR. Hippocampal neurotoxicity of delta9-tetrahydrocannabinol. J Neurosci 1998; 18: 532232 71 Guzmn M, Snchez C, Galve-Roperh I. Control of the cell survival/death decision by cannabinoids. J Mol Med 2001; 78: 61325 72 Di Marzo V. Regulation of endocannabinoid levels under physiological and pathological conditions. A mini-review. Pharm Pharmacol Commun 2000; 6: 23541 73 Tanda G, Goldberg SR. Cannabinoids: reward, dependence, and underlying neurochemical mechanisms - a review of recent preclinical data. Psychopharmacology (Berl) 2003; 169: 11534 74 Barnes D, Palace J, OBrien MD. Stroke following marijuana smoking. Stroke 1992; 23: 1381 75 Mateo I, Pinedo A, Gomez-Beldarrain M, Basterretxea JM, Garcia-Monco JC. Recurrent stroke associated with cannabis use. J Neurol Neurosurg Psychiatry 2005; 76: 4357 76 Mouzak A, Agathos P, Kerezoudi E, Mantas A, VourdeliYiannakoura E. Transient ischemic attack in heavy cannabis smokers - how safe is it? Eur Neurol 2000; 44: 424 77 Thanvi BR, Treadwell SD. Cannabis and stroke: is there a link? Postgrad Med J 2009; 85: 803 78 Zachariah SB. Stroke after heavy marijuana smoking. Stroke 1991; 22: 4069 79 DeLisi LE. The effect of cannabis on the brain: can it cause brain anomalies that lead to increased risk for schizophrenia? Curr Opin Psychiatry 2008; 21: 14050 80 Moore THM, Zammit S, Lingford-Hughes A, Barnes TRE, Jones PB, Burke M, Lewis G. Cannabis use and risk of psychotic or affective mental health outcomes: a systematic review. Lancet 2007; 370: 31928 81 Brust JCM. Opiate addiction and toxicity. In Handbook of Clinical Neurology, Vol. 65: Intoxications of the Nervous

82

83

84

85

86 87

88

89

90

91

92

93

94

95

96

System, Part II. Eds PJ Vinkens, GW Bruyn, FA de Wolff. Amsterdam: Elsevier, 1995; 34961 Ginsberg MD, Hedley-Whyte ET, Richardson EP Jr. Hypoxic-ischemic leukoencephalopathy in man. Arch Neurol 1976; 33: 514 Gosztonyi G, Schmidt V, Nickel R, Rothschild MA, Camacho S, Siegel G, Zill E, Pauli G, Schneider V. Neuropathologic analysis of postmortal brain samples of HIV-seropositive and -seronegative i.v. drug addicts. Forensic Sci Int 1993; 62: 1015 Oehmichen M, Meiner C, Reiter A, Birkholz M. Neuropathology in non-human immunodeciency virus-infected drug addicts: hypoxic brain damage after chronic intravenous drug abuse. Acta Neuropathol 1996; 91: 6426 Pearson J, Richter RW. Addiction to opiates: neurologic aspects. In Handbook of Clinical Neurology. Intoxications of the Nervous System, Part II. Eds PJ Vinken, GW Bruyn. Amsterdam: North-Holland Publishing Company, 1979; 365400 Protass LM. Delayed postanoxic encephalopathy after heroin use. Ann Intern Med 1971; 74: 7389 Andersen SN, Skullerud K. Hypoxic/ischaemic brain damage, especially pallidal lesions, in heroin addicts. Forensic Sci Int 1999; 102: 519 Adle-Biassette H, Marc B, Benhaiem-Sigaux N, Durigon M, Gray F. Infarctus crbraux chez un toxicomane inhalant lhroine. Arch Anat Cytol Pathol 1996; 44: 1217 Brust JCM. Clinical, radiological, and pathological aspects of cerebrovascular disease associated with drug abuse. Stroke 1993; 24 (Suppl. I): I-129-33 Brust JCM, Richter RW. Stroke associated with addiction to heroin. J Neurol Neurosurg Psychiatry 1976; 39: 1949 Caplan LR, Hier DB, Banks G. Current concepts of cerebrovascular disease - stroke: stroke and drug abuse. Stroke 1982; 13: 86972 Kelly MA, Gorelick PB, Mirza D. The role of drugs in the etiology of stroke. Clin Neuropharmacol 1992; 15: 24975 Sloan MA, Kittner SJ, Rigamonti D, Price TR. Occurrence of stroke associated with use/abuse of drugs. Neurology 1991; 41: 135864 Weber M, Modemann S, Schipper P, Trauer H, Franke H, Illes P, Geiger KD, Hengstler JG, Kleemann WJ. Increased polysialic acid neural cell adhesion molecule expression in human hippocampus of heroin addicts. Neuroscience 2006; 138: 121523 Gray F, Lescs MC, Keohane C, Paraire F, Marc B, Durigon M, Gherardi R. Early brain changes in HIV infection: neuropathological study of 11 seropositive, non-AIDS cases. J Neuropathol Exp Neurol 1992; 51: 177 85 Bell JE, Arango JC, Robertson R, Brettle RP, Leen C, Simmonds P. HIV and drug misuse in the Edinburgh

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

130

A. Bttner

97

98

99

100

101

102

103

104

105

106

107

108

109

110

cohort. J Acquir Immune Dec Syndr 2002; 31 (Suppl. 2): S3542 Adelman LS, Aronson SM. The neuropathologic complications of narcotic drug addiction. Bull N Y Acad Med 1969; 45: 22534 McCreary M, Emerman C, Hanna J, Simon J. Acute myelopathy following intranasal insufation of heroin: a case report. Neurology 2000; 55: 31617 Pearson J, Richter RW, Baden MM, Challenor YB, Bruun B. Transverse myelopathy as an illustration of the neurologic and neuropathologic features of heroin addiction. Hum Pathol 1972; 3: 10713 Kriegstein AR, Armitage BA, Kim PY. Heroin inhalation and progressive spongiform leukoencephalopathy. N Engl J Med 1997; 336: 58990 Rizzuto N, Morbin M, Ferrari S, Cavallaro T, Sparaco M, Boso G, Gaetti L. Delayed spongiform leukoencephalopathy after heroin abuse. Acta Neuropathol 1997; 94: 8790 Wolters EC, Stam FC, Lousberg RJ, van Wijngaarden GK, Rengelink H, Schipper MEI, Verbeeten B. Leucoencephalopathy after inhalating heroin pyrolysate. Lancet 1982; 320: 12337 Gabilondo AM, Meana JJ, Barturen F, Sastre M, GarcaSevilla JA. m-opioid receptor and a2-adrenoreceptor agonist binding sites in the postmortem brain of heroin addicts. Psychopharmacology (Berl) 1994; 115: 13540 Garca-Sevilla JA, Ventayol P, Busquets X, La Harpe R, Walzer C, Guimn J. Regulation of immunolabelled mu-opioid receptors and protein kinase C-alpha and zeta isoforms in the frontal cortex of human opiate addicts. Neurosci Lett 1997; 226: 2932 Meana JJ, Gonzlez-Maeso J, Garca-Sevilla JA, Guimn J. m-Opioid receptor and a2-adrenoreceptor agonist stimulation of [35S]GTPgS binding to G-proteins in postmortem brains of opioid addicts. Mol Psychiatry 2000; 5: 30815 Schmidt P, Schmolke C, Musshoff F, Menzen M, Prohaska C, Madea B. Area-specic increased density of m-opioid receptor immunoreactive neurons in the cerebral cortex of drug-related fatalities. Forensic Sci Int 2003; 133: 20411 Hashimoto E, Frlich L, Ozawa H, Saito T, Shichinohe S, Takahata N, Riederer P. Alteration of guanosine triphosphate binding proteins in postmortem brains of heroin addicts. Alcohol Clin Exp Res 1996; 20: 301A4A Law PY, Wong YH, Loh HH. Molecular mechanisms and regulation of opioid receptor signaling. Annu Rev Pharmacol Toxicol 2000; 40: 389430 Shichinohe S, Ozawa H, Hashimoto E, Tatschner T, Riederer P, Saito T. Changes in the cAMP-related signal transduction mechanism in postmortem human brains of heroin addicts. J Neural Transm 2001; 108: 33547 Escrib PV, Sastre M, Garca-Sevilla JA. Increased density of guanine nucleotide-binding proteins in the

111

112

113

114

115

116

117

118

119 120 121 122 123

124

125

postmortem brains of heroin addicts. Arch Gen Psychiatry 1994; 51: 494501 Shichinohe S, Ozawa H, Saito T, Hashimoto E, Lang C, Riederer P, Takahata N. Differential alteration of adenyl cyclase subtypes I, II, and V/VI in postmortem human brains of heroin addicts. Alcohol Clin Exp Res 1998; 22: 84S7S Lane-Ladd SB, Pineda J, Boundy VA, Pfeuffer T, Krupinski J, Aghajanian GK, Nestler EJ. CREB (cAMP response element-binding protein) in the locus coeruleus: biochemical, physiological, and behavioral evidence for a role in opiate dependence. J Neurosci 1997; 17: 7890 901 Ozaita A, Escrib PV, Ventayol P, Murga C, Mayor F Jr, Garca-Sevilla JA. Regulation of g protein-coupled receptor kinase 2 in brains of opiate-treated rats and human opiate addicts. J Neurochem 1998; 70: 124957 Busquets X, Escrib PV, Sastre M, Garca-Sevilla JA. Loss of protein kinase C-ab in brain of heroin addicts and morphine-dependent rats. J Neurochem 1995; 64: 24752 Garca-Sevilla JA, Ventayol P, Busquets X, La Harpe R, Walzer C, Guimn J. Marked decrease of immunolabelled 68 kDa neurolament (NF-L) proteins in brains of opiate addicts. Neuroreport 1997; 8: 156170 Kish SJ, Kalasinsky KS, Derkach P, Schmunk GA, Guttman M, Ang L, Adams V, Furukawa Y, Haycock JW. Striatal dopaminergic and serotonergic markers in human heroin users. Neuropsychopharmacology 2001; 24: 5617 Gerostamoulos J, Burke MP, Drummer OH. Involvement of codeine in drug-related deaths. Am J Forensic Med Pathol 1996; 17: 32735 Karch SB, Stephens BG. Toxicology and pathology of deaths related to methadone: retrospective review. West J Med 2000; 172: 1114 Kintz P. A new series of 13 buprenorphine-related deaths. Clin Biochem 2002; 35: 51316 Milroy CM, Forrest ARW. Methadone deaths: a toxicological analysis. J Clin Pathol 2000; 53: 27781 Prakash A, Das G. Cocaine and the nervous system. Int J Clin Pharmacol Ther Toxicol 1993; 31: 57581 White SM, Lambe CJT. The pathophysiology of cocaine abuse. J Clin Forensic Med 2003; 10: 2739 Biegon A, Dillon K, Volkow ND, Hitzemann RJ, Fowler JS, Wolf AP. Quantitative autoradiography of cocaine binding sites in human brain postmortem. Synapse 1992; 10: 12630 Kalasinsky KS, Bosy TZ, Schmunk GA, Ang L, Adams V, Gore SB, Smialek J, Furukawa Y, Guttman M, Kish SJ. Regional distribution of cocaine in postmortem brain of chronic human cocaine users. J Forensic Sci 2000; 45: 10418 McEvoy AW, Kitchen ND, Thomas DGT. Intracerebral haemorrhage and drug abuse in young adults. Br J Neurosurg 2000; 14: 44954

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

131

126 Petitti DB, Sidney S, Quesenberry C, Bernstein A. Stroke and cocaine or amphetamine use. Epidemiology 1998; 9: 596600 127 Kaku DA, Lowenstein DH. Emergence of recreational drug abuse as a major risk factor for stroke in young adults. Ann Intern Med 1990; 113: 8217 128 Levine SR, Brust JCM, Futrell N, Brass LM, Blake D, Fayad P, Schultz LR, Millikan CH, Ho K-L, Welch KMA. A comparative study of the cerebrovascular complications of cocaine: alkaloidal versus hydrochloride - a review. Neurology 1991; 41: 11737. 129 Davis GD, Swalwell CI. The incidence of acute cocaine or methamphetamine intoxication in deaths due to ruptured cerebral (berry) aneurysms. J Forensic Sci 1996; 41: 6268 130 Fessler RD, Esshaki CM, Stankewitz RC, Johnson RR, Diaz FG. The neurovascular complications of cocaine. Surg Neurol 1997; 47: 33945 131 Konzen JP, Levine SR, Garcia JH. Vasospasm and thrombus formation as possible mechanism of stroke related to alkaloidal cocaine. Stroke 1995; 26: 1114 18 132 Mangiardi JR, Daras M, Geller ME, Weitzner I, Tuchman AJ. Cocaine-related intracranial hemorrhage. Report of nine cases and review. Acta Neurol Scand 1988; 77: 17780 133 Qureshi AI, Akbar MS, Czander E, Safdar K, Janssen RS, Frankel MR. Crack cocaine use and stroke in young patients. Neurology 1997; 48: 3415 134 Rajab R, Stearns E, Baithun S. Autopsy pathology of cocaine users from the Eastern district of London: a retrospective cohort study. J Clin Pathol 2008; 61: 84850 135 Treadwell SD, Robinson TG. Cocaine use and stroke. Postgrad Med J 2007; 83: 38994 136 Herning RI, King DE, Better WE, Cadet JL. Neurovascular decits in cocaine abusers. Neuropsychopharmacology 1999; 21: 11018 137 Kaufman MJ, Levin JM, Ross MH, Lange N, Rose SL, Kukes TJ, Mendelson JH, Lukas SE, Cohen BM, Renshaw PF. Cocaine-induced cerebral vasoconstriction detected in humans with magnetic resonance angiography. JAMA 1998; 279: 37680 138 Schreiber MD, Madden JA, Covert RF, Torgerson LJ. Effects of cocaine, benzoylecgonine, and cocaine metabolites on cannulated pressurized fetal sheep cerebral arteries. J Appl Physiol 1994; 77: 8349 139 Jennings LK, White MM, Sauer CM, Mauer AM, Robertson JT. Cocaine-induced platelets defects. Stroke 1993; 24: 13529 140 Kugelmass AD, Oda A, Monahan K, Cabral C, Ware JA. Activation of human platelets by cocaine. Circulation 1993; 88: 87683 141 Nolte KB, Brass LM, Fletterick CF. Intracranial hemorrhage associated with cocaine abuse: a prospective autopsy study. Neurology 1996; 46: 12916.

142 Peterson PL, Roszler M, Jacob I, Wilner HI. Neurovascular complications of cocaine abuse. J Neuropsychiatry Clin Neurosci 1991; 3: 1439 143 Vannemreddy P, Caldito G, Willis B, Nanda A. Inuence of cocaine on ruptured intracranial aneurysms: a case control study of poor prognostic indicators. J Neurosurg 2008; 108: 4706 144 Martin-Schild S, Albright KC, Hallevi H, Barreto AD, Philip M, Misra V, Grotta JC, Savitz SI. Intracerebral hemorrhage in cocaine users. Stroke 2010; 41: 6804 145 Oyesiku NM, Colohan ART, Barrow DL, Reisner A (1993) Cocaine-induced aneurysmal rupture: an emergent negative factor in the natural history of intracranial aneurysms? Neurosurgery 1993; 32: 51826 146 Merkel PA, Koroshetz WJ, Irizarry MC, Cudkowicz ME. Cocaine-associated cerebral vasculitis. Semin Arthritis Rheum 1995; 25: 17283 147 Dez-Tejedor E, Frank A, Gutirrez M, Barreiro P. Encephalopathy and biopsy-proven cerebrovascular inammatory changes in a cocaine abuser. Eur J Neurol 1998; 5: 1037 148 Gan X, Zhang L, Berger O, Stins MF, Way D, Taub DD, Chang SL, Kim KS, House SD, Weinand M, Witte M, Graves MC, Fiala M. Cocaine enhances brain endothelial adhesion molecules and leukocyte migration. Clin Immunol 1999; 91: 6876 149 Zhang L, Looney D, Taub D, Chang SL, Way D, Witte MH, Graves MC, Fiala M. Cocaine opens the blood-brain barrier to HIV-1 invasion. J Neurovirol 1998; 4: 61926 150 Hurd YL, Herkenham M. Molecular alterations in the neostriatum of human cocaine addicts. Synapse 1993; 13: 35769 151 Little KY, Krolewski DM, Zhang L, Cassin BJ. Loss of striatal vesicular monoamine transporter protein (VMAT2) in human cocaine users. Am J Psychiatry 2003; 160: 4755 152 Little KY, McLaughlin DP, Zhang L, McFinton PR, Dalack GW, Cook EH Jr, Cassin BJ, Watson SJ. Brain dopamine transporter messenger RNA and binding sites in cocaine users: a postmortem study. Arch Gen Psychiatry 1998; 55: 7939 153 Little KY, Patel UN, Clark TB, Butts JD. Alterations of brain dopamine and serotonin levels in cocaine users: a preliminary report. Am J Psychiatry 1996; 153: 121618 154 Little KY, Kirkman JA, Carroll FI, Clark TB, Duncan GE. Cocaine use increases [3H]WIN 35428 binding sites in human striatum. Brain Res 1993; 628: 1725 155 Wilson JM, Levey AI, Bergeron C, Kalasinsky K, Ang L, Peretti F, Adams VI, Smialek J, Anderson WR, Shannak K, Deck J, Niznik HB, Kish SJ. Striatal dopamine, dopamine transporter, and vesicular monoamine transporter in chronic cocaine users. Ann Neurol 1996; 40: 428 39 156 Meador-Woodruff JH, Little KY, Damask SP, Mansour A, Watson SJ. Effects of cocaine on dopamine receptor gene

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

132

A. Bttner

157

158

159

160

161

162

163

164

165

166

167 168

169

170

expression: a study in the postmortem human brain. Biol Psychiatry 1993; 34: 34855 Mash DC, Pablo J, Ouyang Q, Hearn WL, Itzenwasser S. Dopamine transport function is elevated in cocaine users. J Neurochem 2002; 81: 292300 Segal DM, Moraes CT, Mash DC. Up-regulation of D3 dopamine receptor mRNA in the nucleus accumbens of human cocaine fatalities. Mol Brain Res 1997; 45: 3359 Staley JK, Hearn WL, Ruttenber AJ, Wetli CV, Mash DC. High afnity cocaine recognition sites on dopamine transporter are elevated in fatal cocaine overdose victims. J Pharmacol Exp Ther 1994; 271: 167885 Bannon MJ, Pruetz B, Manning-Bog AB, Whitty CJ, Michelhaugh SK, Sacchetti P, Granneman JG, Mash DC, Schmidt CJ. Decreased expression of the transcription factor NURR1 in dopamine neurons of cocaine abusers. Proc Natl Acad Sci USA 2002; 99: 63825 Mash DC, Ouyang Q, Pablo J, Basile M, Itzenwasser S, Lieberman A, Perrin RJ. Cocaine users have an overexpression of a-synuclein in dopamine neurons. J Neurosci 2003; 23: 256472 Staley JK, Rothman RB, Rice KC, Partilla J, Mash DC. k2 opioid receptors in limbic areas of the human brain are upregulated by cocaine in fatal overdose victims. J Neurosci 1997; 17: 822533 Tang W-X, Fasulo WH, Mash DC, Hemby SE. Molecular proling of midbrain dopamine regions in cocaine overdose victims. J Neurochem 2003; 85: 91124 Mash DC, Staley JK, Itzenwasser S, Basile M, Ruttenber AJ. Serotonin transporters upregulate with chronic cocaine use. J Chem Neuroanat 2000; 20: 27180 Ross BM, Moszczynska A, Peretti F, Adams V, Schmunk GA, Kalasinsky KS, Ang L, Mamalis N, Turenne SD, Kish SJ. Decreased activity of brain phospholipid metabolic enzymes in human users of cocaine and methamphetamine. Drug Alcohol Depend 2002; 67: 739 Kalasinsky KS, Bosy TZ, Schmunk GA, Reiber G, Anthony RM, Furukawa Y, Guttman M, Kish SJ. Regional distribution of methamphetamine in autopsied brain of chronic methamphetamine users. Forensic Sci Int 2001; 116: 1639 Heye N, Hankey GJ. Amphetamine-associated stroke. Cerebrovasc Dis 1996; 6: 14955 Karch SB, Stephens BG, Ho C-H. Methamphetaminerelated deaths in San Francisco: demographic, pathologic, and toxicologic proles. J Forensic Sci 1999; 44: 35968 Logan BK, Fligner CL, Haddix T. Cause and manner of death in fatalities involving methamphetamine. J Forensic Sci 1998; 43: 2834 Moriya F, Hashimoto Y. A case of fatal hemorrhage in the cerebral ventricles following intravenous use of methamphetamine. Forensic Sci Int 2002; 129: 104 9

171 Yen DJ, Wang SJ, Ju TH, Chen CC, Liao KK, Fuh JL, Hu HH. Stroke associated with methamphetamine inhalation. Eur J Neurol 1994; 34: 1622 172 Zhu BL, Oritani S, Shimotouge K, Ishida K, Quan L, Fujita MQ, Ogawa M, Maeda H. Methamphetaminerelated fatalities in forensic autopsy during 5 years in southern half of Osaka city and surrounding areas. Forensic Sci Int 2000; 113: 4437 173 Lee YW, Hennig B, Yao J, Toborek M. Methamphetamine induces AP-1 and NF-kappaB binding and transactivation in human brain endothelial cells. J Neurosci Res 2001; 66: 58391 174 Brown JM, Hanson GR, Fleckenstein AE. Methamphetamine rapidly decreases vesicular dopamine uptake. J Neurochem 2000; 74: 22213 175 Davidson C, Gow AJ, Lee TH, Ellinwood EH. Methamphetamine neurotoxicity: necrotic and apoptotic mechanisms and relevance to human abuse and treatment. Brain Res Brain Res Rev 2001; 36: 122 176 Frost DO, Cadet JL. Effects of methamphetamineinduced neurotoxicity on the development of neural circuits: a hypothesis. Brain Res Brain Res Rev 2000; 34: 10318 177 Harvey DC, Lacan G, Tanious SP, Melega WP. Recovery from methamphetamine induced long-term nigrostriatal dopaminergic decits without substantia nigra cell loss. Brain Res 2000; 871: 25970 178 Seiden LS, Sabol KE. Methamphetamine and methylenedioxymethamphetamine neurotoxicity: possible mechanisms of cell destruction. NIDA Res Monogr 1996; 163: 25176 179 Sekine Y, Ouchi Y, Takei N, Yoshikawa E, Nakamura K, Futatsubashi M, Okada H, Minabe Y, Suzuki K, Iwata Y, Tsuchiya KJ, Tsukada H, Iyo M, Mori N. Brain serotonin transporter density and aggression in abstinent methamphetamine abusers. Arch Gen Psychiatry 2006; 63: 90100 180 Tong J, Ross BM, Schmunk GA, Peretti FJ, Kalasinsky KS, Furukawa Y, Ang L-C, Aiken SS, Wickham DJ, Kish SJ. Decreased striatal dopamine D1 receptor-stimulated adenylyl cyclase activity in human methamphetamine users. Am J Psychiatry 2003; 160: 896903 181 Volkow ND, Chang L, Wang G-J, Fowler JS, Leonido-Yee M, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R, Ding Y-S. Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. Am J Psychiatry 2001; 158: 37782 182 Guilarte TR. Is methamphetamine abuse a risk factor in parkinsonism? Neurotoxicology 2001; 22: 72531 183 Thrash B, Thiruchelvan K, Ahuja M, Suppiramaniam V, Dhanasekaran M. Methamphetamine-induced neurotoxicity: the road to Parkinsons disease. Pharmacol Rep 2009; 61: 96677 184 Wilson JM, Kalasinsky KS, Levey AI, Bergeron C, Reiber G, Anthony RM, Schmunk GA, Shannak K, Haycock JW, Kish SJ. Striatal dopamine nerve terminal markers

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

The neuropathology of drug abuse

133

185

186

187

188

189

190 191

192

193

194

195

196

197

198

199

in human, chronic methamphetamine users. Nat Med 1996; 2: 699703 Cadet JL, Krasnova IN. Molecular bases of methamphetamine-induced neurodegeneration. Int Rev Neurobiol 2009; 88: 10119 Christophersen AS. Amphetamine designer drugs an overview and epidemiology. Toxicol Lett 2000; 112: 12731 Parrott AC. Is ecstasy MDMA? A review of the proportion of ecstasy tablets containing MDMA, their dosage levels, and the changing perceptions of purity. Psychopharmacology (Berl) 2004; 173: 23441 De la Torre R, Farre M, Roset PN, Pizarro N, Abanades S, Segura M, Segura J, Cami J. Human pharmacology of MDMA: pharmacokinetics, metabolism and disposition. Ther Drug Monit 2004; 26: 137447 Huether G, Zhou D, Rther E. Causes and consequences of the loss of serotonergic presynapses elicited by the consumption of 3,4-methylenedioxy methamphetamine (MDMA, ecstasy) and its congeners. J Neural Transm 1997; 104: 77194 Kalant H. The pharmacology and toxicology of ecstasy (MDMA) and related drugs. CMAJ 2001; 165: 91728 McKenna DJ, Peroutka SJ. Neurochemistry and neurotoxicity of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy). J Neurochem 1990; 54: 1422 De Letter EA, Espeel M, Craeymeersch M, Lambert WE, Clauwaert K, Dams R, Mortier KA, Piette M. Immunohistochemical demonstration of the amphetamine derivatives 3,4-methylenedioxymethamphetamine (MDMA) and 3,4-methylenedioxyamphetamine (MDA) in human post-mortem brain tissues and the pituitary gland. Int J Legal Med 2003; 117: 29 Fineschi V, Centini F, Mazzeo E, Turillazzi E. Adam (MDMA) and Eve (MDEA) misuse: an immunohistochemical study on three fatal cases. Forensic Sci Int 1999; 104: 6574 Gill JR, Hayes JA, deSouza IS, Marker E, Stajic M. Ecstasy (MDMA) deaths in New York City: a case series and review of the literature. J Forensic Sci 2002; 47: 121 6 Henry JA, Jeffreys KJ, Dawling S. Toxicity and deaths from 3,4-methylenedioxymethamphetamine (ecstasy). Lancet 1992; 340: 3847 Milroy CM, Clark JC, Forrest ARW. Pathology of deaths associated with ecstasy and eve misuse. J Clin Pathol 1996; 49: 14953 Bertram M, Egelhoff T, Schwarz S, Schwab S. Toxic leukoencephalopathy following ecstasy ingestion. J Neurol 1999; 246: 61718 Bitsch A, Thiel A, Rieckmann P, Prange H. Acute inammatory CNS disease after MDMA (ecstasy). Eur Neurol 1996; 36: 3289 Gledhill JA, Moore DF, Bell D, Henry JA. Subarachnoid haemorrhage associated with MDMA abuse. J Neurol Neurosurg Psychiatry 1993; 56: 10367

200 Hanyu S, Ikeguchi K, Imai H, Imai N, Yoshida M. Cerebral infarction associated with 3,4methylenedioxymethamphetamine (ecstasy) abuse. Eur Neurol 1995; 35: 173 201 Harries DP, De Silva R. Ecstasy and intracerebral haemorrhage. Scott Med J 1992; 37: 1502 202 Hughes JC, McCabe M, Evans RJ. Intracranial haemorrhage associated with ingestion of ecstasy. Arch Emerg Med 1993; 10: 3724 203 Manchanda S, Connolly MJ. Cerebral infarction in association with ecstasy abuse. Postgrad Med J 1993; 69: 8745 204 Spatt J, Glawar B, Mamoli B. A pure amnestic syndrome after MDMA (ecstasy) ingestion. J Neurol Neurosurg Psychiatry 1997; 62: 41819 205 Squier MV, Jalloh S, Hilton-Jones D, Series H. Death after ecstasy ingestion: neuropathological ndings. J Neurol Neurosurg Psychiatry 1995; 58: 756 206 Commins DL, Vosmer G, Virus RM, Woolverton WL, Schuster CR, Seiden LS. Biochemical and histological evidence that methylenedioxymethamphetamine (MDMA) is toxic to neurons in the rat brain. J Pharmacol Exp Ther 1987; 241: 33845 207 Ricaurte GA, McCann UD, Szabo Z, Scheffel U. Toxicodynamics and long-term toxicity of the recreational drug, 3,4-methylenedioxymethamphetamine (MDMA, ecstasy). Toxicol Lett 2000; 112: 1436 208 Curran HV. Is MDMA (ecstasy) neurotoxic in humans? An overview of evidence and of methodological problems in research. Neuropsychobiology 2000; 42: 34 41 209 Lyles J, Cadet JL. Methylenedioxymethamphetamine (MDMA, ecstasy) neurotoxicity: cellular and molecular mechanisms. Brain Res Brain Res Rev 2003; 42: 15568 210 Sprague JE, Everman SL, Nichols DE. An integrated hypothesis for the serotonergic axonal loss induced by 3,4-methylenedioxymethamphetamine. Neurotoxicology 1998; 19: 42742 211 Simantov R, Tauber M. The abused drug MDMA (ecstasy) induces programmed cell death of human serotonergic cells. FASEB J 1997; 11: 1416 212 Zhou JF, Chen P, Zhou YH, Zhang L, Chen HH. 3,4Methylenedioxymethamphetamine (MDMA) abuse may cause oxidative stress and potential free radical damage. Free Radic Res 2003; 37: 4917 213 Gouzoulis-Mayfrank E, Daumann J. Neurotoxicity of methylenedioxyamphetamines (MDMA; ecstasy) in humans: how strong is the evidence for persistent brain damage? Addiction 2006; 101: 34861 214 Kish SJ. How strong is the evidence that brain serotonin neurons are damaged in human users of ecstasy? Pharmacol Biochem Behav 2002; 71: 84555 215 McGuire P. Long term psychiatric and cognitive effects of MDMA use. Toxicol Lett 2000; 112113: 1536 216 Parrott AC, Marsden CA. MDMA (3,4methylenedioxymethamphetamine) or ecstasy: the

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134

134

A. Bttner

217

218

219

220

221

222

223

224

225

226

227

228

contemporary human and animal research perspective. J Psychopharmacol 2006; 20: 1436 Parrott AC. Human psychopharmacology of ecstasy (MDMA): a review of 15 years of empirical research. Hum Psychopharmacol Clin Exp 2001; 16: 55777 Reneman L, Booij J, Majoie CBL, van den Brink W, den Heeten GJ. Investigating the potential neurotoxicity of ecstasy (MDMA): an imaging approach. Hum Psychopharmacol Clin Exp 2001; 16: 57988 Verbaten MN. Specic memory decits in ecstasy users? The results of a meta-analysis. Hum Psychopharmacol Clin Exp 2003; 18: 28190 Makrigeorgi-Butera M, Hagel C, Laas R, Pschel K, Stavrou D. Comparative brain pathology of HIVseronegative and HIV-infected drug addicts. Clin Neuropathol 1996; 15: 3249 Boronat MA, Garca-Fuster MJ, Garca-Sevilla JA. Chronic morphine induces up-regulation of the proapoptotic Fas receptor and down-regulation of the antiapoptotic Bcl-2 oncoprotein in rat brain. Br J Pharmacol 2001; 134: 126370 Hu S, Sheng WS, Lokensgard JR, Peterson PK. Morphine induces apoptosis of human microglia and neurons. Neuropharmacology 2002; 42: 82936 Jiang Y, Yang W, Zhou Y, Ma L. Up-regulation of murine double minute clone 2 (MDM2) gene expression in rat brain after morphine, heroin, and cocaine administrations. Neurosci Lett 2003; 352: 21620 Mao J, Sung M, Ji R-R, Lim G. Neuronal apoptosis associated with morphine tolerance: evidence for an opioidinduced neurotoxic mechanism. J Neurosci 2002; 22: 765061 Stumm G, Schlegel J, Schfer T, Wrz C, Mennel HD, Krieg J-C, Vedder H. Amphetamines induce apoptosis and regulation of bcl-x splice variants in neocortical neurons. FASEB J 1999; 13: 106572 Boronat MA, Olmos G, Garca-Sevilla JA. Attenuation of tolerance to opioid-induced antinociception and protection against morphine-induced decrease of neurolament proteins by idazoxan and other I2-imidazoline ligands. Br J Pharmacol 1998; 125: 17585 Ferrer-Alcn M, Garca-Sevilla JA, Jaquet PE, La Harpe R, Riederer BM, Walzer C, Guimn J. Regulation of nonphosphorylated and phosphorylated forms of neurolament proteins in the prefrontal cortex of human opioid addicts. J Neurosci Res 2000; 61: 33849 Ramage SN, Anthony IC, Carnie FW, Busuttil A, Robertson R, Bell JE. Hyperphosphorylated tau and amyloid precursor protein deposition is increased in the brains of

229

230

231

232 233

234

235

236

237

238

239

240

young drug abusers. Neuropathol Appl Neurobiol 2005; 31: 43948 Bttner A, Rohrmoser K, Mall G, Penning R, Weis S. Widespread axonal damage in the brain of drug abusers as evidenced by accumulation of b-amyloid precursor protein (b-APP): an immunohistochemical investigation. Addiction 2006; 101: 133946 Anthony IC, Ramage SN, Carnie FW, Simmonds P, Bell JE. Does drug abuse alter microglial phenotype and cell turnover in the context of advancing HIV infection? Neuropathol Appl Neurobiol 2005; 31: 32538. Tomlinson GS, Simmonds P, Busuttil A, Chiswick A, Bell JE. Upregulation of microglia in drug users with and without pre-symptomatic HIV infection. Neuropathol Appl Neurobiol 1999; 25: 36979 Miguel-Hidalgo JJ. The role of glial cells in drug abuse. Curr Drug Abuse Rev 2009; 2: 7682 Sorensen RG, Lawrence DMP. Glial cells and drugs of abuse in the nervous system. Drug Alcohol Depend 2009; 102: 1669 Anderson CE, Tomlinson GS, Pauly B, Brannan FW, Chiswick A, Brack-Werner R, Simmonds P, Bell JE. Relationship of Nef-positive and GFAP-reactive astrocytes to drug use in early and late HIV infection. Neuropathol Appl Neurobiol 2003; 29: 37888 Stadlin A, Lau JWS, Szeto YK. A selective regional response of cultured astrocytes to methamphetamine. Ann N Y Acad Sci 1998; 844: 10821 Castagnoli N Jr, Castagnoli KP. Metabolic bioactivation reactions potentially related to drug toxicities. NIDA Res Monogr 1997; 173: 85105 Sastre M, Ventayol P, Garca-Sevilla JA. Decreased density of I2-imidazoline receptors in the postmortem brains of heroin addicts. Neuroreport 1996; 7: 50912 Connor MD, Lammie GA, Bell JE, Warlow CP, Simmonds P, Brettle RP. Cerebral infarction in adult AIDS patients: observations from the Edinburgh HIV autopsy cohort. Stroke 2000; 31: 211726 Bttner A, Kroehling C, Mall G, Penning R, Weis S. Alterations of the vascular basal lamina in the cerebral cortex in drug abuse: a combined morphometric and immunohistochemical investigation. Drug Alcohol Depend 2005; 69: 6379 Geibprasert S, Gallucci M, Krings T. Addictive illegal drugs: structural neuroimaging. AJNR Am J Neuroradiol 2010; 31: 8038

Received 26 July 2010 Accepted after revision 30 September 2010 Published online Article Accepted on 6 October 2010

2011 The Author Neuropathology and Applied Neurobiology 2011 British Neuropathological Society, 37, 118134