Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jul 15.
Published in final edited form as: Biol Psychiatry. 2009 Apr 5;66(2):118–127. doi: 10.1016/j.biopsych.2009.02.021

Methamphetamine- and Trauma-Induced Brain Injuries: Comparative Cellular and Molecular Neurobiological Substrates

Mark S Gold 1,2,3,Ψ, Firas H Kobeissy 1,2,Ψ,, Kevin KW Wang 1,5,Ψ, Lisa J Merlo 2, Adriaan W Bruijnzeel 2, Irina N Krasnova 4, Jean Lud Cadet 4
PMCID: PMC2810951  NIHMSID: NIHMS128863  PMID: 19345341

Abstract

The use of methamphetamine (METH) is a growing public health problem because its abuse is associated with long-term biochemical and structural effects on the human brain. Neurodegeneration is often observed in humans as a result of mechanical injuries (e.g. traumatic brain injury, TBI) and ischemic damage (strokes). In this review, we discuss recent findings documenting the fact that the psychostimulant drug, METH, can cause neuronal damage in several brain regions. The accumulated evidence from our laboratories and those of other investigators indicates that acute administration of METH leads to activation of calpain and caspase proteolytic systems. These systems are also involved in causing neuronal damage secondary to traumatic and ischemic brain injuries. Protease activation is accompanied by proteolysis of endogenous neuronal structural proteins (αII-spectrin and MAP-tau protein) evidenced by the appearance of their breakdown products after these injuries. When taken together, these observations suggest that METH exposure, like TBI, can cause substantial damage to the brain by causing both apoptotic and necrotic cell death in the brains of METH addicts who use large doses of the drug during their lifetimes. Finally, because METH abuse is accompanied by functional and structural changes in the brain similar to those in TBI, METH addicts might experience greater benefit if their treatment involved greater emphasis on rehabilitation in conjunction with the use of potential neuroprotective pharmacological agents such as calpain and caspase inhibitors similar to those used in TBI.

Keywords: neurotoxicity, methamphetamine, psychoproteomics, proteolysis, calpain, caspase, αII-spectrin, neuroproteomics, brain injury

Introduction

Substance use disorders are chronic relapsing conditions that are very prevalent throughout the world. These drugs are thought to impact or “highjack” the brain’s motivational and reward centers, which then mediate the long-term usage of these drugs (1). Furthermore, the discontinuation of drug self-administration leads to negative emotional states that may provide additional motivation for the continuation of drug abuse (2). Although these ideas have influenced basic and clinical research related to the rewarding effects of drugs of abuse and drug withdrawal, they have not led to substantial revolution in our treatment of the cognitive deficits in drug abusing individuals. In fact, many fundamental issues regarding the long-term effects of drugs of abuse remain to be elucidated. For example, it has not yet been determined if there exist specific markers of addiction and recovery. It is still not clear if drug addiction is associated with permanent drug-mediated biochemical and structural changes in the human brain. Given the presence of neuropsychiatric disorders in many drug abusers, it will be important to decipher the neuropathological substrates for the signs and symptoms that impact patients’ daily activities. The accumulated evidence suggests that some drugs can cause inflammatory responses, substantial loss of neurotransmitters, as well as neuronal death in animal models using these drugs to mimic the human conditions (3). Thus, the purpose of this review is to compare and contrast the cellular and molecular events that occur in two brain disorders, namely traumatic brain injury (TBI) and methamphetamine (METH) abuse. These data will then be used as a springboard to suggest that METH addicts might benefit substantially from long-term rehabilitative approaches in conjunction with neuroprotective agents similar to those used in trauma patients.

Traumatic Brain Injury

Neuropsychiatric complications of TBI

TBI occurs when the brain is damaged after being impacted by an external force to the head region (4). There are more than two million TBI incidents per year (5). These injuries result in 500,000 hospitalizations, 80,000 patients with long term disabilities, and 100,000 deaths annually in the US. The symptoms of TBI can vary from mild to severe. The patients can complain of headache, confusion, blurred vision, behavioral changes, mood disorders, and other neurological symptoms (6, 7). Longterm deficits correlate with pathological changes on CAT scans or MRI (8, 9). Neuroimaging studies have revealed significant reduction in hippocampal volume and enlargement of the lateral ventricles (8). Detailed neuropsychological assessments have revealed deficits in attention and processing (10). Successful rehabilitation strategies that address cognitive and other functional deficits in these patients include intensive neuropsychological interventions and the use of pharmacological agents to enhance cognitive improvements (11).

Mechanisms of TBI-induced cell death

Studies assessing the bases of TBI-induced neuropathological changes have depended mostly on animal models (12). These investigations have lead to the conclusion that the pathological substrates of TBI are secondary to a two-step mechanism which involved primary and secondary injuries (13). The primary injury results in the compression of neuronal, glial, astrocytic and vascular tissue (13). The primary injury is associated with disruption of cell membrane and disturbances of ionic homeostasis as a consequence of membrane leakiness (13, 14). The initial impact to the brain has been shown to activate multiple death pathways which include activation of calpains (calcium-dependent protease with papain) and caspases protease systems (4, 14).

Secondary injuries proceed within minutes to days after head injury and culminate in widespread cell death (4, 15). TBI-induced cell death is accompanied by release of excitatory amino acids, increased production of reactive oxygen species (ROS), and disruption of mitochondrial bioenergetics, disturbances in calcium homeostasis, and neuroinflammatory responses such as reactive gliosis (14, 16, 17). These disturbances are then accompanied by the activation of a cascade of events which include activation of calcium-dependent enzymes, destruction of the cellular architectures, and ultimately the demise of various cell types in the brain (17, 18). This cellular demise occurs via the activation of both necrotic and apoptotic death pathways (16).

Necrosis and apoptosis are two different forms of cell death with different implications for the surrounding tissues (19). Necrosis is a very rapid form of death that can affect large cell populations (20). It results from environmental perturbations (physical and chemical insults) leading to cellular injury. These injuries result in a massive increase in calcium influx that causes cell swelling and nonspecific DNA damage. This is coupled with spillage of intracellular content to the extra-cytoplasmic space, which leads to surrounding tissue inflammation concomitant with calpain protease activation. Apoptotic cell death, also referred to as programmed cell death or type I cell death, is characterized by cell shrinkage, changes in nuclear morphology, blebbing of the plasma membrane, and formation of apoptotic bodies (20). Biochemically, apoptosis is characterized by the activation of cysteine proteases called caspases. Upon activation, caspases act on a number of cytosolic, cytoskeletal and neuronal protein substrates including DNA function related factor-45 (DFF-45), lamin A, poly (ADP-ribose) polymerase (PARP) and αII-spectrin (21). Apoptosis can occur via activation of intrinsic and extrinsic pathways. The intrinsic pathway involves release of several proteins from the mitochondria and subsequent activation of caspase 9 and caspase 3, and caspase-3-mediated destruction of cytoplasmic and nuclear proteins (22-24). The extrinsic pathway is initiated via ligation of death receptors (FasL/Fas pathway) which results in the aggregation of the adaptor molecule Fas-associated death domain (FADD) and activation of caspase-8 (25, 26). Activation of caspase-8 leads activation of caspase-3 and to the truncation of the pro-apoptotic Bcl-2 family protein, Bid, which activates the release of pro-apoptotic proteins form the mitochondria (26, 27).

TBI-induced cell death has been shown to involve activation of the calpain family of proteases. Calpains are cytoplasmic, calcium-activated neutral cysteine proteases (19). Calpains reside in the cytosol as pro-calpain and translocate to the plasma membrane in response to increased Ca2+ levels (19). Calcium induces structural changes leading to calpain activation via the autolytic processing of calpain molecules, generating a smaller functional unit. Among the 14 identified mammalian calpains, two calpains are known to be active after TBI including μ-calpain or calpain-1 which requires micromolar of Ca2+ for activation and m-calpain or calpain-2 which requires millimolar of Ca2+ for activation (19, 28). Upon their activation, calpains cause the degradation of and the appearance of unique breakdown products of various proteins which include cytosolic collapsin response mediator protein-2 (CRMP-2), cytoskeletal αII-spectrin, neurofilament protein, PARP protein and microtubule associated protein-2 (MAP-tau) (29-38). Although calpain activation has been associated mainly with necrotic cell death, some recent studies have also demonstrated their potential involvement in some models of apoptosis (39, 40). Nevertheless, TBI-induced apoptosis appears to involve mainly the activation of caspase-3 (41, 42).

Neuronal Cell Death and Degradomics Patterns in TBI

Among the proteins, which have been extensively studied in the area of TBI are the cytoskeletal αII-spectrin protein and (MAP)-tau protein. αII-spectrin (280 kDa) is a major component of cell membrane cytoskeleton. Of interest, αII-spectrin is a major substrate for both calpain and caspase-3 proteases (35, 43). Several in vivo and in vitro studies have provided substantial evidence indicating that αII-spectrin is processed by calpain and caspase proteases to generate signature proteolytic breakdown products indicative of necrotic and apoptotic activation after brain injury (37, 44). Calpain degradation of α-II spectrin results in the appearance of two unique and highly stable α-II spectrin breakdown products of 150 kDa and 145 kDa (SBDP150 and SBDP145), which occurs early in neural cell pathology indicative of necrotic/excitotoxic neuronal cell death (Fig. 1). Similarly, caspase-3 activation results in a 150 kDa SBDP, which is further cleaved into a 120 kDa fragment (SBDP120) indicative of apoptotic neuronal cell death (Fig. 1) (43). TBI has been shown to induce calpain- and caspase-dependent degradation of structural proteins in humans and in animal models (Table I). The levels of αII-spectrin breakdown products (SBDPs) in cerebrospinal fluid (CSF) from adults with severe TBI (41 patients) were examined to assess the severity of brain injury and clinical outcome (45). Findings from this study indicated that calpain and caspase-3 increased SBDP levels in CSF were significantly increased in TBI patients at several time points after injury, compared to control subjects. Taken together these data suggest that both necrotic/oncotic and apoptotic cell death mechanisms are activated in humans following severe TBI, but with a different time course after injury. Siman et al (2004) have also examined the CSF of rats for differential protein expression in a model of mild/moderate experimental TBI employing two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) proteomic techniques. Tau protein fragment of 17 kDa, the αII-spectrin breakdown product of 150 kDa (SBDP150) and the collapsing response mediated protein-4 were increased in the CSF after the brain insult. Other proteins released in the CSF included GAP-43 and 14-3-3ζ, which are indicative of necrotic cell death whereas proteins such αII-spectrin breakdown as the products 120 kDa (SBDP120) were suggestive of apoptotic cell death (46).

Figure 1.

Figure 1

Schematic diagram showing calpain (red) and caspase-3 (blue) specific proteolysis following traumatic brain injury and METH expposure. As discussed, TBI and METH exposure will induce activation of the calpain and caspase protease system that leads to the proteolysis of different cell death proteins (PARP and DFF-4) along with structural products (Tau, αII spectrin and lamin A) which will generate signature breakdown products (BDPs) indicative of the selectivitiy of either capase (blue) or calpain (red) activation.

Table I.

Summary of recent traumatic brain injury studies showing evidence of cytoskeletal protein degradation and cell death activation induced by calpain/caspase protease system.

TBI Model Tissue Proteins measured, time-points Cellular cascades References
Rat, controlled cortical impact
(1.6 mm compression)
Cortex ↓280 kDa αII-spectrin, ↑ 120 kDa SBDP,
↓65 kDa kDa synaptotagmin,
↑37 kDa synaptotagmin BDP,
↓62 kDa CRMP-2, ↑55 kDa CRMP-2 BDP,
↑75 kDa transferrin, ↑26 kDa C-reactive protein,
↓200 kDa microtubule-associated proteins
MAP2A/2B, ↓102 kDa hexokinase,
↓GAPDH 48 h after injury
Degradation of
cytoskeleton-
associated proteins
Kobeissy et al., (2006)
Rat, controlled cortical impact,
(1.6 mm compression)
Hippocampus ↑caplpain-2 specific 145&150 kDa SBDP
110 kDa βII-spectrin BDP,
40&35 kDa striatin BDP,
33 kDa synaptotagmin-1 BDP;
↑caspase-3 specific 149&120 kDa SBDP
108&85 kDa βII-spectrin BDP,
100 kDa striatin BDP;
↑70 kDa synaptojanin-1 BDP,
↑30&25 kDa N-ethylmaleimide sensitive
fusion protein BDP at 48 h after injury
Calpain-2 and
caspase-3-specific
proteolysis
Liu et al., (2006)
Rat, controlled cortical impact
(moderate)
Cortex,
hippocampus
↑protein carbonyls, ↑3-nitrotyrosine,
↑4-hydroxynonenal in the hippocampal
mitochondria, ↑oxidized pyruvate dehydrogenase,
voltage gated anion channel, ATP synthase,
cytochrome c oxidase Va, GAPDH and prohibitin
in the cortex
Oxidative damage
to mitochondrial
proteins,
mitochondrial
dysfunction
Opii et al., (2007)
Rat, controlled cortical impact
(2.3 mm compression)
Cortex,
hippocampus,
thalamus,
striatum
↑145 kDa SBDP in the cortex at 3 h-5d,
hippocampus at 24 h-7 d and thalamus
15 min-14 d after injury;
↑120 kDa SBDP in the hippocampus
at 15 min-6 h and striatum at 3 h-2 d
following injury
Caspase-3- and
calpain-dependent
spectrin proteolysis
Pike et al., (1998)
Rat, controlled cortical impact
moderate (2.0 mm) compression
Cortex,
CSF
↓αII spectrin in the cortex,
↑αII spectrin in the CSF,
↑150 & 145 kDa SBDP
in the cortex and CSF 1-3 d afterinjury
Calpain-specific
αII-spectrin
proteolysis
Pike et al., (2001)
Human, severe TBI,
Glasgow Coma Scale score ≤8
CSF ↑145 & 150 kDa SBDP at 6 h-3 d and
↑120 kDa SBDP at 6 h-5 d following TBI
Calpain- and
caspase-3-mediated
αII-spectrin
proteolysis
Pineda et al., (2007)
Rat, lateral fluid percussion;
mild (1.2 atm fluid pressure),
moderate (2.4 atm fluid
pressure)
CSF ↑14-3-3ζ, ↑14-3-3β,
↑αII spectrin (280 kDa),
↑150 kDa SBDP, ↑115-120 kDa SBDP
at 6-24 h after injuty; ↑tau 28 kDa,
↑tau 17 kDa 48 h after injury
Calpain- and
caspase-dependent
proteolysis
Siman et al., (2004)
Rat, controlled cortical impact,
(1.6 mm compression)
Cortex,
hippocampus
↑55 kDa CRMP-2 BDP,
↑58 kDa CRMP-4 BDP in the cortex
at 24-48 hours, CRMP-2 BDP at hippocampus
at 48 hours following injury
calpain-2 treatment caused
↑55 kDa CRMP-2 BDP in naïve brain lysate,
Calpain-2-mediated
CRMP-2 proteolysis
Zhang et al., (2007)
Mouse, Flat Impounder Model Cortex ↑ 85 kDa BDP of PARP and
↓32 kDa Procaspase -3
Caspase Hutchison et al. (2001)

Abbreviations: BDP — breakdown product, CRMP - collapsin response mediator protein, ER — endoplasmic reticulum, GAPDH - glyceraldehyde-3-phosphate hydrogenase, SBDP — αII-spectrin breakdown product, TBI — traumatic brain injury, PARP — 1Poly (ADP-ribose) polymerase-1.

In our laboratories, we have used a novel offline multidimensional separation platform termed, cation-anion exchange chromatography (CAX-PAGE/RPLC-MSMS), which is comprised of tandem ion exchange fractionation followed by 1D-PAGE separation, as a novel approach for identifying TBI biomarkers and protein breakdown products (degradomes) (32, 47). We have used the CAX/neuroproteomic analysis on cortical samples obtained from rats 48 hours after a controlled cortical impact (CCI) model of experimental TBI (32). There were 59 differential protein components, of which 21 showed decreased levels and 38 showed increased levels after TBI. Among these, CRMP-2, MAP-2A/2B, and hexokinase were down-regulated whereas C-reactive protein and transferrin were up-regulated after the trauma. This work also identified novel protease substrates such as CRMP-2 and synaptotagmin,.

CRMPs are major proteins involved in neurite outgrowth and axonal guidance and alterations in their expression have been implicated in several neurological diseases including Alzheimer’s, TBI, and ischemia. The CRMPs were subjected to in vitro calpain and caspase digestion (31). These studies allowed us to identify calpain-2, but not the caspases, as the possible proteolytic mediator of CRMP-2 breakdown following TBI.

Liu et al (2006) conducted a similar line of inquiry using high throughput immunoblotting (HTPI) technology, a novel proteomic methods for studying differential expression of proteins, in an effort to identify protease substrates for calpains and caspase-3 in an experimental TBI model (48). The authors identified 92 proteins, of which 54 were substrates sensitive to calpain-2 digestion and 38 were sensitive to caspase-3 proteolysis. This study revealed an array of proteins including β-spectrin, synaptotagmin-1, and synaptojanin-1 that are vulnerable to proteolysis following TBI (48).

Because mitochondrial dysfunction is thought to integrate various death pathways in TBI-induced neuropathology, Opii et al (2007) utilized the proteomics approach to assess potential TBI-induced changes in mitochondrial proteins in the cortex and hippocampus of rats following moderate TBI (49). Cortical and hippocampal proteins that were oxidatively modified following brain injury include pyruvate dehydrogenase and prohibitin. These data further support a role for oxidative stress as a mediator of TBI-induced brain damage (49).

In conclusion, this review indicates that many of the changes occurring in response to TBI are reflected by complex alterations in protein dynamics of relevance to protein expression, protein interaction, and protein proteolysis. These changes then are the harbinger of necrotic and apoptotic cell death in TBI models of neurodegeneration.

Methamphetamine-Induced Brain Injuries

Neuropsychiatric complications of METH addiction

Amphetamine derivatives including METH are among the most widely abused, illegal form of amphetamines, and are estimated to be abused by 25 million people worldwide. The drugs are used because they cause euphoria, hypersexuality, and increased energy (50). METH abuse is associated with a number of negative consequences, which include cognitive dysfunctions and neurological adverse events (51, 52). Large doses of METH can also cause life-threatening hyperthermia, cerebrovascular hemorrhages, seizures, and death (51, 53). Chronic abuse of METH is associated with withdrawal-induced depression, psychosis, and psychomotor dysfunctions (52, 53). METH-induced neuropsychological abnormalities include attention deficits, memory problems, and poor decision-making (54). The accumulated evidence suggests neuropsychiatric consequences of METH abuse are related to drug-induced neuropathological changes in the brains of these METH-exposed individuals reviewed in (3).

Interestingly, METH abuse has been linked to numerous adverse neuropsychological effects showing deficits in execution memory (novel problem solving), motor skills, and episodic memory, which have been interpreted with regard to the affected dopamine rich fronto-striato-cortical loops. METH associated-episodic memory impairment is among the most susceptible cognitive functions related to METH relapse likely related to METH-induced neurotoxicity leading to brain injury. In addition, METH abusers show evidence of risky decision making and impulsivity, which has been linked to the executive aspects of working memory deficits (52, 55).

Koob and colleagues investigated the effect of METH self-administration on gliogenesis in the medial prefrontal cortex (mPFC) (56). They demonstrated that daily METH self-administration (1 hour or 6 hour / day) decreased gliogenesis and increased cell death in the mPFC. Glial cells play an important role in neuronal survival and therefore a METH-induced decrease in gliogenesis might have a negative effect prefrontal cortex functioning. Schoenbaum and colleagues evaluated the effects of cocaine on orbitofrontal cortex (OFC) functioning (57). It was found that cocaine-treated rats, who demonstrated long-lasting sensitization to the locomotor activating effects of cocaine, display deficits in odor discrimination learning. Similar deficits in this task have been detected in rats with OFC lesions (58). Taken together, these studies suggest that chronic drug intake could lead to impairments in the frontal cortex which could lead to cognitive impairments and increased drug taking behavior (59).

Studies from patients suffering from either acquired brain injury to the frontal cortex or drug addictions support the link between frontal-subcortical systems injury and risk taking behaviors. Carcuel et al showed that patients with acquired frontal cortex brain injury and drug addictions share a range of neuropsychiatric dysfunctions including apathy, poor self-control, and poor executive control, as evaluated by the Frontal Systems Behavioral Scale (FrSBe). It was shown that the addicted subjects, along with the brain injured patients, exhibited greater impairments than control subjects (60). A similar study was conducted by Lange et al, in which 104 patients with mild TBI were compared to 104 substance abuse patients (61). It was shown that there were no differences between the neuropsychological test performances of TBI patients and addicted patients on cognitive measures of visual and verbal memory and executive functioning (61). Interestingly, in one study by Regard et al, gambling behavior was evaluated in pathologic gamblers. It was shown that these “addicted” subjects had fronto-temporal neuropsychological dysfunctions and that the compulsive gambling may be a consequence of brain damage to the fronto-limbic systems (62). Based on the above studies, we argue that brain injuries due to mechanical injury and substance abuse, share similar molecular profiles and tend to increase the risk of developing addictive behavior.

Mechanisms of METH neurotoxicity

In addition to the signs and symptoms of neuropsychiatric disturbances, METH can indeed cause neurodegenerative changes in the human brain (63). These neuropathological changes include persistent decreases in the levels of dopamine transporters (DAT) in the orbitofrontal cortex, dorsolateral prefrontal cortex, and the caudate-putamen (64). They also include decreases in the density of serotonin transporters (5-HTT) in various brain regions, including the orbitofrontal and cingulate cortices of METH-dependent individuals (65). Structural magnetic resonance imaging (MRI) has documented loss of gray matter in various cortical areas (66). METH abusers also suffer from decreases in the neuronal marker, N-acetylaspartate (67) and increases in the glial marker, myoinositol (67). Microglial activation has also been reported in the brain of METH abusers using PET imaging techniques (68). In post-mortem analyses, there were decreases in dopamine (DA), tyrosine hydroxylase, and DAT levels in the basal ganglia of chronic METH users (69).

Studies in a number of animal species have replicated some of the observations obtained from clinical populations (70, 71); see Cadet et al 2007 for detailed review (3). METH can cause depletion of DA, 5-HT, and their metabolites in rodents and nonhuman primates (72). METH also causes degeneration of DA and 5-HT terminals (73, 74). Although the exact mechanisms involved in METH toxicity remain to be fully determined, they are similar to those reported in models of TBI, and include formation of oxidative radicals, release of glutamate, and the activation of apoptotic and necrotic death pathways (75, 76).

METH neurotoxicity is thought to be mediated by the redistribution of DA from vesicular storage vesicles to the cytoplasm and extracellular space and the subsequent generation of ROS, quinine by-products, and associated lipid peroxidation of the membrane of monoaminergic terminals (77). The release of glutamate (78) and the subsequent formation of nitric oxide have also been invoked as culprits in METH-related problems (79). In addition to its toxic effects on monaminergic terminals, METH can also cause cell death via both apoptotic and necrotic mechanisms (3, 71, 77, 80). Cell death is observed in hippocampal remnants (81), the parietal cortex (82), the striatum (83), and in several other brain regions (83).

The bases of METH-induced neuronal cell death have been extensively studied (Table II). METH-induced neuronal cell death is associated with activation of both ER- and mitochondria-dependent death pathways which interact to cause the ultimate demise of cells (80). Specifically, mice injected with METH showed an almost immediate activation of calcium-dependent calpain, caspase-12, increased expression of GRP78/BiP, and of C/EBP homologous protein (CHOP) which are all involved in ER stress-mediated apoptosis (80). There was also release of proteins from the mitochondria including cytochrome c and activation of caspase-9 and caspase 3. These events were followed by the proteolysis of caspase substrates such as DFF-45, lamin A and PARP in the nucleus (Fig. 1) (80). The role of the mitochondrial death pathway was also supported by the demonstration that METH is associated with transcriptional and translational increases in the expression of pro-apoptotic Bcl-2-related genes (Bad, Bax and Bid), but decreases in the expression of Bcl-2 and Bcl-XL in the mouse cortex (84). Similar studies have been conducted in the mouse olfactory bulb where both DA depletion and death of DA neurons were reported by Deng et al (85).

Table II.

Summary of recent reports studying mechanisms of cell death and altered protein dynamics induced by METH exposure.

METH treatment model Tissue Proteins measured, time-points Cellular cascades References
METH i.p. single injection,
40 mg/kg
Mouse
olfactory bulb
↑cleaved caspase-3
↑Bax, Bid protein expression (4h-24h)
↓Bcl-2 protein expression (4h-24h)
Activation of
Bcl-2-dependent
apoptotic pathway
Deng et al., (2007)
METH i.p. single ingection,
40 mg/kg
Mouse cortex ↑Bax, Bak, Bad, Bid mRNA (4 h-24 h) and
protein levels (30 min-2 d);
↓Bcl-2, BclXL, Bclw mRNA (1h -24 h) and
protein levels (1h-7d)
Activation of
Bcl-2 dependent
apoptotic pathway
Jayanthi et al., (2001)
METH i.p. single injection,
40 mg/kg
Mouse
striatum
↑cleaved caspase-12 (40 kDa) at 1h-2d,
↑cleaved caspase-9 (37 kDa) at 8h-2d,
↑cleaved caspase-3 (17 kDa) at 3h-7d
↓calpain (80 kDa) at 30 min-7d, ↑cleaved calpain
(76 kDa) at 30 min-7d; ↑GRP78/BiP mRNA
(30 min-4h) and protein (8h-7d) expression,
↑CHOP/GADD153 mRNA (30 min-2h) and
protein (4h-7d) expression; ↑transition of AIF,
smac/DIABLO, cytochrome c from mitochondria
to cytoplasm; proteolysis of DFF-45,
lamin A (8h-7d), RARP (16h-2d) in nucleus
Activation of
ER-dependent and
mitochondria-
dependent neuronal
apoptosis
Jayanthi et al. (2004)
METH i.p. injections
10 mg/kg × 4, every 2h
Rat striatum ↑145 kDa SBDP at 5-7d after drug
treatment, blocked by AMPA receptor
antagonist, but not by NMDA receptor antagonist
Calpain-mediated,
AMPA receptor-
dependent spectrin
proteolysis
Staszewski and Yamamoto,(2006)
METH i.p. four injections:
10 mg/kg × 2, 7.5 mg/kg and
5 mg/kg 2 h apart
Rat striatum,
hippocampus
↑cleaved tau immunoreactivity
in the astrocytes in striatum, CA2/CA2
and dentate gyrus at 3d post-drug
METH-induced
reactive gliosis
Straiko et al., (2007)
METH i.p. injections,
10 mg/kg × 4, every 2h
Rat striatum,
hippocampus,
cortex
↑cleaved tau in the striatum (3d-7d)
prevented by cold ambient temperature;
↑cleaved tau in the cortex and hippocampus (3d)
METH-induced
skeletal damage
Wallace et al. (2003)
METH i.p. injections,
Doses 10 mg/kg × 1,
10 mg/kg × 2 or 10 mg/kg × 3,
1 h apart
Rat cortex,
hippocampus
↑145 &150 kDa SBDP,
↑36, 32 & 26 kDa tau BDP in the cortex
and hippocampus (24h); ↑120 kDa SBDP
in the hippocampus (24h)
Calpain-dependent
αII-spectrin and tau
proteolysis,
caspse-3-dependent
αII-spectrin
proteolysis
Warren et al., (2005)
METH i.p. injections,
10 mg/kg × 4, every 1h
Rat cortex ↑active calpain-1 and caspase-3 immuno-
reactivity (48h) in neurons; ↑145 and 120 kDa
SBDP immunoreactivity (48h) in neurons
Neuronal calpain-
and caspase-
mediated αII-spectrin
proteolysis
Warren et al.,
(2007a)
METH 1 or 2 mM,
24 & 48 h exposure
Rat primary
cerebro-
cortical
neuronal
culture
↑145 and 120 kDa SBDP,
↑45, 32, 26 & 14 kDa tau BDP (24 & 48 h);
↑150 kDa SBDP (48h);
calpain inhibitor caused ↓formation of 145 kDa
SBDP and 14 kDa tau BDP,
caspase inhibitors caused ↓levels of 120 kDa
SBDP and 45 kDa tau BDP
Calpain- and
caspse-mediated
αII-spectrin and tau
proteolysis
Warren et al.,
(2007b)

Abbreviations: BDP — breakdown product, ER — endoplasmic reticulum, SBDP - αII-spectrin breakdown product.

The possibility of METH-induced necrotic death has been supported by some recent studies. For example, METH administration can cause the release of glutamate, which was shown to cause activate of calcium-dependent calpains via interactions with calcium permeable AMPA receptors, resulting in cleavage of their cytoskelatal substrates including αII-spectrin (86). This activation was blocked by the AMPA receptor antagonist, GYKI 52466 (86). Exposing the animals to stressful events appeared to exacerbate the effects of METH on the breakdown of spectrin (87). METH can also cause proteolysis of the neuronal cytoskelatal protein, microtubule-associated protein (MAP-tau) in the striatum, and in the hippocampus (88). MAP-tau is localized within neuronal axonal compartment and contributes to the formation of microtubule bundles that are structural elements of axonal cytoskeleton (89).

Other studies have also assessed the effects of METH on protein expression and have compared the protein breakdown products observed in TBI models (41, 43). In a study from our laboratory, neurotoxicity was evaluated in rat model, 24 hrs after treatment at different concentrations (10 mg/kg-40 mg/kg). We tried to achieve a dosage paradigm similar to what is considered ‘normal acute’ or ‘binge’ use for humans (90, 91). It has been shown that the 4×10 mg/kg paradigm of METH exposure in naive animals is currently the most frequently used model that mimics acute toxic dosing of METH (71). This paradigm provides excellent relevance to the intravenous and smoked METH in humans, in addition, it demonstrates the toxic effects of METH in non-tolerant users (71).

In our work, cortical and hippocampal brain regions were evaluated for the presence of cytoskelatal structural protein proteolysis as markers for necrotic and apoptotic cell death post METH injections (91). There were significant increases in the levels of products of the breakdown of αII-spectrin (SBDP120 and SBDP150) and of MAP-tau (26 kDa, 32 kDa, and 36 kDa), which are thought to be indicative of neuronal cell death processes (Fig. 1) (92, 93). These breakdown products suggest that METH cause activation of the pro-apoptotic caspase-3 and the pro-necrotic calpain-1 as previously reported in TBI models (91). Immunohistochemical studies have revealed that the SBDPs were localized mainly in the axonal regions of neuronal cells localized mainly within the cortex region of the brain (90). This observed profile mimics the phenotype observed in TBI as shown in previous studies (91).

Proteomics Analysis of Methamphetamine

Proteomic analyses are now considered invaluable tools in studies attempting to elucidate the cellular and molecular underpinnings of complex biological systems (94). As such, these approaches promise to revolutionize our understanding of the effects of pharmacological agents including drugs of abuse on the brain. For example, Sokolov and Cadet have investigated the effects of chronic METH treatment on protein expression using antibody microarrays and Immunoblotting techniques (95). METH administration caused significant decreases in MEK1, Erk2p, GSK3a, and MEK7 proteins in the striata of treated mice. Interestingly, Iwazaki et al used 2D-PAGE and reported that injections of METH caused alterations in the expression of proteins, which were involved in mitochondrial functions, oxidative stress, and degenerative processes (96). Iwazaki et al have also evaluated proteomic changes following METH-induced behavioral sensitization resulting from repeated METH administration, as well as reported changes in proteins involved in apoptotic pathways (97).

In a fashion similar to our investigations of the effects of TBI, we have used the novel multidimensional proteomic platform to evaluate the proteomic changes associated with acute METH-induced neurotoxicity. We found 82 differential protein components, of which 40 were decreased and 42 were increased in abundance following acute METH treatment (47). Identified proteins belonged to pathways involved in oxidative stress, synaptic transmission, and cell death-related proteins. The relative similarities of the proteomic changes associated with both TBI and acute METH treatment (shown in Table III) suggest that there might be common neurobiological events that are involved in causing the long-term neurological and neuropsychiatric effects of these kinds of trauma. Thus, proteomic approaches, when used with appropriate classification of patient populations, hold promise to identify relevant biomarkers that might have predictive therapeutic values.

Table III.

Summary of major altered (upregulated and downregulated) proteins post traumatic brain injury and METH exposure identified via proteomics/biochemical approach Ψ.

Upregulated Proteins common to
TBI and METH Exposuer
Proposed functions References
UCH-L1 Cytosolic neuronal protein involved in proteosomal regulation Kobeissy (2006, 2008)-Iwazaki (2006)
LC3 Homologue of autophagic ATG-8 protein involvedin autophagic cell death Kobeissy (2008) -Sadasivan (2006)
MAP kinase 8 MAP kinase proteins involved in in the cortical and striatal circuits Kobeissy (2006)- Sokolov (2006)
MAP kinase kinase 1 MAP kinase proteins involved in in the cortical and striatal circuits Kobeissy (2006)- Sokolov (2006)
Carbonic anhydrase Ametalloenzyme that catalyze the conversion of carbon dioxide to
bicarbonate and protons,
Kobeissy (2006, 2008)
Phosphoglycerate mutase 1 A glycolytic enzyme and interats with microtubles Jenkins (2002) -Iwazaki (2008),
Kobeissy (2008)
Peroxiredoxin Cytosolic protein participates in cellular growth and metabolism Jenkins (2002) -Iwazaki (2008)
Beta-synuclein cytoplasmic protein involved in neuroplasticity and neurodegenerative
diseases
Jenkins (2002) -Iwazaki (2008)
Gamma enolase Cytoplasmic protein with a trophic role in synaptic remodelling Jenkins (2002) -Kobeissy (2008)
Heat shock cognate 71 An HSP 70 family involved in ATP hydrolysis kobeissy Jenkins (2002) -Kobeissy (2008)
Phosphoglycerate kinase 1 A major glycolytic enzyme with a transferase activity Kobeissy(2006)-Iwazaki (2008)
Creatine kinase Cytoplasmic protein involved in energy metabolism Kobeissy (2006)-Iwazaki (2008)
Lactate dehydrogenase Cytoplasmic protein involved in oxidation of lactate to produce
pyruvate associated with cell death
Kobeissy (2006)- Iwazaki (2006)
Glycerdehyde-3-phosphate
dehydrogenase
Glycolytic cytoplasmic enzyme involved in apoptotic cell death Freeman (2005)- Jenkins (2002)-
Kobeissy (2008)
Downregulated Proteins common to
TBI and METH Exposuer
Proposed functions References
αII - Spectrin cytoskeletal proteins (major substrate for calpain and caspase) Kobeissy (2006, 2008)
Map-Tau cytoskeletal proteins (major substrate for calpain and caspase) Kobeissy (2006, 2008)
CRMP-2 Neuronal axonal/dendritic guidance protein (major substrate for calpain) Kobeissy (2006,2008)- Iwazaki (2008)
Cofilin-1 Cytoplasmic actin binding proteininvolvedin the assembly and
disassembly of actin filaments
Jenkins (2002) -Kobeissy (2008)
Tubulin beta chain Cytoskeletal protein, major component of microtubules Jenkins (2002) -Kobeissy (2008)
Pyruvate kinase isozymes Glycolytic enzyme that catalyzes the transfer of a phosphate groups Kobeissy (2006, 2008)
Glucose-regulated protein ER protein and stress protein facilitates protein assembly in the ER
precursor (GRP 78)
Jenkins (2002) -Iwazaki (2006)
HSP-60 Mitochondrial protein involved in protein import and folding Freeman (2005)- Jenkins (2002)-
Glutamate dehydrogenase 1 Mitochondrial matrix enzyme, with a key role in the energy homeostasis Kobeissy (2006)- Iwazaki (2008)
Aconitase Cytoplasmic protein with an enzymatic activity that catalyses the
stereo-specific isomerization of citrate
Kobeissy (2006)- Iwazaki (2008)
α-enolase A metalloenzyme responsible for the catalysis of 2-phosphoglycerate
(2-PG) to phosphoenolpyruvate (PEP)
Kobeissy (2006)- Freeman (2005)
Ψ

Protein identification was processed using different proteomic methods including 2D-DIGE, CAX-PAGE and antibody array. Protein data represent statistically significant differentially expressed proteins that were showing common trend of alteration (upregulated and downregulated) in both brain insults (METH exposure and TBI). Data were compiled from different studies as represented by the used references. Protein molecular function was derived from the Human protein Reference Database (http://www.hprd.org) and the ExPASy (Expert Protein Analysis System) proteomics server of the Swiss Institute of Bioinformatics (http://au.expasy.org/sprot/)

Conclusion

This review has highlighted similarities between METH-induced neurotoxicity and TBI. As discussed previously, data from Carcuel et al, Lange et al and Schoenbaum et al demonstrated the link between drug abuse and frontal-subcortical systems injury where acquired frontal injury pateints share a wide range of neuropsychiatric dysfunctions such as poor executive control (59-61). Both brain insults are coupled with neuronal injury that are mediated mainly by protease activation leading to cell death in the cortical brain region. These observations have been generated using classical and more modern molecular and proteomic approaches as shown in Tables I, II and III. When taken together, these data suggest that METH abuse can result in neurobiological consequences whose pattern can be classified as a syndrome of chemical brain injury.

Because TBI and METH-induced injuries share some neuropathological abnormalities showing elevated calpain/caspase-mediated neuronal death, it is not farfetched to also suggest that the pathological changes might play an important role in the long-term sequelae of the two syndromes. In the area of TBI, recent studies have demonstrated the use of calpain inhibitors would attenuate the progressive neuronal death and would even improve locomotor functions and reduce the functional and structural deterioration observed after experimental brain injury (98, 99). The use of these pharmacological agents may hint they can be used to ameliorate the neuropsychological dysfunctions observed in these patients. In fact, a recent study by Nimmrich et al (2008) has demonstrated that the use of calpain inhibitors would attenuate NMDA-mediated neuronal injury along with associated behavioral dysfunctions (cognitive deficits) occurring after excitotoxic lesions (100). The importance of these findings is that they demonstrate the hidden link between neuronal injury and neuropsychiatric impairments observed in different types of brain injury (mechanical or drug abuse-induced). This may suggest that combating neuronal loss would lead to better psychiatric treatment (please refer to the supplement section for a detailed discussion). Finally, because TBI patients benefit from rehabilitative interventions in conjunction with the use of specific pharmacological agents (calpain inhibitors), the research discussed in the present review may be interpreted to suggest that similar treatment approaches might be beneficial to METH patients who suffer from drug-induced brain damage.

Supplementary Material

01

Figure 2.

Figure 2

TBI/METH Exposure induces neurotoxicity via two protease-dependent cell death pathways (neural necrosis and apoptosis). The use of calpain inhibitor and caspase inhibitor can provide protection against METH-induced neurotoxicity and would ameliorate the psychiatric deficits observed.

Acknowledgments and Financial Disclaimer

This work was supported in part by the Donald Dizney Eminent Scholar Chair, held by Mark S Gold, M.D. Distinguished Professor, at McKnight Brain Institute, College of Medicine, and the Capebranch and Riverbranch Endowments, Department of Psychiatry at the University of Florida. Jean Lud Cadet, M.D. and Irina N. Krasnova, Ph.D. are supported by the Intramural Research Program of the DHHS/NIH/NIDA. Lisa J. Merlo, Ph.D. is supported in part by National Institute of Drug Abuse (NIDA) training grant T32-DA-07313-10 (PI: Cottler), on which she is a postdoctoral research scholar. This work was supported in part by the Department of Defense (DOD) grant DAMD17-03-1-0066 and National Institutes of Health (NIH) grants R01 NS052831, and R01 051431. The authors report no biomedical financial interests or potential conflicts of interest.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Wise RA. Drug-activation of brain reward pathways. Drug and alcohol dependence. 1998;51:13–22. doi: 10.1016/s0376-8716(98)00063-5. [DOI] [PubMed] [Google Scholar]
  • 2.Koob GF. A role for brain stress systems in addiction. Neuron. 2008;59:11–34. doi: 10.1016/j.neuron.2008.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cadet JL, Krasnova IN, Jayanthi S, Lyles J. Neurotoxicity of substituted amphetamines: molecular and cellular mechanisms. Neurotoxicity research. 2007;11:183–202. doi: 10.1007/BF03033567. [DOI] [PubMed] [Google Scholar]
  • 4.Moppett IK. Traumatic brain injury: assessment, resuscitation and early management. Br J Anaesth. 2007;99:18–31. doi: 10.1093/bja/aem128. [DOI] [PubMed] [Google Scholar]
  • 5.2006 http://www.ninds.nih.gov/disorders/tbi/detail_tbi.htm.
  • 6.Hoffmann B, Duwecke C, von Wild KR. Neurological and social long-term outcome after early rehabilitation following traumatic brain injury. 5-year report on 240 TBI patients. Acta neurochirurgica. 2002;79:33–35. doi: 10.1007/978-3-7091-6105-0_6. [DOI] [PubMed] [Google Scholar]
  • 7.Mateer CA, Sira CS. Cognitive and emotional consequences of TBI: intervention strategies for vocational rehabilitation. NeuroRehabilitation. 2006;21:315–326. [PubMed] [Google Scholar]
  • 8.Himanen L, Portin R, Isoniemi H, Helenius H, Kurki T, Tenovuo O. Cognitive functions in relation to MRI findings 30 years after traumatic brain injury. Brain Inj. 2005;19:93–100. doi: 10.1080/02699050410001720031. [DOI] [PubMed] [Google Scholar]
  • 9.Sherer M, Stouter J, Hart T, Nakase-Richardson R, Olivier J, Manning E, et al. Computed tomography findings and early cognitive outcome after traumatic brain injury. Brain Inj. 2006;20:997–1005. doi: 10.1080/02699050600677055. [DOI] [PubMed] [Google Scholar]
  • 10.Salmond CH, Sahakian BJ. Cognitive outcome in traumatic brain injury survivors. Current opinion in critical care. 2005;11:111–116. doi: 10.1097/01.ccx.0000155358.31983.37. [DOI] [PubMed] [Google Scholar]
  • 11.Tenovuo O. Pharmacological enhancement of cognitive and behavioral deficits after traumatic brain injury. Current opinion in neurology. 2006;19:528–533. doi: 10.1097/WCO.0b013e328010944f. [DOI] [PubMed] [Google Scholar]
  • 12.Finnie JW, Blumbergs PC. Traumatic brain injury. Veterinary pathology. 2002;39:679–689. doi: 10.1354/vp.39-6-679. [DOI] [PubMed] [Google Scholar]
  • 13.Reilly PL. Brain injury: the pathophysiology of the first hours.‘Talk and Die revisited’. J Clin Neurosci. 2001;8:398–403. doi: 10.1054/jocn.2001.0916. [DOI] [PubMed] [Google Scholar]
  • 14.Wennersten A, Holmin S, Mathiesen T. Characterization of Bax and Bcl-2 in apoptosis after experimental traumatic brain injury in the rat. Acta Neuropathol (Berl) 2003;105:281–288. doi: 10.1007/s00401-002-0649-y. [DOI] [PubMed] [Google Scholar]
  • 15.Hall ED, Sullivan PG, Gibson TR, Pavel KM, Thompson BM, Scheff SW. Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: more than a focal brain injury. J Neurotrauma. 2005;22:252–265. doi: 10.1089/neu.2005.22.252. [DOI] [PubMed] [Google Scholar]
  • 16.McIntosh TK, Juhler M, Wieloch T. Novel pharmacologic strategies in the treatment of experimental traumatic brain injury: 1998. J Neurotrauma. 1998;15:731–769. doi: 10.1089/neu.1998.15.731. [DOI] [PubMed] [Google Scholar]
  • 17.Sullivan PG, Rabchevsky AG, Waldmeier PC, Springer JE. Mitochondrial permeability transition in CNS trauma: cause or effect of neuronal cell death? J Neurosci Res. 2005;79:231–239. doi: 10.1002/jnr.20292. [DOI] [PubMed] [Google Scholar]
  • 18.Rego AC, Monteiro NM, Silva AP, Gil J, Malva JO, Oliveira CR. Mitochondrial apoptotic cell death and moderate superoxide generation upon selective activation of non-desensitizing AMPA receptors in hippocampal cultures. Journal of neurochemistry. 2003;86:792–804. doi: 10.1046/j.1471-4159.2003.01898.x. [DOI] [PubMed] [Google Scholar]
  • 19.Czogalla A, Sikorski AF. Spectrin and calpain: a ‘target’ and a ‘sniper’ in the pathology of neuronal cells. Cell Mol Life Sci. 2005;62:1913–1924. doi: 10.1007/s00018-005-5097-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Golstein P, Kroemer G. Cell death by necrosis: towards a molecular definition. Trends Biochem Sci. 2007;32:37–43. doi: 10.1016/j.tibs.2006.11.001. [DOI] [PubMed] [Google Scholar]
  • 21.Hutchison JS, Derrane RE, Johnston DL, Gendron N, Barnes D, Fliss H, et al. Neuronal apoptosis inhibitory protein expression after traumatic brain injury in the mouse. J Neurotrauma. 2001;18:1333–1347. doi: 10.1089/08977150152725632. [DOI] [PubMed] [Google Scholar]
  • 22.Stridh H, Kimland M, Jones DP, Orrenius S, Hampton MB. Cytochrome c release and caspase activation in hydrogen peroxide- and tributyltin-induced apoptosis. FEBS Lett. 1998;429:351–355. doi: 10.1016/s0014-5793(98)00630-9. [DOI] [PubMed] [Google Scholar]
  • 23.Hirsch T, Susin SA, Marzo I, Marchetti P, Zamzami N, Kroemer G. Mitochondrial permeability transition in apoptosis and necrosis. Cell biology and toxicology. 1998;14:141–145. doi: 10.1023/a:1007486022411. [DOI] [PubMed] [Google Scholar]
  • 24.Yuan J, Lipinski M, Degterev A. Diversity in the mechanisms of neuronal cell death. Neuron. 2003;40:401–413. doi: 10.1016/s0896-6273(03)00601-9. [DOI] [PubMed] [Google Scholar]
  • 25.Fulda S, Debatin KM. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 2006;25:4798–4811. doi: 10.1038/sj.onc.1209608. [DOI] [PubMed] [Google Scholar]
  • 26.Wallach D, Boldin M, Varfolomeev E, Beyaert R, Vandenabeele P, Fiers W. Cell death induction by receptors of the TNF family: towards a molecular understanding. FEBS Lett. 1997;410:96–106. doi: 10.1016/s0014-5793(97)00553-x. [DOI] [PubMed] [Google Scholar]
  • 27.Wallach D. Apoptosis. Placing death under control. Nature. 1997;388:123, 125–126. doi: 10.1038/40516. [DOI] [PubMed] [Google Scholar]
  • 28.Huang Y, Wang KK. The calpain family and human disease. Trends in molecular medicine. 2001;7:355–362. doi: 10.1016/s1471-4914(01)02049-4. [DOI] [PubMed] [Google Scholar]
  • 29.McGinnis KM, Gnegy ME, Park YH, Mukerjee N, Wang KK. Procaspase-3 and poly(ADP)ribose polymerase (PARP) are calpain substrates. Biochemical and biophysical research communications. 1999;263:94–99. doi: 10.1006/bbrc.1999.1315. [DOI] [PubMed] [Google Scholar]
  • 30.Saatman KE, Murai H, Bartus RT, Smith DH, Hayward NJ, Perri BR, et al. Calpain inhibitor AK295 attenuates motor and cognitive deficits following experimental brain injury in the rat. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:3428–3433. doi: 10.1073/pnas.93.8.3428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhang Z, Ottens AK, Shankar S, Kobeissy F, Tei F, Hayes RL, et al. Calpain-mediated collapsin response mediator protein-2 proteolysis following acute traumatic or neurotoxic injury. J Neurotrauma. 2007;24:460–472. doi: 10.1089/neu.2006.0078. [DOI] [PubMed] [Google Scholar]
  • 32.Kobeissy FH, Ottens AK, Zhang Z, Liu MC, Denslow ND, Dave JR, et al. Novel differential neuroproteomics analysis of traumatic brain injury in rats. Mol Cell Proteomics. 2006;5:1887–1898. doi: 10.1074/mcp.M600157-MCP200. [DOI] [PubMed] [Google Scholar]
  • 33.Wang KK, Nath R, Raser KJ, Hajimohammadreza I. Maitotoxin induces calpain activation in SH-SY5Y neuroblastoma cells and cerebrocortical cultures. Arch Biochem Biophys. 1996;331:208–214. doi: 10.1006/abbi.1996.0300. [DOI] [PubMed] [Google Scholar]
  • 34.Pike BR, Zhao X, Newcomb JK, Posmantur RM, Wang KK, Hayes RL. Regional calpain and caspase-3 proteolysis of alpha-spectrin after traumatic brain injury. Neuroreport. 1998;9:2437–2442. doi: 10.1097/00001756-199808030-00002. [DOI] [PubMed] [Google Scholar]
  • 35.Farkas O, Polgar B, Szekeres-Bartho J, Doczi T, Povlishock JT, Buki A. Spectrin breakdown products in the cerebrospinal fluid in severe head injury--preliminary observations. Acta Neurochir (Wien) 2005;147:855–861. doi: 10.1007/s00701-005-0559-6. [DOI] [PubMed] [Google Scholar]
  • 36.O F, B P, Szekeres-Bartho J, Doczi T, Povlishock JT, Buki A. Spectrin breakdown products in the cerebrospinal fluid in severe head injury - preliminary observations. Acta Neurochir (Wien) 2005;147:855–861. doi: 10.1007/s00701-005-0559-6. [DOI] [PubMed] [Google Scholar]
  • 37.Pike BR, Flint J, Dutta S, Johnson E, Wang KK, Hayes RL. Accumulation of non-erythroid alpha II-spectrin and calpain-cleaved alpha II-spectrin breakdown products in cerebrospinal fluid after traumatic brain injury in rats. Journal of neurochemistry. 2001;78:1297–1306. doi: 10.1046/j.1471-4159.2001.00510.x. [DOI] [PubMed] [Google Scholar]
  • 38.Liu HM, Sturner WQ. Extravasation of plasma proteins in brain trauma. Forensic Sci Int. 1988;38:285–295. doi: 10.1016/0379-0738(88)90174-0. [DOI] [PubMed] [Google Scholar]
  • 39.Pineda JA, Wang KK, Hayes RL. Biomarkers of proteolytic damage following traumatic brain injury. Brain Pathol. 2004;14:202–209. doi: 10.1111/j.1750-3639.2004.tb00054.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Squier MK, Miller AC, Malkinson AM, Cohen JJ. Calpain activation in apoptosis. Journal of cellular physiology. 1994;159:229–237. doi: 10.1002/jcp.1041590206. [DOI] [PubMed] [Google Scholar]
  • 41.Wang KK, Ottens A, Haskins W, Liu MC, Kobeissy F, Denslow N, et al. Proteomics studies of traumatic brain injury. Int Rev Neurobiol. 2004;61:215–240. doi: 10.1016/S0074-7742(04)61009-9. [DOI] [PubMed] [Google Scholar]
  • 42.Bernath E, Kupina N, Liu MC, Hayes RL, Meegan C, Wang KK. Elevation of cytoskeletal protein breakdown in aged Wistar rat brain. Neurobiol Aging. 2006;27:624–632. doi: 10.1016/j.neurobiolaging.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • 43.Wang KK, Posmantur R, Nath R, McGinnis K, Whitton M, Talanian RV, et al. Simultaneous degradation of alphaII- and betaII-spectrin by caspase 3 (CPP32) in apoptotic cells. J Biol Chem. 1998;273:22490–22497. doi: 10.1074/jbc.273.35.22490. [DOI] [PubMed] [Google Scholar]
  • 44.Pike BR, Zhao X, Newcomb JK, Wang KK, Posmantur RM, Hayes RL. Temporal relationships between de novo protein synthesis, calpain and caspase 3-like protease activation, and DNA fragmentation during apoptosis in septo-hippocampal cultures. J Neurosci Res. 1998;52:505–520. doi: 10.1002/(SICI)1097-4547(19980601)52:5<505::AID-JNR3>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
  • 45.Pineda JA, Lewis SB, Valadka AB, Papa L, Hannay HJ, Heaton SC, et al. Clinical significance of alphaII-spectrin breakdown products in cerebrospinal fluid after severe traumatic brain injury. J Neurotrauma. 2007;24:354–366. doi: 10.1089/neu.2006.003789. [DOI] [PubMed] [Google Scholar]
  • 46.Siman R, McIntosh TK, Soltesz KM, Chen Z, Neumar RW, Roberts VL. Proteins released from degenerating neurons are surrogate markers for acute brain damage. Neurobiol Dis. 2004;16:311–320. doi: 10.1016/j.nbd.2004.03.016. [DOI] [PubMed] [Google Scholar]
  • 47.Kobeissy FH, Warren MW, Ottens AK, Sadasivan S, Zhang Z, Gold MS, et al. Psychoproteomic Analysis of Rat Cortex Following Acute Methamphetamine Exposure. Journal of proteome research. 2008;7:1971–1983. doi: 10.1021/pr800029h. [DOI] [PubMed] [Google Scholar]
  • 48.Liu MC, Akle V, Zheng W, Dave JR, Tortella FC, Hayes RL, et al. Comparing calpain- and caspase-3-mediated degradation patterns in traumatic brain injury by differential proteome analysis. Biochem J. 2006;394:715–725. doi: 10.1042/BJ20050905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Opii WO, Nukala VN, Sultana R, Pandya JD, Day KM, Merchant ML, et al. Proteomic identification of oxidized mitochondrial proteins following experimental traumatic brain injury. J Neurotrauma. 2007;24:772–789. doi: 10.1089/neu.2006.0229. [DOI] [PubMed] [Google Scholar]
  • 50.Homer BD, Solomon TM, Moeller RW, Mascia A, DeRaleau L, Halkitis PN. Methamphetamine abuse and impairment of social functioning: a review of the underlying neurophysiological causes and behavioral implications. Psychological bulletin. 2008;134:301–310. doi: 10.1037/0033-2909.134.2.301. [DOI] [PubMed] [Google Scholar]
  • 51.Albertson TE, Derlet RW, Van Hoozen BE. Methamphetamine and the expanding complications of amphetamines. The Western journal of medicine. 1999;170:214–219. [PMC free article] [PubMed] [Google Scholar]
  • 52.Scott JC, Woods SP, Matt GE, Meyer RA, Heaton RK, Atkinson JH, et al. Neurocognitive effects of methamphetamine: a critical review and meta-analysis. Neuropsychology review. 2007;17:275–297. doi: 10.1007/s11065-007-9031-0. [DOI] [PubMed] [Google Scholar]
  • 53.Darke S, Kaye S, McKetin R, Duflou J. Major physical and psychological harms of methamphetamine use. Drug and alcohol review. 2008;27:253–262. doi: 10.1080/09595230801923702. [DOI] [PubMed] [Google Scholar]
  • 54.Gonzalez R, Rippeth JD, Carey CL, Heaton RK, Moore DJ, Schweinsburg BC, et al. Neurocognitive performance of methamphetamine users discordant for history of marijuana exposure. Drug and alcohol dependence. 2004;76:181–190. doi: 10.1016/j.drugalcdep.2004.04.014. [DOI] [PubMed] [Google Scholar]
  • 55.Bechara A. Decision making, impulse control and loss of willpower to resist drugs: a neurocognitive perspective. Nature neuroscience. 2005;8:1458–1463. doi: 10.1038/nn1584. [DOI] [PubMed] [Google Scholar]
  • 56.Mandyam CD, Wee S, Eisch AJ, Richardson HN, Koob GF. Methamphetamine self-administration and voluntary exercise have opposing effects on medial prefrontal cortex gliogenesis. J Neurosci. 2007;27:11442–11450. doi: 10.1523/JNEUROSCI.2505-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Schoenbaum G, Saddoris MP, Ramus SJ, Shaham Y, Setlow B. Cocaine-experienced rats exhibit learning deficits in a task sensitive to orbitofrontal cortex lesions. Eur J Neurosci. 2004;19:1997–2002. doi: 10.1111/j.1460-9568.2004.03274.x. [DOI] [PubMed] [Google Scholar]
  • 58.Schoenbaum G, Setlow B, Nugent SL, Saddoris MP, Gallagher M. Lesions of orbitofrontal cortex and basolateral amygdala complex disrupt acquisition of odor-guided discriminations and reversals. Learning & memory (Cold Spring Harbor, NY. 2003;10:129–140. doi: 10.1101/lm.55203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Schoenbaum G, Shaham Y. The role of orbitofrontal cortex in drug addiction: a review of preclinical studies. Biological psychiatry. 2008;63:256–262. doi: 10.1016/j.biopsych.2007.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Caracuel A, Verdejo-Garcia A, Vilar-Lopez R, Perez-Garcia M, Salinas I, Cuberos G, et al. Frontal behavioral and emotional symptoms in Spanish individuals with acquired brain injury and substance use disorders. Arch Clin Neuropsychol. 2008;23:447–454. doi: 10.1016/j.acn.2008.03.004. [DOI] [PubMed] [Google Scholar]
  • 61.Lange RT, Iverson GL, Franzen MD. Comparability of neuropsychological test profiles in patients with chronic substance abuse and mild traumatic brain injury. The Clinical neuropsychologist. 2008;22:209–227. doi: 10.1080/13854040701290062. [DOI] [PubMed] [Google Scholar]
  • 62.Regard M, Knoch D, Gutling E, Landis T. Brain damage and addictive behavior: a neuropsychological and electroencephalogram investigation with pathologic gamblers. Cogn Behav Neurol. 2003;16:47–53. doi: 10.1097/00146965-200303000-00006. [DOI] [PubMed] [Google Scholar]
  • 63.Chang L, Alicata D, Ernst T, Volkow N. Structural and metabolic brain changes in the striatum associated with methamphetamine abuse. Addiction (Abingdon, England) 2007;102(Suppl 1):16–32. doi: 10.1111/j.1360-0443.2006.01782.x. [DOI] [PubMed] [Google Scholar]
  • 64.Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, et al. Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. The American journal of psychiatry. 2001;158:377–382. doi: 10.1176/appi.ajp.158.3.377. [DOI] [PubMed] [Google Scholar]
  • 65.Sekine Y, Ouchi Y, Takei N, Yoshikawa E, Nakamura K, Futatsubashi M, et al. Brain serotonin transporter density and aggression in abstinent methamphetamine abusers. Archives of general psychiatry. 2006;63:90–100. doi: 10.1001/archpsyc.63.1.90. [DOI] [PubMed] [Google Scholar]
  • 66.Ballmaier M, Sowell ER, Thompson PM, Kumar A, Narr KL, Lavretsky H, et al. Mapping brain size and cortical gray matter changes in elderly depression. Biological psychiatry. 2004;55:382–389. doi: 10.1016/j.biopsych.2003.09.004. [DOI] [PubMed] [Google Scholar]
  • 67.Ernst T, Chang L, Leonido-Yee M, Speck O. Evidence for long-term neurotoxicity associated with methamphetamine abuse: A 1H MRS study. Neurology. 2000;54:1344–1349. doi: 10.1212/wnl.54.6.1344. [DOI] [PubMed] [Google Scholar]
  • 68.Sekine Y, Ouchi Y, Sugihara G, Takei N, Yoshikawa E, Nakamura K, et al. Methamphetamine causes microglial activation in the brains of human abusers. J Neurosci. 2008;28:5756–5761. doi: 10.1523/JNEUROSCI.1179-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wilson JM, Kalasinsky KS, Levey AI, Bergeron C, Reiber G, Anthony RM, et al. Striatal dopamine nerve terminal markers in human, chronic methamphetamine users. Nature medicine. 1996;2:699–703. doi: 10.1038/nm0696-699. [DOI] [PubMed] [Google Scholar]
  • 70.Choi HJ, Yoo TM, Chung SY, Yang JS, Kim JI, Ha ES, et al. Methamphetamine-induced apoptosis in a CNS-derived catecholaminergic cell line. Molecules and cells. 2002;13:221–227. [PubMed] [Google Scholar]
  • 71.Davidson C, Gow AJ, Lee TH, Ellinwood EH. Methamphetamine neurotoxicity: necrotic and apoptotic mechanisms and relevance to human abuse and treatment. Brain research. 2001;36:1–22. doi: 10.1016/s0165-0173(01)00054-6. [DOI] [PubMed] [Google Scholar]
  • 72.Bowyer JF, Ali S. High doses of methamphetamine that cause disruption of the blood-brain barrier in limbic regions produce extensive neuronal degeneration in mouse hippocampus. Synapse. 2006;60:521–532. doi: 10.1002/syn.20324. [DOI] [PubMed] [Google Scholar]
  • 73.Ricaurte GA, Guillery RW, Seiden LS, Schuster CR, Moore RY. Dopamine nerve terminal degeneration produced by high doses of methylamphetamine in the rat brain. Brain Res. 1982;235:93–103. doi: 10.1016/0006-8993(82)90198-6. [DOI] [PubMed] [Google Scholar]
  • 74.Axt KJ, Molliver ME. Immunocytochemical evidence for methamphetamine-induced serotonergic axon loss in the rat brain. Synapse. 1991;9:302–313. doi: 10.1002/syn.890090405. [DOI] [PubMed] [Google Scholar]
  • 75.Cadet JL, Brannock C. Free radicals and the pathobiology of brain dopamine systems. Neurochemistry international. 1998;32:117–131. doi: 10.1016/s0197-0186(97)00031-4. [DOI] [PubMed] [Google Scholar]
  • 76.Fumagalli F, Gainetdinov RR, Wang YM, Valenzano KJ, Miller GW, Caron MG. Increased methamphetamine neurotoxicity in heterozygous vesicular monoamine transporter 2 knock-out mice. J Neurosci. 1999;19:2424–2431. doi: 10.1523/JNEUROSCI.19-07-02424.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Deng X, Cai NS, McCoy MT, Chen W, Trush MA, Cadet JL. Methamphetamine induces apoptosis in an immortalized rat striatal cell line by activating the mitochondrial cell death pathway. Neuropharmacology. 2002;42:837–845. doi: 10.1016/s0028-3908(02)00034-5. [DOI] [PubMed] [Google Scholar]
  • 78.Albers DS, Sonsalla PK. Methamphetamine-induced hyperthermia and dopaminergic neurotoxicity in mice: pharmacological profile of protective and nonprotective agents. The Journal of pharmacology and experimental therapeutics. 1995;275:1104–1114. [PubMed] [Google Scholar]
  • 79.Di Monte DA, Royland JE, Jakowec MW, Langston JW. Role of nitric oxide in methamphetamine neurotoxicity: protection by 7-nitroindazole, an inhibitor of neuronal nitric oxide synthase. Journal of neurochemistry. 1996;67:2443–2450. doi: 10.1046/j.1471-4159.1996.67062443.x. [DOI] [PubMed] [Google Scholar]
  • 80.Jayanthi S, Deng X, Noailles PA, Ladenheim B, Cadet JL. Methamphetamine induces neuronal apoptosis via cross-talks between endoplasmic reticulum and mitochondria-dependent death cascades. Faseb J. 2004;18:238–251. doi: 10.1096/fj.03-0295com. [DOI] [PubMed] [Google Scholar]
  • 81.Schmued LC, Bowyer JF. Methamphetamine exposure can produce neuronal degeneration in mouse hippocampal remnants. Brain Res. 1997;759:135–140. doi: 10.1016/s0006-8993(97)00173-x. [DOI] [PubMed] [Google Scholar]
  • 82.Eisch AJ, Marshall JF. Methamphetamine neurotoxicity: dissociation of striatal dopamine terminal damage from parietal cortical cell body injury. Synapse. 1998;30:433–445. doi: 10.1002/(SICI)1098-2396(199812)30:4<433::AID-SYN10>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 83.Ellison G, Switzer RC., 3rd Dissimilar patterns of degeneration in brain following four different addictive stimulants. Neuroreport. 1993;5:17–20. doi: 10.1097/00001756-199310000-00004. [DOI] [PubMed] [Google Scholar]
  • 84.Jayanthi S, Deng X, Bordelon M, McCoy MT, Cadet JL. Methamphetamine causes differential regulation of pro-death and anti-death Bcl-2 genes in the mouse neocortex. Faseb J. 2001;15:1745–1752. doi: 10.1096/fj.01-0025com. [DOI] [PubMed] [Google Scholar]
  • 85.Deng X, Ladenheim B, Jayanthi S, Cadet JL. Methamphetamine administration causes death of dopaminergic neurons in the mouse olfactory bulb. Biological psychiatry. 2007;61:1235–1243. doi: 10.1016/j.biopsych.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 86.Staszewski RD, Yamamoto BK. Methamphetamine-induced spectrin proteolysis in the rat striatum. Journal of neurochemistry. 2006;96:1267–1276. doi: 10.1111/j.1471-4159.2005.03618.x. [DOI] [PubMed] [Google Scholar]
  • 87.Quinton MS, Yamamoto BK. Neurotoxic effects of chronic restraint stress in the striatum of methamphetamine-exposed rats. Psychopharmacology. 2007;193:341–350. doi: 10.1007/s00213-007-0796-x. [DOI] [PubMed] [Google Scholar]
  • 88.Straiko MM, Coolen LM, Zemlan FP, Gudelsky GA. The effect of amphetamine analogs on cleaved microtubule-associated protein-tau formation in the rat brain. Neuroscience. 2007;144:223–231. doi: 10.1016/j.neuroscience.2006.08.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kosik KS, Finch EA. MAP2 and tau segregate into dendritic and axonal domains after the elaboration of morphologically distinct neurites: an immunocytochemical study of cultured rat cerebrum. J Neurosci. 1987;7:3142–3153. doi: 10.1523/JNEUROSCI.07-10-03142.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Warren MW, Larner SF, Kobeissy FH, Brezing CA, Jeung JA, Hayes RL, et al. Calpain and caspase proteolytic markers co-localize with rat cortical neurons after exposure to methamphetamine and MDMA. Acta neuropathologica. 2007;114:277–286. doi: 10.1007/s00401-007-0259-9. [DOI] [PubMed] [Google Scholar]
  • 91.Warren MW, Kobeissy FH, Liu MC, Hayes RL, Gold MS, Wang KK. Concurrent calpain and caspase-3 mediated proteolysis of alpha II-spectrin and tau in rat brain after methamphetamine exposure: a similar profile to traumatic brain injury. Life Sci. 2005;78:301–309. doi: 10.1016/j.lfs.2005.04.058. [DOI] [PubMed] [Google Scholar]
  • 92.Nath R, Raser KJ, Stafford D, Hajimohammadreza I, Posner A, Allen H, et al. Non-erythroid alpha-spectrin breakdown by calpain and interleukin 1 beta-converting-enzyme-like protease(s) in apoptotic cells: contributory roles of both protease families in neuronal apoptosis. Biochem J. 1996;319(Pt 3):683–690. doi: 10.1042/bj3190683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wallace TL, Vorhees CV, Zemlan FP, Gudelsky GA. Methamphetamine enhances the cleavage of the cytoskeletal protein tau in the rat brain. Neuroscience. 2003;116:1063–1068. doi: 10.1016/s0306-4522(02)00795-9. [DOI] [PubMed] [Google Scholar]
  • 94.Marcus K, Schmidt O, Schaefer H, Hamacher M, van Hall A, Meyer HE. Proteomics--application to the brain. Int Rev Neurobiol. 2004;61:285–311. doi: 10.1016/S0074-7742(04)61011-7. [DOI] [PubMed] [Google Scholar]
  • 95.Sokolov BP, Cadet JL. Methamphetamine causes alterations in the MAP kinase-related pathways in the brains of mice that display increased aggressiveness. Neuropsychopharmacology. 2006;31:956–966. doi: 10.1038/sj.npp.1300891. [DOI] [PubMed] [Google Scholar]
  • 96.Iwazaki T, McGregor IS, Matsumoto I. Protein expression profile in the striatum of acute methamphetamine-treated rats. Brain Res. 2006;1097:19–25. doi: 10.1016/j.brainres.2006.04.052. [DOI] [PubMed] [Google Scholar]
  • 97.Iwazaki T, McGregor IS, Matsumoto I. Protein expression profile in the striatum of rats with methamphetamine-induced behavioral sensitization. Proteomics. 2007;7:1131–1139. doi: 10.1002/pmic.200600595. [DOI] [PubMed] [Google Scholar]
  • 98.Yu CG, Geddes JW. Sustained calpain inhibition improves locomotor function and tissue sparing following contusive spinal cord injury. Neurochem Res. 2007;32:2046–2053. doi: 10.1007/s11064-007-9347-4. [DOI] [PubMed] [Google Scholar]
  • 99.Ai J, Liu E, Wang J, Chen Y, Yu J, Baker AJ. Calpain inhibitor MDL-28170 reduces the functional and structural deterioration of corpus callosum following fluid percussion injury. J Neurotrauma. 2007;24:960–978. doi: 10.1089/neu.2006.0224. [DOI] [PubMed] [Google Scholar]
  • 100.Nimmrich V, Szabo R, Nyakas C, Granic I, Reymann KG, Schroder UH, et al. Inhibition of Calpain Prevents N-Methyl-D-aspartate-Induced Degeneration of the Nucleus Basalis and Associated Behavioral Dysfunction. The Journal of pharmacology and experimental therapeutics. 2008;327:343–352. doi: 10.1124/jpet.108.142679. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES