<<

A Dissertation entitled The Contribution of Ammonia to by Laura E. Halpin Submitted to the Graduate Faculty as partial fulfillment of the requirements for the

Doctor of Philosophy Degree in Biomedical Sciences

______Bryan K. Yamamoto, Ph.D., Committee Chair

______Andrew Beavis, Ph.D., Committee Member

______Nicolas Chiaia, Ph.D., Committee Member

______David Giovannucci, Ph.D., Committee Member

______Robert Mrak, M.D., Ph.D., Committee Member

______Patricia R. Komuniecki, Ph.D., Dean College of Graduate Studies

The University of Toledo August 2013

Copyright 2013, Laura E. Halpin This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author.

An Abstract of The Contribution of Ammonia to Methamphetamine Neurotoxicity by Laura E. Halpin Submitted to the Graduate Faculty as partial fulfillment of the requirements for the Doctor of Philosophy Degree in Biomedical Sciences The University of Toledo August 2013

Methamphetamine neurotoxicity results from multiple interacting mechanisms of damage, yet the contribution of peripheral organ damage and ammonia to the long-term monoaminergic terminal damage seen after drug exposure has not been previously investigated. We hypothesized that methamphetamine caused acute liver damage and increases in ammonia which contribute to the long-term striatal neurotoxicity produced by the drug. These effects were investigated using a binge dosing paradigm of

methamphetamine in Male Sprague-Dawley rats. Methamphetamine was shown to

produce significant hyperthermia-dependent liver damage that contributed to increases in

plasma and ammonia concentrations. These increases in ammonia were also shown

to contribute significantly to the long term striatal dopamine and serotonin terminal

damage produced by the drug. Ammonia was demonstrated to mediate this neurotoxicity

via increases in glutamate and subsequent excitotoxicity. These findings identify liver

damage and ammonia as novel mediators of the neurotoxicity of methamphetamine and

highlight the importance of considering peripheral organ function in the study of neuronal

damage. iii

For my teachers, professors, mentors, and patients. The opportunity to learn from you has been and always will be the greatest of privileges.

iv

Acknowledgements

I would like to thank my dissertation advisor, Bryan K. Yamamoto, PhD. for helping me design and carry out the experiments necessary to complete this work.

Beyond the completion of these experiments, I would also like to thank him for helping me develop the presentation, decision-making and communication skills needed to prepare me to be an effective scientist. I would also like to recognize the members of the

Yamamoto lab for teaching me the techniques needed to complete these experiments and for their support and feedback though out the past few years.

In addition to my colleagues in the lab, I would also like to thank my friends and family for their support and encouragement as I have been working toward completing this degree.

v

Contents

Abstract iii

Acknowledgements v

Contents vi

List of Figures x

List of Abbreviations xii

1 General Introduction to Methamphetamine Neurotoxicity, Liver Damage and Ammonia 1

1.1 Methamphetamine Use 1

1.2 Acute Effects of METH on release 2

1.3 Acute Behavioral Effects of METH Exposure 3

1.4 Persistent Dopamine and Serotonin Terminal Damage 4

1.5 Neuronal Death 5

1.6 Long Term Behavioral Consequences of METH Exposure 6

1.7 Mechanisms of Neurotoxicity 8

1.7.1 Excitotoxicity 8

1.7.2 Oxidative Stress 9

1.7.3 Altered and Mitochondrial Damage 10 vi

1.8 Emerging Mechanisms of Toxicity 11

1.8.1 Inflammation 11

1.8.2 Brain Barrier Damage 12

1.8.3 Peripheral Organ Damage 13

1.9 METH-Induced Liver Damage 14

1.10 Ammonia and Neurotoxicity 14

1.11 Summary of Past Findings and Gaps in Knowledge 15

1.12 Hypotheses 17

2 Methamphetamine causes Acute Hyperthermia-Dependent Liver Damage 18

2.1 Introduction 18

2.2 Methods 20

Rat Treatment 20

Transmission Electron Microscopy 20

Hematoxylin and Eosin Staining 21

Asparate Aminotransferase and Alanine Aminotransferase Determination 21

Statistical Methods 22

2.3 Results 22

METH and Liver Damage 22

METH and Ultrastructural Liver Damage 23

Environmental Cooling and Methamphetamine-Induced Hyperthermia 23

Hyperthermia and METH-Induced Changes in Liver Morphology 24

vii

Hyperthermia and METH-Induced Hepatocellular Damage 24

2.4 Discussion 32

3 Peripheral Ammonia Mediates Methamphetamine Neurotoxicity 37

3.1 Introduction 37

3.2 Methods 39

Rat Treatment 39

Plasma Ammonia Determination 40

In-Vivo Microdialysis 40

Brain Ammonia Determination 41

HPLC Analysis of Monoamine Tissue Content 42

Western Blot for DAT Immunoreactivity 42

Mass Spectroscopy 43

Statistical Analysis 43

3.3 Results 44

METH and plasma and ammonia 44

Effect of Hyperthermia on METH-Induced Increases in Plasma Ammonia 44

METH and Brain Ammonia 45

METH and Increased Peripheral Ammonia Excretion 45

Local Administration of METH and Ammonia 48

3.4 Discussion 57

4 Contribution of Ammonia to Methamphetamine-Induced Increases in

Extracellular Glutamate and Excitotoxicity 62

4.1 Introduction 62 viii

4.2 Methods 67

Rat Treatment 67 HPLC Analysis of Monoamine Tissue Content 67 HPLC Analysis of Glutamate 68 In-Vivo Microdialysis 68 Spectrin Breakdown Product (SBP) Analysis 69 Glutamine Synthetase Level Measurement 69 Glutamine Synthetase Activity Assay 70 Statistical Analysis 71 4.3 Results 71

Effect of AMPA Receptor Antagonism on Local Administration of METH and Ammonia 71

Effect of Locally Administered Ammonia on Extracellular Glutamate 72

Effect of Ammonia on Systemic-METH-Induced Increases in Extracellular Glutamate 73

Effect of Ammonia on -Mediated Spectrin Proteolysis 73

Effect of MET H on Glutamine Synthetase Protein Levels 74

Effect of METH on Glutamine Synthetase Activity 74

4.4 Discussion 82

5 General Discussion of Findings 88

5.1 Summary of Findings 88

5.2 General Discussion 90

References 96

ix

List of Figures

1.1 Model of METH Neurotoxicity 16

2.1 METH causes Hepatocellular Damage Visible by H&E Staining 26

2.2 Serum AST and ALT after METH Treatment 27

2.3 Ultrastructural Changes in Liver 24 Hours After Methamphetamine Exposure 28

2.4 Effect of Environmental Cooling on Methamphetamine-Induced Hyperthermia 29

2.5 Effect of Hyperthermia on Methamphetamine-Induced Changes in Liver Morphology 30

2.6 Effect of Hyperthermia on Methamphetamine-Induced Increases in Serum AST and ALT 31

3.1 METH and Plasma Ammonia 50

3.2 Effect of Hyperthermia on Methamphetamine-Induced Persistent Increases in Plasma Ammonia 51

3.3 METH and Brain Ammonia 52

3.4 Hyperthermia after METH Treatment 53

3.5 Effect of Lactulose on METH-induced Monoamine Depletions 54

3.6 Effect of Lactulose on METH-induced Reductions in DAT Immunoreactivity 55

3.7 Effects of Local Administration of Ammonia and METH into the Striatum 56

4.1 The Dopamine and Serotonin Terminal Damage Produced by the Combination of NH3 and METH Requires AMPA Receptor Activation 76

x

4.2 Effect of Local Ammonia Administration on Extracellular Glutamate Levels 77

4.3 Effect of Ammonia on Systemic METH-induced Increases in Extracellular Glutamate 78

4.4 Effect of Ammonia on METH-induced Increases in Calpain-Mediated Spectrin Proteolysis 79

4.5 Effect of METH on Glutamine Synthetase Expression 80

4.6 Effect of METH on Glutamine Synthetase Activity 81

5.1 Summary of Findings 89

xi

List of Abbreviations

5HT Serotonin aCSF Artificial Cerebrospinal Fluid

AIDS Acquired Immunodeficiency Syndrome

ALT Alanine Aminotransferase

AMPA 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid

AST Aspartate Aminotransferase

BAS Bioanalytical Systems Inc.

Bcl B-cell lymphoma

BD Becton-Dickinson

BMDS Bio Medic Data Systems

CNS

COX Cyclooxygenase

DAT Dopamine Transporter

EAAT Excitatory Amino Acid Transporter

FDA Food and Drug Adminstration

GABA Gamma-aminobutyric Acid

GLT-1 -1

GLAST Glutamate Aspartate Transporter

xii

GS Glutamine Synthetase

GYKI 52466 4-(8-Methyl-9H-1,3-dioxolo[4,5-h][2,3]benzodiazepin-5-yl)- benzenamine dihydrochloride

H&E Hematoxylin and Eosin

HRP Horseradish Peroxidase

IL Interleukin

Lac Lactulose

LDS Dodecyl Sulfate

MWCO Molecular Weight Cut-Off

OD Outer Diameter

PE Polyethylene

PVDF Polyvinylidene Difluoride

Sal Saline

SAMSHA Substance Abuse and Mental Health Services Administration

SERT Serotonin Transporter

SEM Standard Error of the Mean

METH Methamphetamine

MDMA 3,4-methylenedioxymethamphetamine

NMDA N-methyl-D-aspartate

TNF

TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling

UNODC United Nations Office on Drugs and Crime

Veh Vehicle

xiii

Chapter 1:

General Introduction to Methamphetamine Neurotoxicity,

Liver Damage and Ammonia

1.1 Methamphetamine Use

Methamphetamine (METH) is a widely abused psychostimulant. The synthetic

substituted-amphetamine can be inexpensively manufactured from household ingredients

contributing to the significant prevalence of its use. In 2011, the lifetime prevalence of

METH use was 4.6% and prevalence of current METH use is 0.2% of the population.

Throughout 2011, over 130,000 people tried METH for the first time and the mean age of

first-time METH users was 17.8 years old (SAMSHA, 2012b). METH exposure is also a

clinically relevant concern as nearly 150,000 people presented to emergency departments

in 2010 for complaints associated with amphetamine-type stimulant use (SAMSHA,

2012a). Beyond their highly prevalent use within the United States, amphetamine-type

stimulants are the second most-widely used class of drugs worldwide, second to cannabis

(UNODC, 2012). The prevalence of METH use is greater than 1% of the population

aged 15-64 throughout other North American countries, Asia, and Australia, as well as in

1

some regions of Europe and Africa, where the prevalence of METH-use is increasing

(UNODC, 2012).

Since it was first synthesized in 1919 and made commercially available during the middle of the twentieth century, METH has been used for many indications including asthma treatment, to heighten focus and alertness, and weight loss. The drug has also been used extensively by the military to increase alertness, reduce fatigue and decrease appetite (Vearrier et al., 2012). Soon after METH was commercialized, the addictive and other adverse effects became apparent. The Food and Drug Adminstration (FDA) first restricted the distribution of METH in 1959, requiring prescriptions for patients to receive METH-containing inhalers. In 1970, the drug was given Schedule II classification under the Comprehensive Drug Abuse Prevention and Control Act of 1970

(Rasmussen, 2008). Today, the drug is manufactured under the name desoxyn and its

FDA approved indications include attention deficit hyperactivity disorder and for the short-term, adjunctive treatment of exogenous obesity. Currently, nearly all recreational

METH users obtain illicitly manufactured drug that was prepared in homemade laboratories (Gonzales et al., 2010).

1.2 Acute Effects of METH on Neurotransmitter Release

METH causes acute increases in synaptic dopamine, serotonin and norepinephrine. These increases result from the binding of the drug to the dopamine transporter (DAT), serotonin transporter (SERT), and norepinephrine (NET). In addition to binding these transporters, METH is a substrate that is transported into the axon terminal (Rothman and Baumann, 2003). Following intracellular transport, amphetamines contribute to vesicle disruption, causing increased cytoplasmic dopamine and serotonin

2

(Sulzer and Rayport, 1990). Subsequent to increases in cytoplasmic dopamine and

serotonin, reversal of the dopamine and serotonin transporters causes significant, action

potential-independent neurotransmitter release (Sulzer et al., 1995). In addition to

reversal, acute decreases in neurotransmitter transporter function also contribute to

increases in dopamine (Fleckenstein et al., 1997a). Secondary to increases in dopamine,

METH causes acute increases in glutamate as a result of D1 dopamine receptor-mediated disinhibition of corticostriatal glutamate release (Nash and Yamamoto, 1992, Mark et al.,

2004).

1.3 Acute Behavioral Effects of METH Exposure

The acute behavioral effects of METH exposure result primarily from the effect of the drug on monoamine concentrations. Increases in norepinephrine during drug exposure are associated with arousal, reduced fatigue and appetite, as well as increases in heart rate, respiration rate, blood pressure and pupil dilation (Shappell et al., 1996, Berridge,

2006, Mendelson et al., 2006). METH-induced increases in dopamine contribute to the euphoria, paranoia, auditory and visual hallucinations, improved attention, manic symptoms, and psychomotor activation produced by the drug (Bell, 1973, Perez-Reyes et al., 1991, Shappell et al., 1996, Mendelson et al., 2006). Depending on the receptor subtypes that are activated, acute increases in serotonin can contribute to either relaxation and increased social interaction, or anxiety (Martin et al., 1971, Harris et al., 2003,

Mendelson et al., 2006). The drug also produces significant increases in body temperature in the range of 39-42 °C as a result of both central effects within the hypothalamus involving serotonin and norepinephrine, and peripheral vasoconstriction, hyperlocomotion and altered metabolism (Wang et al., 1990, Brown et al., 2007, Sprague

3

et al., 2007, Benamar et al., 2008). Chronic unpredictable stress, which produces

significant increases in corticosterone, has also been shown to potentiate METH-induced

hyperthermia in a 5HT2 receptor dependent manner, further highlighting the role of

serotonin in METH-induced hyperthermia (Doyle and Yamamoto, 2010). METH-induced hyperthermia is associated with the neurotoxicity produced by the drug. Blockade of

drug induced increases in hyperthermia during METH-treatment protects against the long-term monoaminergic terminal damage produced by the drug, although hyperthermia alone is not adequate to produce this terminal damage (Bowyer et al., 1992, Bowyer et al., 1994, Albers and Sonsalla, 1995). Accordingly, hyperthermia appears to potentiate

many of the mechanisms contributing to METH-induced neurotoxicity including action of the drug at the dopamine and serotonin transporters, as well as increases in oxidative

stress and brain edema (Fleckenstein et al., 1997b, Haughey et al., 2000, Xie et al., 2000,

Kiyatkin and Sharma, 2009). METH-induced hyperthermia may also contribute tissue

damage produced by the drug as significant increases in core temperature alone can

damage the liver and other peripheral organs.

1.4 Persistent Dopamine and Serotonin Terminal Damage

Subsequent to the acute effects of exposure, METH produces long-term damage

to dopaminergic and serotonergic axon terminals in the striatum, hippocampus and

prefrontal cortex (Ricaurte et al., 1980, Wagner et al., 1980, Seiden et al., 1988). This

damage has been shown to persist for at least 2 years after drug exposure in rodents, non-

human primates and humans (Seiden et al., 1988, Woolverton et al., 1989, McCann et al.,

1998, Volkow et al., 2001a, McCann et al., 2005) Neurochemical markers of this

toxicity include decreases in dopamine and serotonin tissue content, decreases in DAT

4

and SERT expression coupled with decreases in transporter Vmax without changes in Km,

and decreases in the expression of tyrosine and tryptophan hydroxylase, the rate limiting

enzymes for dopamine and serotonin, respectively (Hotchkiss and Gibb, 1980, Wagner et

al., 1980, Ricaurte et al., 1982, Ricaurte et al., 1985, Commins et al., 1987) . Beyond

changes in tissue content and neurotransmitter specific , morphological changes

indicative of axon terminal damage have been reported including the presence of swollen,

distorted nerve terminals and positive Fink-Heimer staining, as well as edematous and degenerative changes identified by electron microscopy (Lorez, 1981, Ricaurte et al.,

1982, Sharma and Kiyatkin, 2009). Recent METH self-administration studies with

longer access paradigms further confirm the terminal damage produced by substituted

amphetamine exposure and show decreases in DAT and SERT and tyrosine hydroxylase,

as well as decreases in tissue content, all indicative of drug-induced monoaminergic

terminal damage (Krasnova et al., 2010, McFadden et al., 2012a, McFadden et al.,

2012b) .

1.5 Neuronal

Though not as extensively studied, there is also supporting evidence that METH

may produce cell death, in addition to damaging dopamine and serotonin terminals.

Increases in terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), a

marker for apoptotic cell death, has been reported after exposure to METH in the

prefrontal cortex and striatum (Kadota and Kadota, 2004, Cadet et al., 2005, Zhu et al.,

2006b). This cell death has been identified in different subpopulations of GABA-

interneurons. Within the hippocampus, cell death of calbindin-containing GABA

interneurons has been demonstrated, and in the striatum, METH-contributes to the death

5

of parvalbumin containing GABA interneurons (Zhu et al., 2006a, Kuczenski et al.,

2007). Mitochondrial damage and endoplasmic reticulum stress have been associated

with this METH-induced (Cadet et al., 2005). Specifically, the drug has been

shown to contribute to apoptosis through increases in -3 activity and the Fas/FasL

cell death pathways (Jayanthi et al., 2005). The drug also produces DNA damage and

alterations in the expression of Bcl-2 related genes, which may contribute to GABA

interneuron cell death (Jayanthi et al., 2001, Jeng et al., 2006).

1.6 Long Term Behavioral Consequences of METH Exposure

Repeated METH use contributes to abuse and dependence on the drug. In

addition to intense craving, withdrawal from the drug is associated with disturbed sleep,

anxiety, depressed mood, agitation and reduced energy (McGregor et al., 2005).

Exposure to the drug is also associated with numerous long-term neuropsychological

impairments associated with the persistent and significant dopamine and serotonin terminal damage seen after drug exposure. These persistent neuropsychological effects include impaired impulse control, working memory, decision making, attention, and motor coordination (Rogers et al., 1999, Volkow et al., 2001b, Simon et al., 2002, Clark et al., 2006, Johanson et al., 2006). Abstinent patients who were previously dependent on the drug also have a high potential for relapse and impairments in cortex-mediated

decision making which may be the result of decreases in dopamine activity (Paulus et al.,

2005, Wang et al., 2012). While each of these impairments are exhibited by numerous

METH-dependent subjects, it is also worth noting that the presence of these symptoms

before METH use is unknown and may precede and contribute to METH abuse and

dependence.

6

The long-term damage produced by METH may also represent a vulnerability factor and contribute to protracted psychosis and psychotic disorders, in addition to the acute psychotic effects of the drugs (Flaum and Schultz, 1996). Abuse of amphetamines is associated with a significant increase in the prevalence of psychosis and this association is especially strong in the context of METH use and amphetamine abuse in the mid-late teen years (Harris and Batki, 2000, McKetin et al., 2006). Psychosis presenting subsequent to amphetamine-type stimulant use or in the context of schizophrenia share many common features and have been suggested to share a common neural substrate (Hermens et al., 2009). Of these, substituted-amphetamine induced alterations to dopamine, GABA and glutamate neurotransmitter systems are likely the most significant contributors to protracted psychosis and vulnerability to psychotic disorders. Hyperactivity of the mesolimbic dopamine pathway is strongly associated with the positive symptoms of schizophrenia and a target of many antipsychotic drugs. At first glance, the hyperactive mesolimbic dopamine activity postulated to contribute to psychosis appears to be paradoxically associated with the dopaminergic depletions produced by METH; however mesolimbic dopamine projections are relatively spared from METH-induced dopamine terminal damage (Granado et al., 2010). It is also worth noting that the mesocortical dopamine depletions produced by METH may contribute to the negative, cognitive and affective symptoms associated with schizophrenia.

Beyond changes in dopamine function, the alteration of glutamate and GABA neurotransmission produced by METH represent mechanisms by which the drug contributes to psychosis. Sensory gating, which is important for modulating the perceptual distortions contributing to psychosis, has been shown to be affected by METH

7

(Hadamitzky et al., 2011). METH-induced decreases in sensorimotor gating have been attributed to alterations in both glutamate and downstream GABA neurotransmission in cortico-striatal-thalamic-cortical sensory filtering circuits (Arai et al., 2008, Mizoguchi et al., 2009).

1.7 Mechanisms of Neurotoxicity

1.7.1 Excitotoxicity

Excitotoxicity is a mechanism of neurotoxicity that has been implicated in the monoaminergic terminal damage produced by METH. Excitotoxicity results from the activation of calcium permeable glutamate receptors and excessive influx of calcium leading to activation, mitochondrial damage and nitric oxide formation (Choi,

1994). METH has been demonstrated to produce significant increases in extracellular glutamate within the striatum as a result of dopamine-mediated disinhibition of glutamate release from corticostriatal projections (Nash and Yamamoto, 1992, Mark et al., 2004).

Subsequent to increases in glutamate, calcium permeable AMPA receptors located on dopamine and serotonin terminals in the striatum may be activated contributing to calcium influx (Keefe et al., 1993, Betarbet and Greenamyre, 1999, Hernandez et al.,

2003). This is supported by findings that METH-induced activation of calpain, a calcium activated protease and marker of excitotoxic terminal damage, is decreased when AMPA receptor activation is prevented in the context of METH treatment (Staszewski and

Yamamoto, 2006). NMDA and metabotropic receptor antagonists have also been shown to protect against the damage produced by METH, suggesting that the activation of multiple types of glutamate receptors may contribute to the neurotoxicity of METH

(Farfel et al., 1992, Battaglia et al., 2002). In addition to increases in protease activity,

8

increases in glutamate and excitotoxicity have been linked to free radical and nitric oxide

formation (Dawson and Dawson, 1996, Deng and Cadet, 1999, Battaglia et al., 2002) and

the resultant oxidative stress represents a mechanism by which METH-induced increases in dopamine and glutamate synergize to cause monoaminergic terminal damage (Tata and

Yamamoto, 2007)

1.7.2 Oxidative Stress

Oxidative stress is another mechanism by which METH causes neuronal damage.

Reactive species are formed subsequent to substituted amphetamine exposure

through numerous mechanisms. Acute increases in dopamine contribute to oxidatively

damaged axon terminals after METH exposure (Schmidt et al., 1985). Direct oxidation

of dopamine leading to quinone formation, iron-catalyzed dopamine metabolism via the

Fenton Reaction, and metabolism of dopamine by monoamine oxidase-A contribute to

superoxide and hydrogen peroxide production (Graham, 1978, Yamamoto and Zhu, 1998,

LaVoie and Hastings, 1999). In addition to dopamine, METH treatment leads to

oxidative stress via increases in reactive nitrogen species resulting from nitric oxide

synthase activity (Imam et al., 2000) Increases in free radical formation and oxidative

stress after METH are further supported by depletions in antioxidant enzymes after drug

exposure (Jayanthi et al., 1998, Jayanthi et al., 1999). Subsequent to METH exposure,

increased oxidative damage including lipid peroxidation and protein carbonyl formation

has been shown (Yamamoto and Zhu, 1998, Gluck et al., 2001). Specific nitration and

nitrosylation of proteins important for monoamine synthesis and release including

VMAT-2 and tyrosine and tryptophan hydroxylase has also been reported (Kuhn et al.,

1999, Kuhn and Geddes, 1999, Eyerman and Yamamoto, 2007). The oxidative

9

modification of these proteins is significant as it disrupts their function and contributes to

their degradation, thereby contributing to amphetamine neurotoxicity. Antioxidant

treatments have been shown to be neuroprotective for the damage produced by METH,

highlighting the significant contribution of oxidative stress to the neurotoxicity of

substituted amphetamines (Fukami et al., 2004)

1.7.3 Altered Metabolism and Mitochondrial Damage

METH also contributes to monoaminergic terminal damage though altered energy

metabolism, including mitochondrial damage. During exposure to METH, there is a high

demand for energy and acute increases in energy metabolism in brain regions where the

drugs increase neuronal activity (Pontieri et al., 1990). Soon after the excessive increase

in energy metabolism reported during drug exposure, there is evidence of compromised

energy metabolism and depleted energy stores (Chan et al., 1994, Huang et al., 1999).

Damage to mitochondria and specific complexes of the electron transport chain including

complex II-III and IV contribute to these decreases in energy metabolism (Burrows et al.,

2000a). Glutamate and nitric oxide-dependent decreases in complex II have also been shown to play a significant role in METH neurotoxicity during psychostimulant exposure, as well as excitotoxically-mediated mitochondrial damage (Burrows et al.,

2000b, Brown et al., 2005). The dysfunctional energy metabolism observed subsequent

to METH exposure is believed to result from the increased neuronal energy requirements

during drug exposure, as well as mitochondrial damage from glutamate-mediated excitotoxicity (Brown et al., 2005). When additional energy sources are supplied to during drug exposure or increases in glutamate are blocked, excitotoxic mitochondria damage, as well as dopamine and serotonin terminal damage is prevented,

10

supporting a significant role for these effects in axon terminal damage (Stephans et al.,

1998).

1.8 Emerging Mechanisms of Toxicity

Numerous, complex interacting mechanisms have been identified as significant

contributors to the neurotoxic effects of METH, yet many studies only examine these

effects within neurons. Because the drug is always administered systemically and affects

other cells within the brain, as well as peripheral organ function, it is important to

consider how METH produces non-neuronal damage and how this damage may

contribute to the neurotoxicity of the drug. Identification and understanding of these non-

neuronal contributors is important because they have the potential to significantly

contribute to the excitotoxicity, oxidative stress and metabolic compromise produced by

METH. Furthermore, elucidating these non-neuronal mediators of toxicity may reveal novel targets for the treatment of the neurotoxicity produced by METH.

1.8.1 Inflammation

In addition to increasing neuronal activity, METH has been shown to increase microglial activity, which can contribute significantly to neuroinflammation (Escubedo et al., 1998, Thomas et al., 2004a). METH-induced increases in both dopamine and glutamate may contribute to microglial activation and inflammation via the formation of dopamine quinones and/or NMDA receptor activation (Thomas and Kuhn, 2005b, Kuhn et al., 2006). Microglial activation increases pro-inflammatory cytokines and is associated with the neuronal terminal damage produced by METH (Thomas et al.,

2004b). In the context of METH treatment, the expression of IL-1β, IL-6, and TNF-α have all been shown to increase in the striatum. Conversely, knockout of IL-6 production

11 attenuates the damage produced by the drug (Ladenheim et al., 2000, Sriram et al., 2006).

Reduction of TNF-α and direct inhibition of microglial activation, however, is not protective, suggesting that inflammation plays a complex role in neurotoxicity and may both contribute to and protect against monoaminergic terminal damage (Nakajima et al.,

2004, Sriram et al., 2006). Although METH appears to activate microglia and increase cytokine formation, the degree to which this inflammation contributes to the neurotoxicity of the METH alone is unclear. It has been suggested that increases in cytokine formation may contribute to the neurotoxicity of METH as a result of increases in cyclooxygenase (COX) activity. Furthermore, the peroxidase activity resulting from

COX activity may contribute to the oxidative damage produced by the drug (Thomas and

Kuhn, 2005a). However, the role of COX activity in the monoaminergic terminal damage produced by METH remains unclear as only knockout, but not pharmacologic inhibition of COX activity is protective (Thomas and Kuhn, 2005a). In fact, selective inhibition of COX-2 has been shown to potentiate METH-induced dopaminergic toxicity

(Zhang et al., 2007). Accordingly, further studies are needed to understand the contribution of inflammation to METH neurotoxicity.

1.8.2 Blood-Brain Barrier Damage

Another non-neuronal consequence of METH exposure is damage to the blood brain barrier. The blood brain barrier is responsible for separating the neuronal parenchyma from the arterial blood supply of the brain and is comprised of endothelial cells with tight junctions, pericytes and astrocytic end feet (Saunders et al., 2008).

METH alone produces a transient increase in blood-brain barrier permeability, allowing for the influx of molecules into the brain that would otherwise be limited by this

12

functional barrier (Bowyer and Ali, 2006, Sharma and Ali, 2006, Silva et al., 2010). The

mechanism by which the drug contributes to this damage has not been fully elucidated;

however hyperthermia, oxidative stress, increases in nitric oxide and protease activity

targeting endothelial tight junctions have all been implicated in increases in blood-brain barrier permeability (Bowyer et al., 2008, Ramirez et al., 2009, Martins et al., 2011,

Martins et al., 2013). Accordingly, METH may be causing blood-brain barrier damage though many of the same mechanisms by which the drug causes neuronal damage.

Increases in blood brain barrier permeability may also contribute to the neurotoxicity of

METH. Alterations in blood-brain barrier function have been shown to correlate with hypoxia as well as depletions in tyrosine hydroxlase (Kousik et al., 2011). METH- induced increases in blood-brain barrier permeability may also contribute to increased neurologic involvement in infections including NeuroAIDS, herpes simplex-2, and cryptococcal meningitis (Cisneros and Ghorpade, 2012, Conant et al., 2012, Valencia et al., 2012, Eugenin et al., 2013).

1.8.3 Peripheral Organ Damage

Although studies of METH focus on the central nervous system effects of the drug, METH has been reported to cause significant peripheral organ damage (Smith and

Fischer, 1970). Clinical reports suggest that the cardiac, renal, muscular, and hepatic organ systems may be especially susceptible to damage by METH (Jones et al., 1994,

Kamijo et al., 2002, Wijetunga et al., 2003, Ago et al., 2006). METH is metabolized in the liver by the cytochrome p450 2D6 system and excreted by the kidneys (Caldwell et al., 1972, Kim et al., 2004). In fact, the highest and longest lasting concentrations of

METH after drug exposure have been seen in the liver (Volkow et al., 2010). Further

13

understanding of the peripheral organ damage produced by METH is important because

the brain is highly dependent on peripheral organ function. Therefore, damage to

peripheral organs may contribute significantly to the established neurotoxicity of the drug

and represents a novel target for interventions to treat the neurotoxicity of METH and its

neuropsychiatric consequences.

1.9 METH-Induced Liver damage

Of the organ systems reported to be damaged by METH, we have focused on the

liver because of the significant concentrations of METH measured there, as well as the

fact that liver damage, alone in other contexts, is associated with neurotoxicity (Felipo

and Butterworth, 2002, Volkow et al., 2010). The liver is important for the metabolism

and detoxification of chemicals in the body, as well as protein synthesis, bile production,

and energy storage. METH has the potential to damage the liver though many direct or

indirect mechanisms. The metabolism of METH via the cytochrome p450 system may

result in oxidative and mitochondrial damage to hepatocytes (Pourahmad et al., 2010,

Letelier et al., 2011). Hepatocytes may also be damaged by extra-hepatic effects of

METH, including hyperthermia or changes in cardiovascular function (Kew et al., 1970,

Skibba et al., 1989, Wang et al., 1990, Chen, 2007).

1.10 Ammonia and Neurotoxicity

The significance of drug-mediated peripheral organ damage is highlighted by the dependence of the brain on peripheral organ function and that alterations in peripheral physiology can contribute to neuronal pathology. Clinically, the neurologic symptoms that present in the context of liver failure are termed hepatic encephalopathy. A key feature of the pathology that contributes to neuronal damage is ammonia (Al Sibae and

14

McGuire, 2009). Liver damage is associated with increases in ammonia as the liver

metabolizes ammonia to urea via the urea cycle so it can be efficiently excreted via the

kidneys (Felipo and Butterworth, 2002). Similar to METH, ammonia alone has been shown to cause neurotoxicity via excitotoxicity, oxidative damage, and metabolic compromise (Hawkins et al., 1973, Kosenko et al., 1995, Kosenko et al., 1997, Saez et al., 1999, Llansola et al., 2007). Accordingly, ammonia could be a peripherally derived mediator of METH neurotoxicity which may contribute synergistically to the neurotoxic mechanisms that were classically thought to result from the direct neuronal effects of the drug.

1.11 Summary of Past Findings and Gaps in Knowledge

As reviewed above, METH produces long-term dopamine and serotonin terminal

damage and numerous mechanisms by which the drug produces this damage have been

identified. These include oxidative stress, mitochondrial damage and excitotoxicity

(Figure 1.1). However, it remains to be established if the drug contributes to peripheral

organ damage and whether peripheral organ damage contributes to the neurotoxicity of

the drug. Furthermore, the potential role played by ammonia in the neurotoxicity of

METH has not been examined. Ammonia may represent a novel, peripherally derived

mediator of METH-neurotoxicity. Investigation of the role of peripheral organ damage

and ammonia in the neurotoxicity of METH is significant because they represent novel

targets for the treatment of the damage produced by the drug and more broadly highlights

the significant contribution of altered peripheral organ function to neuropsychiatric

disorders.

15

Figure 1.1:Model of METH Neurotoxicity: METH causes increases in dopamine, serotonin, glutamate and hyperthermia which contribute to established mechanisms of neurotoxicity including oxidative stress, metabolic compromise and excitotoxicity. These mechanisms converge to produce the long-term, persistent neurotoxicity associated with drug use (solid arrows). In the current series of studies, we hypothesize that METH causes liver damage and increases ammonia which play a critical role in the established mechanisms of damage and neurotoxicity (arrows and ?). We also examine if liver damage results from hyperthermia and if increases in ammonia contribute to increased glutamate (arrow and ?).

16

1.12 Hypotheses

The overarching hypothesis of this dissertation research was that the peripheral

organ effects of METH contribute to the long-term neurotoxicity of the drug. More specifically, METH produces liver damage and increases ammonia to cause long term striatal dopamine and serotonin terminal damage mediated by established excitotoxic mechanisms. Chapter 2, features experiments that examined the effects of METH on the liver and determined whether METH-induced hyperthermia is a mechanism by which the drug contributes to the hepatic damage. Chapter 3, includes experiments that examined if the liver damage produced by METH affects ammonia metabolism and whether the increases in ammonia contribute to the neurotoxicity of the drug. Chapter 4 highlights experiments that determined if ammonia contributes to the neurotoxicity of METH via increases in extracellular glutamate and subsequent excitotoxic damage. These series of related studies were designed to establish a role for peripheral organ damage and ammonia in the neurotoxicity of METH.

17

Chapter 2:

Methamphetamine causes Acute Hyperthermia-Dependent

Liver Damage

2.1 Introduction

Methamphetamine (METH) is a widely abused psychostimulant that causes

neuronal damage (Ricaurte et al., 1980, Seiden et al., 1988), yet few studies have

examined peripheral organs for damage caused by the drug. Most studies of METH have

focused on the central nervous system effects of the drug, as the drug contributes to

altered neuronal function, addiction, and cellular damage. An early report on the clinical

presentation of intravenous METH abusers noted hepatitis as the second most common

presenting complaint (Smith and Fischer, 1970) but since that finding, little more has

been studied along these lines until a clinical report demonstrating that METH causes

acute liver failure in the absence of viral hepatitis (Kamijo et al., 2002). Thus, there is a

high potential for METH to produce hepatotoxicity (Smith and Fischer, 1970, Kamijo et

al., 2002, Wijetunga et al., 2003, Ago et al., 2006). Despite these findings, the

hepatocellular damage produced by METH has yet to be extensively characterized and

the mechanism by which the drug contributes to this damage is unknown. 18

METH-induced hepatocellular damage may be the consequence of the direct effects of the drug on the liver, other peripheral effects on cardiac function, or alterations in body temperature. In fact, METH causes considerable hyperthermia which is strongly linked to the neuronal damage produced by the drug (Bowyer et al., 1992, Bowyer et al.,

1994, Albers and Sonsalla, 1995, Haughey et al., 2000, Xie et al., 2000, Kiyatkin and

Sharma, 2009). The significant hyperthermia (39-42 °C) produced by amphetamine exposure likely results from drug-induced increases in monoamine concentrations and the complex integration of the subsequent hyperlocomotion, altered metabolism, changes in hypothalamic neurotransmission, and vasoconstriction (Wang et al., 1990, Brown et al.,

2007, Sprague et al., 2007, Benamar et al., 2008). Also worth noting is that hyperthermia, in the context of heat , produces significant changes in hepatocellular morphology (Kew et al., 1970, Bianchi et al., 1972, Weigand et al., 2007).

Changes in hepatocellular morphology can be detected using hematoxylin and eosin staining for acidic and basic components of the tissue at the light microscopic level.

In addition, specific ultrastructural changes to hepatocytes can be characterized using electron microscopy (Katz, 1989, Batt and Ferrari, 1995). Hepatocellular enzyme serum concentrations can also be utilized as selective biomarkers for hepatic tissue damage

(Ozer et al., 2008). Therefore, to examine if METH causes liver damage, we used H&E staining and AST and ALT measurements to examine hepatocellular morphology after

METH treatment. To further characterize acute liver damage after a binge dose of

METH, we used transmission electron microscopy to assess damage at 24 hours after

METH. We have also quantified AST and ALT at this same time point to examine if

19

METH produces persistent increases in these measures. Finally, we examined if METH-

induced hyperthermia contributes to drug-induced liver damage.

2.2 Methods

Rat Treatment

Male Sprague Dawley rats were treated with METH (10 mg/kg i.p.every 2 hr x4 ) or

saline (1 mL/kg i.p. every 2 hr x4). This dose simulates the concentration and binge

dosing paradigm reported in METH-dependent humans and produces the same long-term neuronal damage seen in humans after METH exposure (McCann et al., 1998, Volkow et al., 2001, McKetin et al., 2006, Cruickshank and Dyer, 2009). Rats were killed by rapid

decapitation 24 hours after the last injection of drug or were anesthetized for ventricular

perfusion and exsanguination for ultrastructural investigation of tissues. Body temperature

was remotely monitored during the experiments using transponders that were

subcutaneously implanted 2 days before drug treatment (IPTT-300 transponder, BDMS).

Implantation of temperature transponders allowed for remote, repeated, and noninvasive

monitoring of temperature during drug treatment. In some experiments, METH-induced hyperthermia was blocked by cooling the external environment with ice packs placed around the test cages. Temperature was measured every 10 minutes throughout drug treatment and for 5 hours after the last injection in all groups to ensure that the body temperature of METH-treated normothermic rats was kept at the same temperature as saline treated rats. All treatments were carried out in accordance with the National

Institute of Health Guide for Care and Use of Rats. All treatments have also been approved by the University of Toledo Institutional Animal Care and Use Committee.

Transmission Electron Microscopy

20

Twenty-four hours after drug treatment, rats were anesthetized (5 mg/kg of xylazine and

75 mg/kg of ketamine) and transcardially perfused via the left ventricle with 100 mL of phosphate buffered saline (pH 7.4) subsequently followed by 200 mL of 3% glutaraldehyde buffered with 0.2 M sodium cacodylate (pH 7.2). After perfusion, livers were harvested and processed using standard electron microscopy procedures including fixation with 1% osmium tetroxide followed by an en bloc stain using saturated aqueous uranyl acetate (pH 3.3), and dehydrated using a series of gradient ethanol concentrations

(30%, 50%, 70%, 90%, 95% and 100%) and acetone. A combination of acetone and resin

Embed It™ Low Viscosity Resin Kit, Polysciences Inc. was used to infiltrate the tissue with resin. Tissue was then embedded in 100% resin and polymerized overnight at 80 ºC.

Ultra thin sections (60-90 nm) were acquired using an OMU3 C. Reichert

Ultramicrotome and stained using saturated uranyl acetate and Reynold’s lead citrate.

Images were acquired using a Philips CM 10 transmission electron microscope.

Hematoxylin and Eosin Staining

Liver tissue was postfixed overnight in 10% buffered formalin (Fisher Scientific) and

embedded in paraffin. Blocks were sectioned at 4 µm and stained with hematoxylin and

eosin using a Leica Autostainer XL.

Asparate Aminotransferase and Alanine Aminotransferase Determination

Trunk blood was collected after rapid decapitation for measuremen of serum aspartate

aminotransferase (AST) and alanine aminotransferase (ALT). Serum was prepared by

allowing blood to coagulate and then centrifuged for 10 minutes at 3000 x g to separate

the serum. AST and ALT levels were determined using a UniCel DxC800 Synchron

Clinical System (Beckman Coulter).

21

Statistical Methods

For samples examined at 2 hours after METH, analyses for AST and ALT levels in saline and METH treated rats and brain concentration of METH in rats treated with lactulose or vehicle and METH were carried out using a t-test. For samples examined at 24 hours after treatment, analysis of AST and ALT levels in the 3 different treatment groups was carried out using a one-way ANOVA with post-hoc Tukey tests to determine differences between groups. These tests were performed using SigmaPlot 11.0 Software (Systat

Software, Inc. SigmaPlot for Windows). All data are presented as Mean ±SEM. Sample sizes were determined based on a power of 80% or greater and α level in all studies was

0.05 or less.

Results

METH and liver damage

Hematoxylin and eosin staining were used to examine the effects of METH or

saline on the livers of treated rats. Livers were examined at 2 and 24 hours after the last

of the 4 injections. At 2 hours, there is moderate vascular and sinusoidal congestion

evident throughout the livers of all METH treated rats (Figure 2.1). This vascular and

sinusoidal congestion were observed in the livers of all METH treated rats examined at

this timepoint and the image in Figure 2.1 is representative of the congestion seen in the

livers of all rats. Similarly, at 24 hours, extensive cytoplasmic damage is observed in the

livers of 100% of METH treated rats.

To confirm that the morphologic changes depicted in Figure 2.1 represent

significant hepatocellular damage, the serum concentration of hepatocyte cytosolic

enzymes was measured at 2 hours after the last injection of METH (10 mg/kg i.p. every 2

22

hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x 4) in the same rats examined in Figure 2.1.

When hepatocytes are damaged, cytosolic contents such as aspartate aminotransferase

(AST) and alanine aminotransferase (ALT) are released into the blood and can be measured. At 2 hrs after the last injection of METH, serum AST concentration was 193.4

± 15.7 IU/L in saline treated rats and significantly elevated to 447.8 ±74.8 IU/L in METH treated rats (t=215.00, p<0.001). At this time point, ALT concentration was 70.6 ± 3.2

IU/L in saline treated rats and significantly increased to 109.3 ± 9.8 IU/L in METH treated rats (t=95.0, p<0.005). Accordingly, AST and ALT levels are significantly elevated by 141.0 ± 44.1% and 54.9 ± 15.2% respectively, in METH treated rats (Figure

2.2).

METH and Ultrastructural Liver Damage

Transmission electron microscopy was used to characterize the significant

METH-induced alterations in hepatocellular morphology seen at 24 hours after treatment on an ultrastructural level. Ultrastructural changes seen in hepatocytes of 100% of

METH-treated rats include microvesicular fatty change, mild hydropic change, increased cytoplasmic glycogen and mitochondrial aggregation (Figure 2.3).

Environmental Cooling and Methamphetamine-Induced Hyperthermia

To examine if METH-induced hyperthermia contributes to the liver damage observed 24 hours after exposure to METH, METH-induced acute hyperthermia was prevented by cooling METH-treated rats to the body temperatures of saline control rats throughout

METH treatment and for 5 hours after the end of drug treatment. METH treatment caused significant hyperthermia throughout and after METH treatment, which was fully blocked

23

by cooling the external environment of rats (Figure 2.4). Cooling resulted in METH

treated rats with normothermic body temperatures.

Hyperthermia and METH-Induced Changes in Liver Morphology

Hematoxylin and eosin staining was used to determine if hyperthermia contributes to the

alterations in hepatocellular morphology throughout the hepatic lobule occurring 24 hrs

after METH in hyperthermic, normothermic, and saline control rats. At 24 hr after METH

exposure, there was considerable clearing of cytoplasmic staining in hepatocytes

throughout the hepatic lobule. The prevention of METH-induced hyperthermia blocked

the changes in cellular morphology seen in the livers of METH treated rats (Figure 2.5).

The cellular alterations in hyperthermic METH-treated rats were evident in 100% of the

METH treated rats and were prevented in all normothermic METH-treated rats.

Hyperthermia and METH-Induced Hepatocellular Damage

To examine if the changes observed in hepatocellular morphology at 24 hrs after drug treatment are indicative of cellular damage and if hyperthermia contributes to METH- induced cellular damage, serum concentrations of the hepatocellular enzymes aspartate aminotransferase (AST) and alanine aminotransferase (ALT) (Ozer et al., 2008) were measured 24 hours after treatment with METH in rats that were hyperthermic, cooled to normothermia, or saline control rats (Figure 2.6). Serum AST concentration was 335.5 ±

29.1 IU/L in hyperthermic METH treated rats and 177.4 ±13.3 IU/L in saline treated rats, representing an increase of 89.1% ± 16.4 % in hyperthermic METH-treated rats. In normothermic METH-treated rats, AST levels were 231.9 ± 20.4 IU/L, representing an increase of only 30.7% ± 11.5%, compared to Saline treated rats. A one-way ANOVA revealed significant differences between the 3 groups (F(2, 29) = 13.395, p<0.001). A post-

24 hoc Tukey test showed significant differences in serum AST levels in hyperthermic

METH versus saline treated rats (q=7.205, p<0.001), and hyperthermic METH versus normothermic METH treated rats (q=4.721, p<0.05); however there was no significance differences in serum AST in saline treated versus normothermic METH treated rats

(q=2.484, p=0.203).

ALT concentration was 83.9 ± 6.3 IU/L in hyperthermic METH treated rats and

67.5 ±2.6 IU/L in saline treated rats, representing a increase of 24.3% ± 9.3% in METH- treated rats. In normothermic METH-treated rats, ALT levels were 64.8 ± 2.8 IU/L. A one-way ANOVA revealed significant differences between the 3 groups (F(2, 29) = 10.005, p<0.001). A post hoc Tukey test revealed significant differences in serum ALT levels in hyperthermic METH versus saline treated rats (q=5.122, p<0.05), and hyperthermic

METH versus normothermic METH treated rats (q=5.800, p<0.05), however there was no significance difference between saline treated and normothermic METH treated rats

(q=0.678, p=0.882).

25

Figure 2.1: METH causes hepatocellular damage visible by H&E Staining: H&E staining was used to examine the effects of METH (10 mg/kg i.p. every 2 hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x 4) on the livers of treated rats. Livers were examined at 2 and 24 hrs after the last injection of METH or saline. A representative image of a hepatic lobule near the central vein is shown (* denotes central vein). The images in the figure are representative of all rats examined for each treatment and at each time point. At 2 hrs, there is moderate vascular and sinusoidal congestion evident throughout the livers of METH treated rats (white arrows). At 24 hrs, this congestion leads to extensive cytoplasmic disappearance in the livers of all METH treated rats (black arrows). (40x magnification)

26

Figure 2.2: Serum AST and ALT after METH treatment: Rats were treated with METH (10 mg/kg i.p. every 2 hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x4). At 2 hrs after the last injection of METH or saline, A) ALT and B) AST levels are significantly elevated to 54.9 ± 15.2% and 141.0 ± 44.1% in METH treated rats compared to saline controls, respectively. Values are Mean + SEM, *p<0.05 (n= 12 rats per group)

27

Figure 2.3: Ultrastructural Changes in Liver 24 Hours After Methamphetamine Exposure: Representative images from METH or Saline treated groups are shown. Hepatocytes of METH treated rats have increased microvesicular lipid (#), hydropic change (&), increased intracellular glycogen stores (*) and mitochondrial aggregation (arrow) compared to control animals. (5200 x magnification)

28

Figure 2.4: Effect of Environmental Cooling on Methamphetamine-Induced Hyperthermia: Rats were treated with METH or saline (Arrows denote injections). The external environment of a group of METH treated rats was cooled throughout drug treatment and for 5 hours after the last injection to prevent METH-induced hyperthermia. Cooling was regulated to ensure that the body temperatures of METH-treated cooled rats were similar to that of saline treated rats. METH produced a significant elevation in body temperature throughout treatment compared to the saline treated and METH cooled groups. Body temperature of METH cooled rats did not differ significantly from saline treated rats. * p<0.05 (n=10 per group)

29

Figure 2.5: Effect of Hyperthermia on Methamphetamine-Induced Changes in Liver Morphology: Hepatocellular morphology was evaluated using H&E sections from livers collected at 24 hours after the last drug injection. A representative picture from all groups of the hepatic lobule near the central vein is shown (asterisk denotes central vein). The livers of METH treated rats show considerably less hepatocellular cytoplasmic staining (arrows denote cytoplasm) compared to saline treated rats. The cytoplasmic clearing was prevented when METH-induced hyperthermia was blocked by cooling the rats to normothermic temperatures.

30

Figure 2.6: Effect of Hyperthermia on Methamphetamine-Induced Increases in Serum AST and ALT: Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were measured 24 hours after treatment with METH or saline. A group of METH treated rats were cooled during and after treatment to prevent METH induced hyperthermia. A) AST was significantly elevated by 89.1% ± 16.4 % (Mean ± SEM) in METH treated rats compared to saline treated rats and this increase was blocked when hyperthermia was prevented in METH-treated rats (METH-Cooled). B) ALT was significantly elevated by 24.3% ± 9.3% (Mean ± SEM) in METH-treated rats compared to saline treated rats. This increase was blocked when METH-induced hyperthermia was prevented (METH-Cooled). * p<0.05 compared to saline treated rats, & p<0.05 compared to METH treated rats (n=10 per group)

31

Discussion:

This study examined if METH caused acute liver damage, characterized METH- induced hepatocellular damage, and examined if METH-induced hyperthermia contributes significantly to this structural liver damage and hepatotoxicity. METH caused changes in hepatocellular morphology as well as increases in AST and ALT. METH also produced changes in hepatocellular ultrastructure indicative of generalized cellular stress and damage at 24 hours after drug exposure. Increases in serum AST and ALT at this same time point confirmed that there was cellular damage concurrent with these structural changes. To determine if METH-induced hyperthermia contributes to this hepatocellular damage, hyperthermia was prevented in rats treated with METH by cooling the external environment. The prevention of hyperthermia in METH treated rats blocked METH-induced changes in hepatocellular morphology, as well as increases in

AST and ALT, suggesting that hyperthermia plays a significant role in METH-induced liver damage.

Liver damage after exposure to METH has not been examined in-depth previously. While the effects of METH on the brain have been extensively studied, the possible damage to peripheral organs has received less attention. Consistent with clinical reports that METH causes peripheral organ damage (Smith and Fischer, 1970,

Kamijo et al., 2002), the current study is the first to show evidence of hepatotoxicity in a rat model of binge METH exposure. Within 2 hours after METH, there is considerable venous and sinusoidal congestion in the liver (Figure 2.1). This congestion is a typical early marker of hepatotoxicity and may be caused by cardiac dysfunction (Hong et al.,

1991, Wijetunga et al., 2003), post-hepatic vascular constriction (Wang et al., 1990,

32

Chen, 2007), and/or direct drug-induced hepatotoxic damage to the liver. Similarly, serum AST and ALT are well established and sensitive markers for liver damage and were elevated at 2 hrs after METH (Figure 2.2). Although increases in AST or ALT could represent damage to other tissues, the elevations in both along with the hepatic histopathological findings, support the view that METH alone causes acute liver damage

(Ozer et al., 2008). At 24 hours after the last METH injection, the damage escalated to severe global, decreases in cytoplasmic staining (Figure 2.1), suggestive of autophagy

(Rautou et al., 2008).

Electron microscopy of the liver at 24 hours after METH shows microvesicular fatty and hydropic change, increased intracellular glycogen and mitochondrial aggregation (Figure 2.3). These changes are consistent with the generalized hepatocellular damage reported after exposure to hyperthermia and/or heatstroke (Kew et al., 1970, Weigand et al., 2007) evidenced by increases in electron-lucent vacuoles along the sinusoidal border that are indicative of membrane damage and alterations in mitochondria (Wills et al., 1976, Kew et al., 1978). Aggregation of mitochondria may indicate mitochondrial dysfunction and cellular damage (Haga et al., 2003, 2005).

Dysfunction of hepatocellular mitochondria may contribute significantly to increases in ammonia as carbamoylphosphate synthetase-1 and ornithine transcarbamylase, two key enzymes in the urea cycle, are located within mitochondria (Adeva et al., 2012). In addition to these markers of damage, METH also increases intracellular glycogen. This may be representative of increased glycogen formation or decreased glycogen breakdown after METH-exposure. It is unclear if this increase in glycogen is a physiologic or pathologic response to METH treatment. Glycogen is made in the liver as a form of

33 energy storage, increases postprandially, and is regulated by insulin (Parkes and

Grieninger, 1985). Glycogen can also accumulate pathophysiologically in conditions of insulin insensitivity or after exposure to alkaloid compounds which inhibit glycogen breakdown (Saul et al., 1985, Messeri et al., 2012). Regardless, METH-induced increases in liver glycogen may represent significant METH-induced alterations in energy metabolism, hepatic function and warrant further study.

To examine the extent of alterations in hepatocellular morphology, H&E staining was used to visualize if this damage is generalized throughout the hepatic lobule. A considerable decrease in H&E staining of the hepatocellular cytoplasm throughout the lobule was observed at 24 hours after METH exposure (Figure 2.5). This decrease in staining is a likely consequence of increased accumulation of hydrophobic substances in the cytoplasm. METH-induced alterations in hepatocellular ultrastructure (Figure 2.3), including increases in glycogen, microvesicular fat and hydropic change all represent increased accumulation of hydrophobic substances, which do not stain with the acidic and basic H&E staining. This extensive lack of H&E staining throughout the hepatic lobule in METH treated rats demonstrates that the effects of METH on hepatocellular morphology are widespread.

To confirm if these changes in morphology are indicative of damage, we measured serum concentrations of aspartate aminotransferase and alanine aminotransferase (ALT and AST) at 24 hours. Both were found to be significantly elevated at 24 hours after METH treatment (Figure 2.6) and support the conclusion that the changes in cellular morphology observed at this time point represent cellular damage.

These findings also support the observation of acute liver damage reported at 2 hrs after

34

METH exposure (Figure 2.2) and suggest that METH is capable of producing persistent

hepatocellular damage.

We examined if METH hepatotoxicity was a result of METH-induced

hyperthermia based on the fact that the alterations in hepatocellular morphology can be a

response to hyperthermia. Amphetamines produce significant increases in body

temperature as a result of alterations in both brain and peripheral physiology (Wang et al.,

1990, Brown et al., 2007, Sprague et al., 2007, Benamar et al., 2008). The current

findings show that when METH-induced hyperthermia is prevented, the changes and hepatocellular morphology and increases in AST and ALT seen 24 hours after METH treatment are blocked (Figure 2.5 and Figure 2.6). These findings indicate that hyperthermia contributes significantly to the hepatotoxicity of METH and extend previous findings that increases in body temperature also play a significant role in

METH-induced brain damage (Bowyer et al., 1993, Bowyer et al., 1994) and hepatocellular damage after the METH analogue, 3,4-methylenedioxymethamphetamine

(MDMA) (Carvalho et al., 2001).

The primary mechanism by which hyperthermia is believed to contribute to cellular damage is through oxidative stress, as evidenced by increases in lipid peroxidation and depletions in reduced glutathione after hyperthermia exposure (Skibba et al., 1991, Carvalho et al., 1997). Although hyperthermia appears to be an essential component of METH hepatotoxicity, METH may contribute to hepatocellular damage through other mechanisms. METH has been shown to reach some of the highest and longest lasting concentrations in liver and accordingly, has the potential to be directly hepatotoxic (Volkow et al., 2010). METH is metabolized in the liver by the cytochrome

35

p450 system (Lin et al., 1997) that can lead to the formation of oxidative byproducts and

result in cellular damage (Moon et al., 2008, Pourahmad et al., 2010, Letelier et al.,

2011). METH has also been shown to have extensive effects on cardiovascular function,

causing cardiac dysfunction and widespread vasospasm (Wang et al., 1990, Wijetunga et

al., 2003, Chen, 2007), both of which may impede hepatic blood flow, leading to

and contributing to hepatotoxicity.

In conclusion, these studies established that METH does produce acute liver

damage, characterized the toxic effects of METH on the liver, and demonstrated that

these effects can persist for several hours after drug exposure. Furthermore, METH-

induced hyperthermia was identified as a key mechanism for METH-induced hepatotoxicity. These findings highlight the potential role of peripheral organ damage in the neuronal effects and toxicity of drugs of abuse and the importance of addressing hyperthermia in the clinical management of acute METH exposure. To address the consequences of the liver damage identified in this chapter, the measurements of ammonia and how it relates to the neurotoxicity of METH is examined in the following chapter (Chapter 3).

36

Chapter 3:

Peripheral Ammonia Mediates Methamphetamine

Neurotoxicity

3.1 Introduction:

Much research has been directed towards understanding the fundamental processes of addiction to methamphetamine (METH) and the mechanistic underpinnings of its neurotoxicity. The causes of the neurotoxicity are conventionally thought to originate in the brain; however, due to the metabolic dependence of the brain on peripheral organ function and the high susceptibility of the brain to alterations in systemic physiology, the liver damage produced by METH that was identified in Chapter

2 may contribute significantly to the neurological damage caused by the drug.

METH produces relatively specific damage to dopamine and 5-HT terminals in the striatum, hippocampus and prefrontal cortex (Ricaurte et al., 1980, Wagner et al.,

1980, Seiden et al., 1988). Established mechanisms include excitotoxicity, oxidative damage, inflammation, and metabolic compromise (Yamamoto and Raudensky, 2008), all of which are assumed to be initiated by factors in the CNS. However, little consideration is given to peripheral organ damage and it is unknown if increases in brain ammonia mediate the neurobiological actions of METH despite knowledge that it can 37

compromise CNS function as evidenced in hepatic encephalopathy. Because the liver is

essential for ammonia metabolism and elimination, is a key

pathological feature of hepatic encephalopathy and is responsible for the neurological

sequelae depicted by motor and cognitive deficits (Felipo and Butterworth, 2002).

Beyond understanding the mechanism of METH-induced structural hepatocellular damage, it is also important to determine if there is a correlation between morphologic alteration and diminished liver function which can affect METH-induced neurotoxicity.

A key function of the liver is the metabolism of ammonia to urea via the urea cycle.

When liver function is compromised, ammonia accumulates and has neurotoxic consequences (Felipo and Butterworth, 2002). Accordingly, it is crucial to examine if

METH increases plasma ammonia concentrations and if the prevention of METH- induced liver damage also blocks the increases in peripheral ammonia. In Chapter 2, we

found that preventing METH-induced hyperthermia protected against the liver damage

caused by the drug. Here, we will examine if preventing liver damage via blocking drug-

induced hyperthermia also protects against METH-induced increases in plasma ammonia.

Ammonia, much like METH, causes neuronal damage via excitotoxicity,

oxidative stress, and inflammation. Ammonia alone has been shown to decrease the

expression of the astrocytic glutamate transporter, EAAT-1, thereby increasing extracellular glutamate, depolarizing glutamate receptors, and producing excitotoxicity

(Fan and Szerb, 1993, Chan and Butterworth, 1999, Chan et al., 2000). Ammonia also increases glutamate release from astrocytes (Gorg et al., 2010) and ammonia neurotoxicity is attenuated by antagonists (Saez et al., 1999, Kosenko et al., 2003a). Ammonia also causes oxidative damage through alterations in oxidative

38 metabolism (Hawkins et al., 1973, McCandless and Schenker, 1981), increases superoxide radicals, and elevates glutathione peroxidase, superoxide dismutase and catalase activities (Kosenko et al., 1997). Moreover, inflammation mediates ammonia neurotoxicity as ibuprofen attenuates the behavioral consequences of the neuronal damage caused by ammonia (Cauli et al., 2007).

The parallels between the mechanisms underlying ammonia and METH neurotoxicities prompted our hypothesis that METH increases plasma ammonia concentrations, in a liver damage-dependent manner. We have also examined if METH increases brain ammonia concentrations, which converge with the neuropharmacological effects of METH to produce neuronal damage. To this end, we assessed plasma and brain ammonia, and the direct effects of METH and/or ammonia on striatal dopamine and

5HT content. To further substantiate a causative role for ammonia, peripheral ammonia excretion was enhanced with lactulose and the long term effects on dopamine and 5HT content were examined after METH.

3.2 Methods:

Rat Treatment:

Male Sprague Dawley rats received METH (10mg/kg i.p. every 2 hr X 4,) or saline (1 mL/kg i.p. every 2 hr X 4,) (Sigma-Aldrich). This dose of METH was chosen because it produces long term neuronal damage similar to what is seen in studies of human METH users (McCann et al., 1998). In lactulose experiments, rats received lactulose

(Pharmaceutical Associates Inc., Greenville, SC) (5.3 g/kg) or vehicle (100 mg/mL galactose and 80 mg/mL lactose) (Fisher Scientific) via oral gavage every 12 hours for 2 days before METH treatment to the day before rats were killed. Lactulose enhances

39

ammonia excretion and attenuates increases in plasma ammonia and the neurological

symptoms of hepatic encephalopathy (Jia and Zhang, 2005, Nicaise et al., 2008, Al Sibae

and McGuire, 2009). In some experiments, METH-induced hyperthermia was blocked

by cooling the external environment with ice packs placed around the test cages.

Temperature was measured every 10 minutes throughout drug treatment and for 5 hours

after the last injection in all groups to ensure that the body temperature of METH-treated normothermic rats was kept at the same temperature as saline treated rats. All rats were killed by rapid decapitation 2 hrs after the end of treatment or 24 hrs or 7 days after the start of the METH or saline treatment. Temperature transponders (IPTT-300 transponder,

BMDS Inc.) were subcutaneously implanted into the rats 2 days before the beginning of each experiment to allow for equilibration. This method of remote temperature monitoring minimizes stress to the rats produced by repeated rectal recordings. All treatments were carried out in accordance with the National Institute of Health Guide for

Care and Use of Rats. All treatments have also been approved by the University of

Toledo Institutional Animal Care and Use Committee.

Plasma Ammonia Determination

Trunk blood was collected after rapid decapitation for measurement plasma ammonia.

Plasma was prepared by collecting blood in BD Microtainer Plasma Separation Tubes

(Becton, Dickenson and Company) and centrifuging for 2 minutes at 10,000 x g.

Ammonia concentration was determined using a UniCel DxC800 Synchron Clinical

System (Beckman Coulter).

In-Vivo Microdialysis:

40

Intracranial probes for use in microdialysis were constructed using PE 20 tubing (Becton

Dickinson), silica tubing (OD of 150 µM, Polymicro Technologies), 26 ga stainless steel hypodermic tubing (Small Parts), Hollow Fiber Microdialysis Membrane (4 mm of active membrane, MWCO 13,000, 216 µm, Spectrum Labs), 2 ton waterproof epoxy and tygon microbore tubing (Yamamoto and Pehek, 1990). Surgeries were performed on the morning of the day before drug treatment. Rats were anesthetized using xylazine (5 mg/kg) and ketamine (75 mg/kg) and the probe was stereotaxically lowered into the striatum. For brain ammonia measurements, one day later and after a 1 hr. equilibration period at a flow rate of 1.5 µL/min, baseline samples were collected every hour for 3 hrs.

Samples were collected every hour during drug treatment until 2 hrs after the last drug injection. For reverse dialysis experiments, METH (100 µM), and/or ammonium chloride (570µM) were reverse dialyzed for 8 hrs. Probe placement was verified histologically. For reverse dialysis experiments, 450 µM thick sections containing the probe tract cut using a cryostat and tissue adjacent to the tract (1mm) was dissected while the tissue was kept frozen.

Brain Ammonia Determination:

Ammonia concentrations in microdialysate were determined using the Sigma-Aldrich

Ammonia Assay Kit. This assay uses L-glutamate dehydrogenase to measure ammonia concentration as a stoichiometric product of NADPH levels via the reaction: α-

+ + ketoglutarate + NH4 + NADPH  L-Glu + NADP + H2O. NADPH levels were monitored by measuring absorbance at 340 nm using a Molecular Devices Spectramax microplate spectrophotometer. An internal standard made of 10 µg/mL of ammonium

41

sulfate was used in each determination and only assays in which the concentration of the

internal standard was within 5% of the true concentration of ammonia were used.

HPLC Analysis of Monoamine Tissue Content:

Tissue was sonicated in 1.0 mL of 0.25 N perchloric acid and centrifuged at 14000 x g

for 20 mins at 4°C. For reverse dialysis experiments, 300 µL of perchloric acid was used.

Supernatant was then analyzed for 5HT and dopamine using HPLC (Breier et al., 2006).

Supernatant was injected on a C18 column (100 x 2 mm, 3µm particle diameter,

Phenomenex) and eluted with a mobile phase containing 32 mM citric acid, 54.3 mM

sodium phosphate, 0.215 mM octyl sodium sulfate, and 11% methanol (pH 4.4).

Concentration of monoamines was determined with a LC-4C amperometric detector

(Bioanalytical Systems, Inc.) and data were recorded using EZ Chrom Software.

Monoamine levels were normalized to protein content of samples. The pellet from the centrifugation was neutralized using 1.0 mL of 1 M NaOH and protein amount was determined using a Bradford Assay. For reverse dialysis experiments, 100 µL of NaOH was used.

Western Blot for DAT Immunoreactivity:

To prepare synaptosomes, striatal tissue was homogenized in 0.32M sucrose.

Homogenate was then centrifuged at 800 x g for 24 minutes. The pellet was discarded

(P1) Supernatant (S1) was decanted and centrifuged at 22,000 x g for 17 minutes.

Supernatant was discarded (S2) and the pellet (P2) was resuspended in cold Millipore

water. A Bradford assay was then used to determine protein concentration and samples

were diluted (1:4) with LDS sample buffer (Invitrogen). 10 µg of each protein was

loaded onto a NuPAGE 4-12%, 1.5 mm x 10 wells, Bis-Tris Gel (Invitrogen). Proteins

42

were then run at 150V for 80 minutes and transferred to polyvinylidine difluoride

membranes at 28 V for 2 hours. Membranes were then washed with TBS-T (20 mM Tris,

137 mM NaCl and 0.5% Tween 20) and blocked for 1 hour at room temperature using

TBS-T with 5% powered milk. Membranes were incubated overnight at 4˚C with

primary antibody to dopamine transporter (DAT, C-20 Santa Cruz Biotechnology).

Membranes were then washed with TBS-T incubated with the appropriate horseradish

peroxidase secondary antibodies (1:2500 dilution) at room temperature for 1 hour, and

developed using the HyGLO-enhanced chemilumescence (Denville Scientific). All

membranes were imaged using the Fuji LAS-4000 mini system and software. The band

at 70 kDa was quantified and all bands were normalized to an α-tubulin internal loading control.

Mass Spectroscopy:

Mass spectroscopy was used to determine METH concentrations in striata. Tissue was

prepared as in the 5HT and dopamine tissue content assays. Supernatant was then

injected on to a MetaChem MetaSil Basic column (3.0×100 mm; 3 μM pore size; Varian,

Lake Forest, CA) and measured using electrospray ionization as the ionization source.

Statistical Analysis:

Analysis of plasma ammonia, hyperthermia, and tissue content of dopamine and 5HT in

the striatum was performed using a two-way ANOVA followed by post hoc Tukey

multiple comparison tests or t-tests. These tests were performed using SigmaPlot 11.0

Software (Systat Software, Inc. SigmaPlot for Windows). Statistical analysis of brain

ammonia levels was carried out using a repeated measures ANOVA test, with each time

point as a within-subjects factor and lactulose and METH treatment as between-subjects

43 factor. These tests were performed using SPSS Statistics 17.0 Software (IBM

Corporation).All data are presented as Mean±Standard Error of the Mean (SEM). α level in all studies is 0.05 or less and sample sizes were chosen to result in a power of 80% or greater.

3.3 Results:

METH and plasma and ammonia

To determine if METH affected ammonia metabolism, plasma ammonia was measured at 2 hrs after the end of METH treatment (10 mg/kg i.p. every 2 hrs x 4) or

Saline (1 mL/kg i.p. every 2 hrs x 4). A two-way ANOVA indicated a significant effect of METH treatment (F(1,31) =18.25, p<0.001), lactulose treatment (F(1,31) =13.60, p<0.001), and a significant interaction between METH and lactulose treatment (F(1,31)

=22.98, p<0.001), At 2 hrs after the last injection of METH, plasma ammonia levels were significantly elevated by 64.04±9.98%, and this increase was blocked with lactulose treatment (Figure 3.1).

Effect of Hyperthermia on METH-Induced Increases in Plasma Ammonia

To examine if METH-induced increases in plasma ammonia persist for 24 hours and if hyperthermia contributes to those increases, plasma ammonia was measured 24 hours after treatment with METH in hyperthermic and normothermic rats, and saline controls (Figure 3.2). Plasma ammonia concentration was 154.17 ± 7.51 µM in hyperthermic METH treated rats and 71.78± 6.51 µM in saline treated rats, representing an increase of 114.8 ± 10.5%. In normothermic METH-treated rats, plasma ammonia concentration was 80.50 ± 6.02 µM, representing only a 12.2 ± 8.3% increase, compared with saline treated rats. A one-way ANOVA revealed significant differences between the

44

3 groups (F(2, 24) = 38.45, p<0.001). A post hoc Tukey test examining group differences

revealed significant differences in plasma ammonia in hyperthermic METH versus saline

treated rats (t=9.198, p<0.001) and hyperthermic METH versus normothermic METH

rats (t=7.481, p<0.001), however there was no significant difference in plasma ammonia

in saline treated versus METH-normothermic rats (t=0.996, p=0.330).

METH and Brain Ammonia

To examine if elevations in plasma ammonia translate to elevations in brain

ammonia, the brain concentration of ammonia was measured using in-vivo microdialysis

during METH or saline treatment. Rats were also treated with lactulose or vehicle every

12 hrs for 2 days before and on the morning of drug treatment. Baseline brain ammonia

levels in all groups were 29.34±5.33 µM. A repeated measures ANOVA indicated a

significant effect of METH treatment (F(7,279)=2.43, p<0.050) and a significant interaction

between METH and lactulose treatment (F(7,279)=2.79, p<0.050) on brain ammonia

indicating that METH increased brain ammonia and that this increase was blocked by the

peripheral administration of lactulose. Brain ammonia levels in METH treated rats are

significantly elevated beginning at the 5 hr time point (F(3,31)=4.22, p<0.05). At the 8 hr time point (Figure 3.3), brain concentration of ammonia in saline treated rats was

34.5±8.0 µM and was 67.9±8.7 µM in METH treated rats representing a 196.8±18.0% increase in ammonia in the striata of METH treated rats. Based on the observation that the in vivo recovery of ammonia by the microdialysis probe is 35% (data not shown), brain ammonia at 8 hrs after the first METH injection is approximately 200 µM.

METH and Increased Peripheral Ammonia Excretion

45

Rats were given lactulose or vehicle every 12 hrs, p.o. for 2 days before and 6

days after METH or saline treatment to ensure that ammonia excretion was consistently

increased throughout the presumed time course over which METH causes neuronal

damage.

During drug treatment, the effects of METH and lactulose on body temperature

were measured one hour after each METH injection. METH treatment caused significant

hyperthermia in both vehicle+METH and lactulose+METH treated rats. Lactulose had

no significant effect on hyperthermia (Figure 3.4) indicating that the effects of lactulose

are not dependent on alterations in hyperthermia.

To examine if lactulose affected the brain concentration of METH, the

concentration of METH in the striatum was measured at 2 hrs after the last injection of

METH using LC-MS-MS. Each of the 4 injections were within the half-life of METH

and at this time point the drug has presumably reached an approximate steady-state

concentration in the brain, thus optimizing the detection of any effect that lactulose may

have on the brain concentration of METH. Striatal concentration of METH in

vehicle+METH treated rats was 2.11x10-2 ±0.28x10-2 ppm/mg protein and 1.96x10-

2±0.13x10-2 ppm/mg protein in lactulose+METH treated rats. The concentrations of

METH in the brain did not differ significantly between the vehicle+METH and

lactulose+METH groups supporting the view that lactulose does not alter the

concentration of METH in the brain.

To examine the effects of METH and lactulose on striatal tissue content of dopamine and 5-HT, rats were killed at 7 days after METH or saline treatment and tissue content of dopamine and 5HT was measured. A two-way ANOVA indicated a

46

significant effect of METH treatment (F(1,23)=30.51, p<0.001) and a significant

interaction between METH and lactulose treatment (F(1,23)=11.03 p<0.005) on dopamine

tissue content indicating that the effects of METH on dopamine content varied according

to whether the rats were treated with lactulose. Tukey post-hoc analysis revealed that

there was no effect of lactulose treatment within saline treated groups (q=2.5; p=0.093);

however lactulose attenuated the dopamine depletion caused by METH (q=8.08;

p<0.001). Treatment with lactulose significantly attenuated the striatal dopamine

depletions caused by METH as METH depleted striatal dopamine by only 34.5 ± 7.9% in

lactulose treated rats (Figure 3.5A), suggesting that ammonia may play a role in the

dopaminergic toxicity produced by METH.

Likewise, a two way ANOVA showed a significant effect of METH treatment on

striatal 5-HT content (F(1,23)=27.59 , p<.0.001) and a significant interaction between

METH treatment and lactulose treatment on striatal 5-HT (F(1,23)=2.28 p<0.050). Tukey

post-hoc tests indicated that lactulose had no effect within saline treated rats (q=0.98;

p=0.34); however, lactulose attenuated the 5-HT depletion caused by METH (q=3.80;

p<0.001). METH treatment produced a depletion of 5-HT by only 15.2 ± 8.2% in

lactulose treated rats, which was not significantly different compared to saline treated rats

(Figure 3.5B), indicating that ammonia may play a key role in METH-induced serotonergic damage.

Both pre- and post-treatment with lactulose were required for attenuation of

METH-induced neuronal terminal damage (data not shown), indicating that elevations in

brain ammonia both during and in the days subsequent to METH treatment may play a

role in monoaminergic terminal damage. Pre-treatment with lactulose was required as

47

the drug takes 24 to 48 hours of dosing to be effective (Jia and Zhang, 2005, Nicaise et

al., 2008, Al Sibae and McGuire, 2009).

To confirm the effects of lactulose on dopamine tissue content, DAT immunoreactivity was measured in the striatum at 7 days after METH treatment. A two-

way ANOVA indicated a significant effect of METH treatment (F(1,23)=4.708, p<0.05)

and a significant interaction between METH and lactulose treatment (F(1,23)=4.708,

p<0.05). Post-hoc tests reveal no effect of lactulose within saline treatment groups

(q=2.129, p=0.311) and a significant effect of lactulose within METH treatment groups

(q=5.729, p<0.05). METH treatment produced a 50.5±19.5% depletion in

Vehicle+METH treated rats and a non-statistically significant 19.5±9.8% depletion in

Lactulose+METH treated rats (Figure 3.6).

Local Administration of METH and Ammonia

To examine if ammonia directly contributes to the neuronal damage caused by

METH, METH (100 µM) and/or ammonia (570 µM) was reverse dialyzed into the striatum for 8 hours. Dopamine and 5-HT tissue content of the tissue adjacent to the microdialysis probe was dissected 7 days later. Our previous studies showed that the local administration of 100 µM METH into the striatum increased striatal dopamine release to the same levels caused by the systemic administrations of 10 mg/kg, every 2 hrs x 4 injections (Burrows et al., 2000). Ammonia (570 µM) was reverse dialysed to approximate the same brain concentration of ammonia previously measured after systemic METH treatment (Figure 3.7), considering the in-vivo recovery of ammonia by the microdialysis probe is 35%. The effects of reverse dialysis of METH and ammonia on dopamine and 5-HT tissue content in the striatum were determined (Figure 3.5). A

48 two-way ANOVA showed a significant interaction between METH and ammonia on dopamine tissue content (F(1,32)=4.5, p<0.050) and post-hoc Tukey tests revealed a significant difference between the combination of METH and ammonia compared to

METH alone (q=4.8, p<0.005) or ammonia alone (q=4.9, p<0.005) (Figure 3.7A).

Similarly, a two-way ANOVA showed a significant interaction between METH and ammonia on 5HT tissue content (F(1,32)=5.6, p<0.050). Tukey post hoc tests revealed a significant difference between the combination of METH and ammonia compared to

METH alone (q=4.4, p<0.005) or ammonia alone (q=6.0, p<0.001) (Figure 3.7 B).

Specifically, the combination of ammonia and METH produced a 51.5±12.5% depletion of dopamine and 41.8±7.0% depletion of 5-HT, when compared with artificial CSF controls, demonstrating that ammonia synergizes with METH to directly contribute to the neuronal damage caused by METH.

49

Figure 3.1: METH and plasma ammonia: Rats were treated with METH (10 mg/kg i.p. every 2 hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x 4) and Lactulose (5.3 g/kg p.o. every 12 hrs) or vehicle (8 mL/kg, every 12 hrs). At 2 hrs after the last injection of METH or saline, plasma ammonia levels are significantly elevated by 64.04±9.98% (Mean±SEM) and this increase is blocked by lactulose treatment (*= p<0.001 compared to Lac Sal, &=p<0.001 compared to Veh METH). (n=8 rats per group) Veh=Vehicle, Lac=Lactulose, Sal=Saline

50

Figure 3.2: Effect of Hyperthermia on Methamphetamine-Induced Persistent Increases in Plasma Ammonia: Plasma ammonia was measured 24 hours after treatment with METH or saline. A group of METH treated rats were cooled to prevent METH-induced hyperthermia. METH treatment significantly elevated plasma ammonia concentrations by 114.8 ± 10.5%. This increase was blocked by preventing METH-induced hyperthermia (METH-Cooled) * p<0.05 compared to saline treated rats, & p<0.05 compared to METH treated rats (n=10 per group)

51

Figure 3.3: METH and brain ammonia: Rats were treated with METH (10 mg/kg x 4 i.p every 2 hrs) or saline (1 mL/kg x4 i.p. every 2 hrs) and lactulose (5.3 g/kg p.o. every 12 hrs) or vehicle (8 mL/kg, every 12 hrs). During drug treatment (arrows denote injections), striatal ammonia concentration was measured using in-vivo microdialysis. Vehicle+METH treatment significantly increases brain ammonia concentrations starting at timepoint 5 and Lactulose pretreatment attenuates this increase (*=p<0.05). Baseline=29.34±5.33 µM. (n=8-10 rats per group) Veh=Vehicle, Lac=Lactulose, Sal=Saline

52

Figure 3.4: Hyperthermia after METH Treatment: Rats were treated with METH (10 mg/kg i.p. every 2 hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x 4) and lactulose (5.3 g/kg p.o. every 12 hrs) or vehicle (8 mL/kg, every 12 hrs). During drug treatment, temperature was measured one hour after each METH injection (denoted by arrows). METH treatment caused significant hyperthermia in both vehicle+METH and lactulose+METH treated rats, and lactulose had no significant effect on hyperthermia. (*=p<0.001 compared with Veh Sal) (n=6-9 per group) Veh=Vehicle, Lac=Lactulose, Sal=Saline

53

Figure 3.5: Effect of Lactulose on METH-induced monoamine depletions: Rats were treated with METH (10 mg/kg i.p. every 2 hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x 4) and lactulose (5.3 g/kg p.o. every 12 hrs) or vehicle and tissue content of dopamine or 5- HT in the striatum was measured 7 days later. A) Treatment with lactulose significantly attenuated the striatal dopamine depletions caused by METH as METH depletes dopamine by only 34.5 ± 7.9% (Mean±SEM) in lactulose treated rats. B) METH depletes 5-HT by 15.2 ± 8.2% (Mean±SEM) in lactulose treated rats, which was not statistically significant compared to saline treated rats. (* p<0.001 compared to Lac Sal. & p<0.001 compared to Veh METH). (n=6-9 rats per group) Veh=Vehicle, Lac=Lactulose, Sal=Saline

54

Figure 3.6: Effect of Lactulose on METH-induced reductions in DAT Immunoreactivity: Rats were treated with METH (10 mg/kg i.p. every 2 hrs x 4) or saline (1 mL/kg i.p. every 2 hrs x 4) and lactulose (5.3 g/kg p.o. every 12 hrs) or vehicle and tissue content of dopamine or 5-HT in the striatum were measured 7 days later. A) Treatment with lactulose significantly blocked the decrease in DAT immunoreactivity as METH produced only a 19.5±9.8% depletion in Lactulose+METH treated rats. B) Representative western blot for DAT immunoreactivity and α-tubulin internal control (* p<0.05 compared to Lac Sal. & p<0.05 compared to Veh METH). (n=6 rats per group) Veh=Vehicle, Lac=Lactulose, Sal=Saline

55

Figure 3.7: Effects of local administration of ammonia and METH into the striatum: Rats were reverse dialyzed with artificial cerebrospinal fluid, METH(100 µM) and/or Ammonia (570 µM), and/or GKYI 52466 (100 µM) in the striatum for 8 hrs and dopamine and 5HT tissue content of the striatal tissue around the probe membrane (1 mm) was measured 7 days later. A) Treatment with the combination of METH and ammonia depletes dopamine in the tissue around the probe by 51.5±12.5% (Mean±SEM). B) Treatment with the combination of METH and Ammonia depletes 5-HT in the tissue around the probe by 41.8±7.0% (Mean±SEM) compared to aCSF.

56

3.4 Discussion:

This study examined the relationship between changes in ammonia in periphery and the brain and the long term depletions of dopamine and 5HT in the brain typically associated with a high dose binge regimen of METH. METH caused increases in plasma ammonia that were paralleled by increases in brain ammonia. These increases were decreased by the enhancement of peripheral ammonia excretion that in turn, attenuated the long-term depletions of brain dopamine and 5HT produced by systemic exposure to

METH. Furthermore, the local administration of ammonia with METH reproduced the depletions of striatal dopamine and 5HT produced by the systemic administrations of

METH alone.

Peripheral ammonia levels were also measured at 2 and 24 hours after METH exposure to determine if the hepatotoxicity at this time point had functional consequences. The liver is the primary organ of ammonia metabolism via the urea cycle.

When liver function is compromised, ammonia accumulates and leads to neuronal damage (Felipo and Butterworth, 2002). Plasma ammonia levels were significantly elevated at 2 and 24 hours after METH exposure. The finding that increases seen at 24 hours after METH were blocked by preventing METH-induced hyperthermia (Figure 3.2) and is consistent with the blockade of METH-induced liver damage described in Chapter

2. Interestingly, the increases in ammonia seen at 24 hours after METH treatment were comparable to those reported at just 2 hours after METH treatment (Figure 3.1). These findings demonstrate that the hepatocellular damage produced by METH is sufficient to cause persistent increases in plasma ammonia. Furthermore, the findings that both

METH-induced liver damage and increases in ammonia were blocked by the prevention

57 of hyperthermia further strengthen the association between METH-induced liver damage and increases in ammonia to levels that can damage the brain. This also suggests that increases in ammonia may be a mechanism by which hyperthermia contributes to METH neurotoxicity. It is worth noting that while the liver is the primary ammonia metabolizing organ, damage to other organ systems which have been reported to be altered by METH, namely the cardiac, renal and musculoskeletal systems, also may contribute directly or indirectly to increases in ammonia (Smith and Fischer, 1970,

Kamijo et al., 2002, Wijetunga et al., 2003).

No previous studies have examined if and how liver damage and/or ammonia contribute to the long-term neuronal damage produced by METH. The current findings show that METH produced elevations of ammonia in the plasma and striatum that were temporally and physiologically related to the liver damage produced by the drug. In parallel with the liver pathology at 2 hrs after the last METH injection, brain ammonia increased to about 70 µM in the striatal microdialysate (Figure 3.3). This translates to a brain ammonia concentration of approximately 200 µM, considering the recovery of ammonia from microdialysis is approximately 35 % . In cases of hepatic encephalopathy, ammonia concentrations in brain range from 300-1000 µM (Swain et al.,

1992, Clemmesen et al., 1999). Although the concentrations of ammonia observed in the brain after METH are less than those in hepatic encephalopathy, lower concentrations of ammonia in combination with METH exposure appear to cause neurotoxic effects. Thus, liver damage may be an unrecognized consequence of METH exposure and a cause of neuronal damage mediated by peripherally derived ammonia even in the absence of hepatic encephalopathy per se. Also worth noting is that METH-induced damage to other

58

peripheral organ systems may contribute to increases in ammonia either directly, as in the

case of renal damage and rhabdomyolysis, or indirectly via influence on hepatic function,

as in the case cardiac dysfunction and especially, right-sided heart failure (Smith and

Fischer, 1970, Kamijo et al., 2002, Wijetunga et al., 2003, Ago et al., 2006). Thus, studies of the possible contributions by these organ systems and ammonia to METH- induced neurotoxicity are warranted.

We also investigated if ammonia derived from the periphery contributes to the elevations in brain ammonia and the neuronal damage after METH exposure. Peripheral ammonia excretion was increased using lactulose. Lactulose is a synthetic disaccharide that is not absorbed systemically but remains in the gut. It is degraded by bacteria in the colon thereby acidifying the colonic contents, converting ammonia to the ammonium ion

+ (NH4 ), thus trapping ammonia and preventing its absorption. Lactulose also functions as

a laxative to reduce time for absorption and metabolism of protein, which is a source of

ammonia. Lactulose prevented the acute increases in brain ammonia and the long term

decreases in dopamine and 5-HT content, without affecting METH-induced hyperthermia

(Figure 3.4, 3.5) and the brain concentrations of METH. This supports the view that the

effects of lactulose are likely due to its effects on peripheral ammonia and not alterations

in the direct pharmacological actions of METH in the brain.

Excretion of ammonia partially protected against dopamine, but blocked the

depletions of 5-HT tissue content produced by METH treatment. The partial protection

of dopamine terminals can be explained by the fact that the established causes of

dopaminergic terminal damage, such as the interaction of METH with the dopamine

transporter (DAT) and vesicular monoamine transporter-2 (VMAT-2), (Fleckenstein et

59

al., 1997, Hogan et al., 2000, Brown et al., 2002) might be relatively less affected by

ammonia. Therefore, METH-induced increases in intra- and extracellular dopamine can

produce ROS and dopamine-quinones and oxidatively damage dopamine terminals

regardless of the presence of ammonia (Schmidt et al., 1985, Fornstedt et al., 1989,

LaVoie and Hastings, 1999). In contrast, there may be causes of serotonergic terminal

damage that are especially ammonia-dependent. Specifically, hyperammoniemia

increases the uptake of L-tryptophan (Grippon et al., 1986), the expression and activity of

monoamine oxidase (Mousseau et al., 1997), and production of 5-HIAA (Batshaw et al.,

1986). Accordingly, ammonia-induced increases in 5-HT turnover may increase the production of oxidative intermediates. Furthermore, hyperammonemia increases the production of the excitotoxin and L-tryptophan metabolite, (Moroni et al.,

1986).

The central application of METH with ammonia recapitulated the neuronal damage produced by systemic METH exposure, even though administration of METH or ammonia alone did not produce neuronal damage (Figure 3.7). The apparent synergy between ammonia and METH indicates both are necessary, but alone are not sufficient to produce toxicity. Ammonia could be toxic through excitotoxic and oxidative damage

(Fan and Szerb, 1993, Kosenko et al., 1995, Kosenko et al., 1997, Chan and Butterworth,

1999) while METH confers selectivity to dopamine and 5HT terminals through its effects on dopamine and 5HT transporters and the dopamine-dependent activation of the striatonigral pathway resulting in increases in extracellular glutamate in the striatum

(Mark et al., 2004).

60

In conclusion, these studies identify a small molecule derived from the periphery

that mediates the neuronal damage produced by METH and note the importance of

considering the periphery in the context of METH-induced neurological dysfunction and damage. These findings suggest that co-morbid conditions affecting liver function or ammonia metabolism may negatively contribute to the neurotoxic consequences of

METH exposure. In particular, co-morbid alcoholic cirrhosis or viral hepatitis with

METH use may exacerbate acute METH-mediated liver damage and subsequent neurotoxicity. Furthermore, these findings highlight acute peripheral organ damage as a possible mediator of damage in other neurological disorders.

61

Chapter 4:

Contribution of Ammonia to Methamphetamine-Induced

Increases in Extracellular Glutamate and Excitotoxicity

4.1 Introduction

Ammonia may contribute to METH-neurotoxicity by mediating drug-induced

increases in glutamate and excitotoxicity. Excitotoxicity is a mechanism of neuronal

damage resulting from increases in extracellular glutamate and glutamate receptor

activation, which leads to increases in intracellular calcium (Choi, 1994). Excitotoxicity

has been identified as a significant mechanism of neuronal damage in many disorders

including Alzheimer’s’ disease, Huntington’s disease, ischemic stroke, and

(Choi, 1988). Glutamate is the primary excitatory neurotransmitter in the central nervous system and is released in a calcium-dependent manner from both pre-synaptic terminals in response to depolarization, as well as from astrocytes (Magistretti, 2006, Santello and

Volterra, 2009). Subsequent to vesicular release, glutamate interacts with both pre- and

post-synaptic receptors including the AMPA, NMDA, and Kainate receptors. Glutamate

released into the synapse is removed by excitatory amino acid transporters located primarily on astrocytes (Danbolt, 2001).

62

Of particular relevance to excitotoxicity are the calcium permeable glutamate

receptors which include the NMDA receptor and non-GluR2 subunit containing AMPA receptors (Hollmann et al., 1991). Physiologically, these calcium-permeable glutamate receptors are important for the development of synaptic plasticity, but are overactivated in the context of significantly elevated extracellular glutamate leading to significant increases in intracellular calcium (Liu and Zukin, 2007). Within the striatum, the expression of calcium permeable AMPA receptors is increased compared to NMDA receptor expression (Fan et al., 1999, Dunah et al., 2000). In particular, non-Glu-R2 subunit containing, calcium permeable AMPA receptors have been identified on dopamine and serotonin terminals within the striatum, making these terminals susceptible to the excitotoxic terminal damage (Keefe et al., 1993, Betarbet and Greenamyre, 1999,

Hernandez et al., 2003). Non-GluR2 containing AMPA receptors may also be located on

GABA interneurons, also increasing the vulnerability of those cells to excitotoxic damage

(Segovia et al., 1997).

Increases in intracellular calcium contribute to cellular damage through numerous mechanisms including the activation of and production of free radicals. The activity of the calcium activated protease, calpain, increases in response to increased intracellular calcium. Conversely, when calpain activity is inhibited, protection against excitotoxicity is provided (Siman et al., 1989, Lee et al., 1991, Squier et al., 1994).

Calpain contributes to neuronal damage through the degradation of essential cytoskeletal proteins, including spectrin, actin, MAP2, neurofilament and Tau (Schlaepfer et al., 1985,

Billger et al., 1988, Harris and Morrow, 1988, Johnson et al., 1989, Villa et al., 1998).

Free radicals are produced in response to increases in intracellular calcium through

63

increases in phospholipase A2 activity, mitochondrial damage, and nitric oxide production

(Chan et al., 1985, Dumuis et al., 1988, Dugan et al., 1995, Gow et al., 1997). Increases in free radicals leads to the oxidative damage of membranes and proteins (Stamler and

Hausladen, 1998). Oxidative modification of specific proteins can reduce the activity of those proteins and further perpetuate cellular damage (Gow et al., 1996).

Subsequent to release, excess glutamate in the synapse is then removed via

excitatory amino acid transporters, located primarily on astrocytes, and metabolized

intracellularly. Decreased astrocytic uptake or intracellular metabolism of glutamate can

contribute to increases in extracellular glutamate and excitotoxicity. Excitatory amino

acid transporters (EAATs) are responsible for the uptake of glutamate and include

EAAT1/glutamate aspartate transporter (GLAST), EAAT2/glutamate transporter-1

(GLT1), and EAATs 3-5 (Had-Aissouni, 2012). The primary glutamate transporters on

astrocytes include EAAT-1/GLAST and EAAT-2/GLT-1 (Lehre et al., 1995) In addition to these glutamate transporters, the cystine-glutamate antiporter system is also important for astrocytic glutamate uptake (Sato et al., 1999). Typically, these transporters move extracellular glutamate into astrocytes from the synapse, however the directionality of these transporters has been shown to be dependent on extracellular potassium levels, such that an increase in extracellular potassium reverses the directionality of these transporters preventing uptake and producing non-vesicular release of glutamate from astrocytes

(Barbour et al., 1988, Szatkowski et al., 1990, Grewer et al., 2008). Once taken up into cells, glutamate, along with ammonia, is metabolized to glutamine via glutamine synthetase (Haberle et al., 2006). Glutamine produced from this reaction is then transported into presynaptic terminals and used for the synthesis of glutamate as part of

64

the glutamate-glutamine cycle. The glutamine produced from this reaction also serves as

a nitrogen source for the biosynthesis of many other important compounds including

tryptophan, glucosamine-6-phosphate, histadine and carbamoyl phosphate (Woolfolk et

al., 1966). Interestingly, glutamine synthetase activity is reduced by nitration subsequent

to glutamate receptor activation and increases in nitric oxide (McBean et al., 1995, Gorg

et al., 2007).

Increases in extracellular glutamate and excitotoxicity contribute significantly to

the neuronal damage produced by METH. METH produces significant increases in

extracellular glutamate levels and inhibition of glutamate receptor activation, or blocking

increases in extracellular glutamate is neuroprotective for the damage produced by the

drug (Farfel et al., 1992, Nash and Yamamoto, 1992, Mark et al., 2004, Tata and

Yamamoto, 2007). METH has also been show to increase calpain-mediated spectrin proteolysis, a marker of excitotoxicity in the striatum, in an AMPA receptor-activation manner (Staszewski and Yamamoto, 2006). Increases in extracellular glutamate are observed acutely during treatment with METH, and result from dopamine D1-receptor mediated disinhibition of corticostriatal glutamate release (Mark et al., 2004). In-vitro, high doses of METH have also been shown to differentially affect glutamine synthetase expression (Stadlin et al., 1998).

As with METH, the neuronal damage produced by ammonia may result from

increases in glutamate and excitotoxicity as this damage is also attenuated with glutamate

treatment (Saez et al., 1999, Kosenko et al., 2003). Ammonia induced increases in extracellular glutamate result from alteration of the local uptake, metabolism and release of glutamate from astrocytes. Ammonia has been shown to decrease the

65

expression of both GLAST and GLT-1 (Chan and Butterworth, 1999, Chan et al., 2000).

High concentrations of ammonia have also been shown to increase calcium-dependent exocytotic glutamate release from astrocytes in-vitro (Gorg et al., 2010). More acutely, the ammonium ion, which has a similar ionic radius to the potassium ion, may contribute to increases in glutamate by reversing potassium-gradient dependent glutamate transporters (Rose, 2002). The metabolism of glutamate within astrocytes is also affected by ammonia. Glutamine synthetase is the key glutamate and ammonia metabolizing enzyme in the brain, is decreased after ammonia exposure and is modified by nitration or

nitrosylation (Kosenko et al., 2003, Gorg et al., 2007). Inhibition of glutamine synthetase

results in the accumulation of both glutamate and ammonia and therefore, represents a

feed-forward mechanism by which ammonia may mediate METH-induced neurotoxicity.

Beyond affecting extracellular glutamate levels, ammonia has been shown to increase

NMDA receptor activation via membrane depolarization and removal of the Mg2+ block

of NMDA receptors (Fan and Szerb, 1993).

Accordingly, the contribution of ammonia to METH-induced elevations in extracellular glutamate was examined. It was hypothesized that ammonia contributes directly to the increases in extracellular glutamate seen during METH treatment as a result of alteration of local glutamate processing, and that this altered glutamate processing, in addition to the previously identified increases in corticostriatal glutamate

release, is required for METH-induced increases in glutamate. In addition to examining

the contribution of ammonia to METH-induced increases in extracellular glutamate, we

have determined if ammonia has a net, downstream effect on METH-induced excitotoxicity by examining calpain-mediated spectrin proteolysis. Finally, we have

66 examined if ammonia and METH contribute to decreased expression or activity of glutamine synthetase and as potential mechanism by which the drug and/or ammonia can contribute to feed-forward mechanisms of neurotoxicity, thereby amplifying the damage caused by both METH and ammonia.

4.2 Methods:

Rat Treatment:

Male Sprague Dawley rats received a high dose of METH (10mg/kg q 2 hr X 4, i.p.) or saline (1 mL/kg q 2 hr X 4, i.p.). In lactulose experiments, rats received lactulose (5.3 g/kg) or vehicle via oral gavage every 12 hours for 2 days before METH treatment until the day before rats were killed. All rats were killed by live decapitation 2 hrs, 5 days or 7 days after the start of the Meth or Saline treatment. Subcutaneous temperature transponders were implanted into the rats 2 days before the beginning of each experiment to allow for accurate monitoring of temperature and to reduce animal stress when temperature was taken. The transponders used were 2.2x14 mm and weigh 120 mg

(IPTT-300 transponder, BMDS Inc.). All treatments were carried out in accordance with the National Institute of Health Guide for Care and Use of rats. All treatments are approved by the University of Toledo Institutional Animal Care and Use Committee.

HPLC Analysis of Monoamine Tissue Content:

Tissue within 1 mm of probe track was dissected and sonicated in 300 µL of 0.25 N

Perchloric Acid and centrifuged at 14000 x g for 20 minutes at 4°C. Supernatant was injected on a C18 column (100x2 mm, 3µm particle diameter, Phenomenex) and eluted with a mobile phase containing 32 mM citric acid, 54.3 mM sodium phosphate, 0.215

67

mM octyl sodium sulfate, and 11% methanol (pH 4.4). Concentration of monoamines

was determined with a LC-4C amperometric detector (BAS Bioanalytical) and data was

recorded using EZ Chrom Software. Monoamine levels were normalized to protein

content of samples. The pellet from the centrifugation was neutralized using 100 µL of 1

M NaOH and protein amount was determined using a Bradford Assay.

HPLC Analysis of Glutamate:

Dialysate (20 µL) was injected on a C18 column (150x2 mm, 3µm particle diameter,

Phenomenex) and eluted with a mobile phase containing 0.1 M Na2HPO4, 0.1 mM

EDTA, and 7.5% Methanol (pH 3.2). Glutamate was derivatized for electrochemical

detection with o-pthaldialdehyde. Concentration of glutamate was determined with a LC-

4C amperometric detector (BAS Bioanalytical) and data was recorded using EZ Chrom

Software. Glutamate levels were normalized to baseline levels where appropriate.

In-Vivo Microdialysis:

Intracranial probes were constructed using PE 20 tubing (Becton Dickinson), silica tubing

(OD of 150 µM, Polymicro Technologies), 26 ga stainless steel hypodermic tubing

(Small Parts), Hollow Fiber Microdialysis Membrane ( 4 mm of active membrane,

MWCO 13,000, 216 µm, Spectrum Labs), 2-ton waterproof epoxy and tygon microbore

tubing. Surgeries were performed the day before drug treatment. Rats were anesthetized

using xylazine (5 mg/kg) and ketamine (75 mg/kg) and the probe was stereotaxically

lowered into the striatum (AP+1.2, ML+3.0, DV-6.5 mm). One day later, and after a 1 hour equilibration period, baseline samples were collected every hour for 3 hrs at a flow rate of 1.5 µL/min. Samples were collected every hour during drug treatment until 2 hrs after the last drug injection. Probe placement was verified histologically.

68

Spectrin Breakdown Product Analysis:

5 days after METH or saline and lactulose or vehicle treatment, rats were be killed by

rapid decapitation and striata were dissected. Tissue was homogenized in a buffer

containing 10mM Tris, 10 mM EGTA, 250 mM sucrose, and Halt protease inhibitor

cocktail (Thermo Scientific). 30 µg of protein of each sample was separated via

electrophoresis, and transferred to a PVDF membrane. Spectrin breakdown products were

examined using an antibody for non-erythroid α-Spectrin (MAB1622; Chemicon) and a

HRP-conjugated secondary antibody. The spectrin breakdown product specific to calpain

mediated proteolysis (145 kDa) was analyzed using the Fuji LAS-400 Mini System.

Immunoreactivity of calpain-specific spectrin breakdown product was normalized to α-

tubulin to as a loading and transfer control. Data are expressed as a percentage of

vehicle-saline treated animals.

Glutamine Synthetase Protein Level Measurement:

2 hours after METH or saline treatment rats were killed by rapid decapitation and striata

were dissected. Tissue was homogenized in RIPA buffer (1% Igepal CA-630, 0.5%

Sodium Deoxycholate, and 0.1% Sodium Dodecyl Sulfate) containing Halt Protease

Inhibitor Cocktail (Thermo Scientific). A Bradford assay was used to determine protein content and samples were diluted (1:4) with LDS sample buffer. 5 µg of protein was separated via electrophoresis and transferred to a PVDF membrane. Glutamine

Synthetase levels were examined using a primary antibody to Glutamine Synthetase

(MAB302, Chemicon) and a HRP-conjugated secondary antibody. The single band present at 45 kDa was analyzed using the Fuji LAS-400 Mini System. Immunoreactivity

69

of glutamine synthetase was normalized to α-tubulin to as a loading and transfer control.

Data are expressed as a percentage of vehicle-saline treated animals.

Glutamine Synthetase Activity Assay:

Glutamine synthetase activity was measured using the glutamyltransferase activity of the

enzyme which catalyzes the reaction producing γ-glutamylhydroxymate from glutamine and hydroxylamine (Webb and Brown, 1976). 2 hours after METH or saline treatment rats were killed by rapid decapitation and striata were dissected. The striatum from a single hemisphere was homogenized in 400 μL of ice cold RIPA buffer (1% Igepal CA-

630, 0.5% Sodium Deoxycholate, and 0.1% Sodium Dodecyl Sulfate). Protease inhibitor

was not used because it alone produced detectable product that was not dependent on

glutamine synthetase activity. RIPA buffer alone does not produce any product. 50 μL

of tissue homogenate was added to the reaction mixture which contained 60 mM L-

glutamine, 15 mM hydroxylamine-HCl, 20 mM Sodium Arsenite, 0.4 mM ADP, 3 mM

Manganese Chloride, and 60 mM Imidazole-HCl (pH 6.8) to a total volume of 500 μL.

After combining tissue with the reaction mixture, the reaction ran for 30 minutes at 37

°C. After 30 minutes, the reaction was stopped by addition of 0.37 mM Iron Chloride,

0.67 mM HCl and 0.2 mM trichloracetic acid. The solution was then centrifuged at

14,000 x g for 10 minutes to separate protein. Product formation in the supernatant was measured and quantified by measuring the absorbance at 500 nm and using a standard curve of γ-glutamylhydroxymate (Sigma-Aldrich). To determine the amount of product formation that was specific to glutamine synthetase activity the reaction was run for each sample with and without the arsenite cofactor. Product formation in the absence of arsenite for each sample was subtracted from the product formation with arsenite to

70

determine the glutamine synthetase-dependent product formation. On average, about

20% of the product formation from tissue was not glutamine synthetase dependent.

Product formation was also normalized to protein concentration of each sample, which was determined using a Bradford assay. Data are expressed as a percentage of vehicle-

saline treated controls

Statistical Analysis:

Analysis of tissue content of dopamine and 5HT in the striatum, and spectrin proteolysis

was performed using a two-way ANOVA followed by post hoc Tukey multiple

comparison tests or t-tests. These tests were performed using SigmaPlot 11.0 Software

(Systat Software, Inc. SigmaPlot for Windows). Statistical analysis of brain glutamate

levels was carried out using a repeated measures ANOVA test, with each time point as a

within-subjects factor and ammonia, lactulose, or METH treatment as between-subjects factors. These tests were performed using SPSS Statistics 17.0 Software (IBM

Corporation). All data are presented as Mean±Standard Error of the Mean (SEM). α level in all studies is 0.05 or less and sample sizes were chosen to result in a power of

80% or greater.

4.3 Results:

Effect of AMPA Receptor Antagonism on Local Administration of METH and

Ammonia

To identify a mechanism by which ammonia and METH may contribute to striatal damage, ammonia (570 µM)+ METH (100 µM), and/or GYKI 52466 (100 µM) was reverse dialyzed for 8 hrs and the tissue adjacent to the microdialysis probe was dissected

7 days later for measurement of dopamine and 5-HT tissue content. GYKI 52466 is an

71

AMPA receptor antagonist that has been shown to prevent METH-induced increases in calpain-mediated spectrin proteolysis (Staszewski and Yamamoto, 2006). The effect of

GYKI 52466 on METH- and ammonia-induced decreases in dopamine and 5HT tissue

content was determined. A two-way ANOVA revealed a significant interaction between

the combination of METH +ammonia, and GYKI 52466 treatment on dopamine tissue

content (F(1,39)=3.392, p<0.05). A post hoc t-test revealed that there is a significant effect

of 52466 GYKI treatment on the METH+ammonia-induced depletions in dopamine

tissue content (t=3.298, p<0.05) (Figure 4.1A). Similarly, when the effects of GYKI

52466 on METH+ammonia-induced depletions in 5HT were examined, a two-way

ANOVA revealed a significant interaction between the combination of METH+ammonia,

and GYKI 52466 treatment (F(1,39)=3.624, p<0.05). A post-hoc Tukey test revealed a

significant effect of GYKI 52466 treatment on METH and ammonia-induced depletions

in 5HT content (q=5.541, p<0.05) (Figure 4.1B). Thus, GYKI 52466 blocked the

decreases in DA and 5HT content produced by ammonia+METH.

Effect of Locally Administered Ammonia on Extracellular Glutamate

To examine if the increases in brain ammonia seen during systemic METH treatment

were capable of altering extracellular glutamate levels, 200 µM of ammonia, which is the

striatal concentration measured during systemic METH treatment, was administered into

the striatum for 8 hours using reverse dialysis and changes in extracellular glutamate

were measured (Figure 4.2). A repeated measures ANOVA shows that ammonia

produced significant elevations in extracellular glutamate, compared to animals reverse

dialysed with artificial cerebrospinal fluid throughout all time points (F(1,13)= 13.531,

72

p<0.005). These increases represent an approximate 2-3-fold increase in extracellular

glutamate.

Effect of Ammonia on Systemic-METH-Induced Increases in Extracellular

Glutamate

To determine to what degree ammonia contributes to the increases in extracellular glutamate produced during systemic METH treatment, rats were treated with lactulose

(5.3 g/kg p.o. every 12 hours) or vehicle (8mL/kg) and treated with METH (10 mg/kg i.p. every 2 hours x 4 injections) or saline (1 mL/kg i.p. every 2 hrs x 4). Extracellular glutamate was measured during treatment with METH or Saline using in-vivo microdialysis (Figure 4.3). Baseline glutamate levels in all groups were 1365.24±183.69 pg of glutamate. A repeated measures ANOVA shows a main effect of METH treatment

(F(7,167)=5.033, p<0.050) and a significant interaction between METH and lactulose

treatment (F(7,167)=3.295 p<0.050). Striatal glutamate levels were significantly elevated

in METH treated animals at the 6 hour time point (F(3,20)=4.768 p<0.050). These data

demonstrate that increasing peripheral ammonia excretion with lactulose fully attenuates

METH-induced elevations in glutamate.

Effect of Ammonia on Calpain-Mediated Spectrin Proteolysis

Calpain is a calcium activated protease that produces a specific spectrin breakdown

product at 145 kDa and is a marker for excitotoxic damage (Harris and Morrow, 1988,

Staszewski and Yamamoto, 2006). To examine the contribution of ammonia to METH-

induced excitotoxic terminal damage, the spectrin breakdown product at 145 kDa was

measured in the striatum of rats treated with lactulose (5.3 g/kg p.o. every 12 hours) or

vehicle (8mL/kg) and treated with METH (10 mg/kg i.p. every 2 hours x 4 injections) or

73

saline (1 mL/kg i.p. every 2 hrs x 4) 5 days after treatment with METH or Saline. METH

produced 123.4 ± 4.9% increase in calpain-specific spectrin breakdown, which was

completed blocked by lactulose treatment. A two-way ANOVA revealed a significant interaction between METH and lactulose treatment (F(3,31)=4.436, p<0.05). Post-hoc tukey tests indicate that METH has a significant effect on calpain-specific spectrin breakdown (q= 4.927, p<0.005) and this effect is blocked by lactulose treatment

(q=4.364, p<0/05) (Figure 4.4A). A representative western blot for spectrin and α- tubulin immunoreactivity is also shown (Figure 4.4B)

Effect of METH on Glutamine Synthetase Protein Levels

To examine if METH produces acute decreases in glutamine synthetase expression, rats were treated with METH (10 mg/kg i.p. every 2 hours x 4 injections) or saline (1 mL/kg i.p. every 2 hrs x 4) and glutamate synthetase immunoreactivty was quantified in the striatum at 2 hours after drug treatment. Glutamine synthetase immunoreactivity did not significantly differ between groups treated with METH or saline (Figure 4.5A). A representative western blot for glutamine synthetase and α-tubulin immunoreactivity

(Figure 4.5B) is also shown.

Effect of METH on Glutamine Synthetase Activity

To determine if METH-induced produces acute decreases in glutamine synthetase activity, rats were treated with METH (10 mg/kg i.p. every 2 hours x 4 injections) or saline (1 mL/kg i.p. every 2 hrs x 4). At 2 hours after drug treatment, glutamate synthetase activity was quantified in the striatum using an assay that measures glutamine synthetase mediated γ-glutamylhydroxmate (γ-GH) formation. Both total γ-GH (Figure

74

4.6A) and γ-GH specific to glutamine synthetase activity (Figure 4.6B) were quantified and METH did not have a significant effect on glutamine synthetase activity.

75

Figure 4.1: The Dopamine and Serotonin Terminal Damage Produced by the Combination of NH3 and METH Requires AMPA Receptor Activation: Rats were reverse dialyzed with METH (100 µM), Ammonia (570 µM), and/or GKYI 52466 (100 µM) in the striatum for 8 hrs and dopamine and 5HT tissue content of the striatal tissue around the probe membrane (1 mm) was measured 7 days later. A) Treatment with GYKI 52466 significantly attenuates depletions in dopamine tissue content produced by METH and ammonia. B) Treatment with GKYI 52466 significantly attenuates depletions in 5HT tissue content produced by the combination of METH and ammonia. (* p<0.05 compared to aCSF, & p<0.05 compared to Ammonia METH) (n=10 rats per group) aCSF=artificial cerebrospinal fluid, GYKI=GYKI 52466

76

Figure 4.2: Effect of Local Ammonia Administration on Extracellular Glutamate Levels: Artificial cerebrospinal fluid (aCSF) or NH3 (570 µM) was reverse dialysed into the striatum for 8 hours and extracellular glutamate was measured. Reverse dialysis of NH3 significantly elevates striatal glutamate levels (*p<0.05). (n=6-8 rats per group)

77

Figure 4.3: Effect of Ammonia on Systemic METH-induced Increases in Extracellular Glutamate: Rats were treated with METH (10 mg/kg x 4 every 2 hrs) or saline (1 mL/kg x4 every 2 hrs) and Lactulose (5.3 g/kg every 12 hrs) or vehicle (8 mL/kg every 12 hrs). During METH treatment (arrows denote injections), striatal glutamate concentration was monitored using in-vivo microdialysis. Vehicle+METH treatment significantly increases brain glutamate concentrations starting at timepoint 6. Lactulose pretreatment blocks the increase. Baseline=1345.05 ± 162.55 pg/20 µL dialysate (*p<0.05). (n=8-10 rats per group).

78

A)

B)

Figure 4.4: Effect of Ammonia on METH-induced Increases in Calpain-Mediated Spectrin Proteolysis: Rats were treated METH (10 mg/kg x 4 every 2 hrs) or saline (1 mL/kg x4 every 2 hrs) and Lactulose (5.3 g/kg every 12 hrs) or vehicle (8 mL/kg every 12 hours) and killed 5 days after drug treatment A) Laculose prevents METH induced increases in the spectrin breakdown product (SBP) which is specific for calpain proteolysis (* p< 0.05 compared to Veh Sal) (n=6-9 per group) B) Representative western blot for spectrin breakdown products and α-tubulin loading control.

79

Figure 4.5: Effect of METH on Glutamine Synthetase Expression: Rats were treated with METH (10 mg/kg x 4 every 2 hrs) or saline (1 mL/kg x4 every 2 hrs) and glutamine synthetase immunoreactivity was measured at 2 hours after treatment. A) METH treatment had no significant effect on glutamine synthetase immunoreactivity. (n=8 per group) B) Representative western blot for glutamine synthetase and α-tubulin loading control. (GS=glutamine synthetase)

80

Figure 4.6: Effect of METH on Glutamine Synthetase Activity: Rats were treated with METH (10 mg/kg x 4 every 2 hrs) or saline (1 mL/kg x4 every 2 hrs) and glutamine synthetase activity was measured using an assay for γ glutamylhydroxymate formation. METH treatment had no significant effect on total γ-glutamyltransferase activity from tissue or γ-glutamyltransferase activity specific to glutamine synthetase. (n= 8 per group) (γ-GH= γ-glutamylhydroxymate)

81

4.4 Discussion:

These findings demonstrate that ammonia mediates METH neurotoxicity via

increases in extracellular glutamate and excitotoxicity. Activation of the AMPA

glutamate receptor was found to be required for the long term dopamine and serotonin

terminal damage produced by the direct administration of ammonia and METH.

Ammonia, at the brain concentrations measured during systemic METH exposure, was also found to be significant enough to alter local extracellular glutamate levels in the striatum. To examine to what degree ammonia affected increases in extracellular

glutamate seen during systemic METH treatment, rats were treated with lactulose to

block increases in brain ammonia during METH treatment. Blocking increases in brain

ammonia during METH treatment also blocked METH-induced increases in extracellular

glutamate, suggesting that increases in brain ammonia are required for increases in

glutamate. Consistent with these findings, ammonia was also found to be required for

METH-induced excitotoxicity, as measured by calpain-mediated spectrin proteolysis. To

determine a mechanism by which ammonia may contribute to METH-induced increases

in extracellular glutamate, we examined if METH treatment affected glutamine

synthetase protein levels and activity, yet found that neither was affected by METH

treatment. This suggests that the ammonia contributes to METH-induced increases in

glutamate through a mechanism other than effects on glutamine synthetase.

The neuronal damage produced by the local administration of ammonia with

METH into the striatum was blocked by the local administration of the AMPA

antagonist, GYKI52466 (Figure 4.1). This suggests that the combination of ammonia

and METH could activate presynaptic AMPA receptors through elevations in

82

extracellular glutamate. Systemic but not the central administration of METH treatment

has been shown to increase extracellular glutamate (Tata and Yamamoto, 2007).

Increased extracellular glutamate can then activate calcium-permeable AMPA receptors on striatal dopamine and 5HT terminals (Hollmann et al., 1991, Lai et al., 2003) resulting in both increased dopamine and 5HT release (Ohta et al., 1994), and excitotoxic terminal damage (Staszewski and Yamamoto, 2006). Ammonia has been shown to increase glutamate release from astrocytes (Gorg et al., 2010) and increase extracellular glutamate by decreasing glutamate transporter expression (Chan et al., 2000) and glutamine synthetase activity (Kosenko et al., 2003), all of which can combine with the effects of

METH on dopamine and 5HT terminals to produce long term 5HT and dopamine depletions.

To determine if ammonia was directly affecting extracellular glutamate levels, we reversed dialysed ammonia, at the same brain concentrations measured during systemic

METH treatment, directly into the striatum and measured extracellular glutamate. High concentrations of ammonia affect the processing of glutamate by astroctyes in vitro (Gorg et al., 2010), however it was unknown if ammonia alone, at the concentrations produced by systemic METH treatment, was capable of producing similar effects on extracellular glutamate. We found that reverse dialyzing ammonia into the brain for 8 hours produced a significant and consistent 2-3 fold increase in brain glutamate (Figure 4.2). This increase is comparable to the 4-5 fold increase in brain glutamate seen with systemic

METH treatment, yet suggests that systemic METH treatment contributes to increases extracellular glutamate through other mechanisms other than those mediated by ammonia alone. In addition to the effect of ammonia reported in the present study, METH-induced

83

increases in dopamine have been shown to contribute to increased presynaptic glutamate

release from corticostriatal projections as a result of D1-receptor mediated disinhibition

(Mark et al., 2004). Accordingly, it appears that METH-induced increases in brain ammonia are capable of contributing to drug-induced elevations in extracellular glutamate.

Interestingly, local administration of ammonia alone is not capable of producing the long term terminal dopamine and serotonin damage seen after systemic METH treatment although we show that ammonia alone is capable of increasing extracellular glutamate (Figure 3.7). This finding is consistent with previous reports that the direct administration of glutamate alone into the striatum does not produce long term depletions in dopamine tissue content (Tata and Yamamoto, 2007). The direct administration of either ammonia or glutamate into the striatum only produces monoaminergic terminal damage when combined with METH. Accordingly, it appears that increases in ammonia, via increases extracellular glutamate, as well as other effects of METH, especially increases in dopamine, interact synergistically to produce the long term monoaminergic terminal damage caused by the drug (Abekawa et al., 1994, Stephans and Yamamoto,

1994). This synergistic interaction explains why the damage produced by METH, which appears to be significantly mediated by global increases in ammonia, is selective for brain regions that contain the typical targets for METH, such as dopamine and serotonin transporters as well as glutamatergic projections.

To examine to what degree ammonia-induced elevations in glutamate are required for the increases in extracellular glutamate seen during systemic METH treatment, we blocked METH-induced increases in brain ammonia and found that this also fully

84

blocked METH-induced increases in extracellular glutamate (Figure 4.3). These findings

suggest that increases in ammonia are required for METH-induced increases in extracellular glutamate. Ammonia has been shown to increase glutamate through alteration of astrocytic uptake, metabolism, and release (Chan and Butterworth, 1999,

Kosenko et al., 2003, Gorg et al., 2010). This is important for extracellular glutamate levels because astrocytic reuptake and metabolism is the primary mechanism by which glutamate is removed from the synapse subsequent to presynaptic release (Benjamin and

Quastel, 1975, Danbolt, 2001). Although increased corticostriatal glutamate release subsequent to METH-induced increases in dopamine has been identified as a mechanism by which the drug increases glutamate (Stephans and Yamamoto, 1994), the current findings have identified another significant mechanism by which the drug contributes to increases in glutamate in a potentially dopamine-independent manner. Accordingly, these findings suggest that both dopamine-mediated increases in glutamate release from corticostriatal projections, as well an ammonia-dependent metabolism of glutamate in astrocytes, are required for METH-induced increases in extracellular glutamate.

To confirm if ammonia also significantly contributes to METH-induced excitotoxicity, we measured whether increases in brain ammonia were also required for increases in calpain-mediated spectrin proteolysis. When increases in brain ammonia were blocked during and after METH treatment with lactulose, this prevented METH induced increases in calpain-mediated spectrin proteolysis (Figure 4.4). Calpain is protease that targets essential cytoskeletal proteins, including spectrin, which is activated subsequent to calcium influx in the context of excitotoxicity (Siman et al., 1989). METH has previously been shown to increase calpain-mediated spectrin proteolysis in an AMPA

85

receptor activation dependent manner (Staszewski and Yamamoto, 2006). The finding

that ammonia is also required for METH-induced calpain-mediated spectrin proteolysis suggests that it is important for both increases in glutamate as well as excitotoxicity.

We also examined if systemic METH treatment affects glutamine synthetase protein levels or activity. Glutamine synthetase is an astrocytic enzyme responsible for metabolizing both ammonia and glutamate to produce glutamine. This enzyme has been shown to be decreased long-term after ammonia exposure, as well as after METH treatment in vitro (Stadlin et al., 1998, Kosenko et al., 2003). We examined glutamine synthetase protein levels and activity at 2 hours after METH treatment because altered enzyme activity at this time point would represent a mechanism by which altered astrocytic metabolism of glutamate may be contributing to acute increases in glutamate.

We found that neither glutamine synthetase protein levels (Figure 4.5) nor activity

(Figure 4.6) was altered. This supports the conclusion that the acute ammonia-induced increases in glutamate observed subsequent to METH are not due to decreased glutamine synthetase protein levels or activity. Glutamine synthetase activity has been shown to decrease in response to nitration subsequent to oxidative damage (Gorg et al., 2007).

METH-induced oxidative damage affecting protein function has been reported at time points as early as 1 hour after the binge dosing regimen, however these increases in oxidative damage are reported in neurons and are likely the result of increases in intracellular dopamine and nitric oxide formation (LaVoie and Hastings, 1999a, b,

Eyerman and Yamamoto, 2007). As glutamine synthetase is an astrocytic enzyme the lack of an effect on this protein suggests that METH-induced oxidative damage, nitric oxide formation, and possibly increases in intracellular dopamine may be specific to

86

terminals at early time points whereas the effects of ammonia on non-neuronal cells such as astrocytes are more protracted based on evidence that increases in glial

fibrillary acidic protein (GFAP) are only observed days after drug exposure (Hess et al.,

1990, O'Callaghan and Miller, 1994, Northrop and Yamamoto, 2012). This finding also

suggests a different mechanism is responsible for the contribution ammonia to METH

induced increases in glutamate. Ammonia may be affecting astrocytic glutamate

reuptake, upstream of glutamate synthase at the level of the excitatory amino acid

transporters. Although ammonia has been shown to affect transporter expression at later

time points after exposure, it may also affect the function of the transporter more acutely

by altering the ionic gradients that are responsible for determining the directionality of

the transporter (Barbour et al., 1988, Szatkowski et al., 1990, Chan and Butterworth,

1999, Grewer et al., 2008). Alteration of directionality of the transporter would prevent

the removal of glutamate from the synapse, allow glutamate within astrocytes to be

released, and contribute to increases in extracellular glutamate.

Overall, these findings identify ammonia as a key mediator for METH-induced

increases in glutamate and excitotoxicity. These findings have broader significance as

they demonstrate that acute increases in brain ammonia contribute to increases in

extracellular glutamate, suggesting ammonia may function as small molecule modulator

of excitatory neurotransmission, as well as a significant contributor to excitotoxicity.

These findings may also have significance in the context of other disorders where

excitotoxicity is believed to play a role including Alzheimer’s disease, ischemic stroke,

Huntington’s chorea and others (Choi, 1988).

87

Chapter 5:

General Discussion of Findings

5.1 Summary of Findings:

We hypothesized that the effects of METH on peripheral organ function contribute to the established striatal dopamine and serotonin terminal damage produced by the drug. The findings in this dissertation support the conclusion that METH-induced

liver damage and increases in ammonia play a significant role in the neurotoxicity of the

drug. We demonstrate that METH produces significant acute, liver damage that persists

for at least 24 hours after drug exposure. This liver damage appears to result from

METH-induced hyperthermia. Concurrent with this liver damage, the drug also produces

significant increases in both plasma and brain ammonia. These increases in ammonia

synergize with METH to contribute to the long term striatal dopamine and serotonin

terminal damage produced by the drug. Ammonia appears to contribute significantly to

this neuronal damage through increases in extracellular glutamate and excitotoxic

damage. These findings identify ammonia as a novel, peripherally-derived mediator of

the neurotoxicity of METH (Figure 5.1).

88

Figure 5.1: Summary of Findings: We have examined the role that METH-induced peripheral organ damage plays in the neurotoxicity of the drug. METH produced acute, hyperthermia-dependent liver damage which contributes to increases in plasma ammonia. Increases in brain ammonia synergize with METH to produce long-term dopamine and serotonin terminal damage though increases in glutamate and subsequent excitotoxicity.

89

5.2 General Discussion

In chapter 2, we established that METH produces alterations in hepatocellular morphology and increases in AST and ALT that persist for at least 24 hours after a binge drug exposure. These findings support that METH exposure causes acute liver damage

(Ozer et al., 2008). METH-induced hyperthermia was also shown to play a significant role in this liver damage as the changes in morphology and increases in AST and ALT were prevented when drug-induced hyperthermia was blocked. Prior to these studies, liver damage after METH had been noted in clinical reports, but the liver damage produced by the drug had yet to be extensively studied (Smith and Fischer, 1970, Kamijo et al., 2002). The findings in this dissertation illustrate for the first time that the drug produces significant and persistent damage to the liver. Because liver function is critical for many aspects of physiology and metabolism, it prompted us to hypothesize liver damage in mediating the neurotoxicity of the drug. Beyond the liver, METH-induced hyperthermia may contribute to the damage of other peripheral organs, warranting the study of the contribution of METH-induced hyperthermia to the damage of other peripheral organs reported to be affected by METH, including the renal, cardiovascular and muscular systems (Kamijo et al., 2002, Wijetunga et al., 2003, Ago et al., 2006,

Yarmolenko et al., 2011).

The liver is essential in the metabolism of ammonia and drove us to hypothesize that the METH-induced liver damage reported in Chapter 2 may affect ammonia metabolism (Felipo and Butterworth, 2002). Therefore, we examined if plasma and brain ammonia levels were elevated at the same time points as the liver damage and

90 found that METH also produces significant elevations in ammonia (Chapter 3). We also found that when METH-induced liver damage was prevented by blocking hyperthermia, this also prevented increases in ammonia and further strengthened the link between

METH-induced liver damage and increases in ammonia. To examine if these increases in ammonia contributed to the dopamine and serotonin terminal damage produced by

METH, we blocked drug-induced increases in ammonia using lactulose and found that this also attenuated the long-term neurotoxicity of the drug. To determine if ammonia was contributing directly to the neurotoxicity of METH, we directly administered ammonia alone or the combination of ammonia and METH to the striatum in concentrations similar to those measured during systemic METH treatment. We found that only the combination of METH and ammonia was able to produce the long-term dopamine and serotonin terminal damage seen after systemic METH treatment, suggesting that ammonia acts as a peripherally derived mediator of METH-neurotoxicity and identifying ammonia as a novel mediator of the neurotoxicity of METH. This suggests that ammonia alone is not capable of producing neuronal damage but can synergize with other toxic compounds to contribute to neurotoxicity. This highlights the potentially important role of acute liver damage and previously assumed subclinical increases in ammonia in mediating other neuropsychiatric disorders (Swain et al., 1992,

Clemmesen et al., 1999). In the context of METH-neurotoxicity, these findings also suggest that disorders which compromise liver function, including cirrhosis and hepatitis, which are prevalent in poly-substance and intravenous drug abusers, may exacerbate the neurotoxic consequences of METH (Bao et al., 2010, Gao and Bataller, 2011, Parikh et al., 2012).

91

We have also examined how increases in brain ammonia contribute to the neurotoxicity of METH (Chapter 4). We examined if the activation of calcium permeable AMPA receptors contributed to the dopamine and serotonin terminal damage produced by the direct administration of METH and ammonia into the striatum and found that when these receptors were blocked, the damage produced by the combination of ammonia and METH was prevented. We also found that ammonia, at the brain concentrations produced by systemic METH treatment, was adequate to increase local glutamate levels, and that during systemic METH treatment, ammonia was required for

METH-induced increases in extracellular glutamate and the subsequent excitotoxic terminal damage produced by the drug. These findings suggest that ammonia may synergize with METH to increase extracellular glutamate and cause excitotoxicity to dopamine and serotonin terminals. These findings are significant because they determine a mechanism by which ammonia plays a significant role in METH neurotoxicity.

Beyond this, identification of ammonia as a significant mediator of excitotoxic terminal damage suggests that it may play a role in other disorders where excitotoxicity has been identified as a mechanism of neuronal damage including stroke, seizures, Alzheimer ’s disease and Huntington’s disease (Choi, 1988). Beyond suggesting a role for ammonia in pathologic conditions, these findings also suggest a physiological role for ammonia in excitatory neurotransmission. In addition to being a metabolic intermediate, these findings support that ammonia may significantly modulate glutamate levels and neurotransmission, even at moderate concentrations in the brain. Although we have yet to determine how ammonia is affecting glutamate concentrations in the brain, we hypothesize that ammonia may be affecting the local reuptake or release of glutamate

92

within astrocytes (Barbour et al., 1988, Szatkowski et al., 1990, Chan and Butterworth,

1999, Grewer et al., 2008, Gorg et al., 2010).

These current findings also have important implications for the acute treatment of

METH exposure. Recommendations for the acute management of METH exposure focus

on reducing patient agitation, tachycardia, tachypnea, and hypertension (Richards et al.,

1997). The findings in this dissertation support the proposition that hyperthermia, liver

damage, and increases in ammonia should also be addressed during acute drug exposure

to reduce the neurotoxicity of the drug. Reducing the dopamine and serotonin terminal

damage produced by the drug is important because this damage has been associated with

cognitive deficits as well as likelihood of relapse in patients (Volkow et al., 2001, Wang

et al., 2012). Our current findings determined that METH-induced hyperthermia

contributes significantly to the liver damage and increases in ammonia seen after drug

exposure. Accordingly, reducing acute METH-induced hyperthermia may be important

for reducing the hepatotoxicity of the drug. We also found that treatment with lactulose

prevented drug induced increases in ammonia and striatal glutamate, as well as the long

term dopamine and serotonin terminal damage produced by METH. Lactulose is

currently used clinically for the treatment of hepatic encephalopathy in both in- and

outpatient settings (Al Sibae and McGuire, 2009). These findings suggest that treating

patients with lactulose during acute METH exposure may reduce the neurotoxicity of the

drug. Lactulose also has the potential for use in METH-dependent patients to reduce the neuronal damage caused by repeated drug use. Treatment with lactulose may reduce the dopamine and serotonin terminal damage caused by the drug and potentially reduce the neuropsychiatric consequences of drug exposure including impaired decision making,

93

reduced impulse control, and addiction (Rogers et al., 1999, Simon et al., 2002, Paulus et

al., 2005, Clark et al., 2006, Wang et al., 2012). While our findings support the clinical

use of lactulose to reduce the neurotoxicity of METH, one limitation with this treatment

is that lactulose, in our studies, as well as clinically, requires 24-48 hours of treatment to

reduce ammonia levels and neuronal damage (Al Sibae and McGuire, 2009). In addition

to lactulose, there are other fast-acting formulations which are intravenously administered

to rapidly reduce ammonia, such as ornithine phenylacetate (Jalan et al., 2007). This may

represent an even more effective inpatient intervention to reduce METH-induced

increases in ammonia and neurotoxicity.

Finally, these findings also broadly highlight the importance of examining the

connection between peripheral organ and neuronal damage. In contrast to the traditional

line of thinking that the neurotoxicity of METH is a result of the drug’s direct action on

the brain, these findings illustrate that liver damage and increases in peripheral ammonia

contribute significantly to the neurotoxicity of METH. In addition to identifying a

peripherally-derived mediator of toxicity, we have also established a peripherally-acting intervention (i.e. lactulose) for this neuronal damage. These findings support the importance of considering the periphery in the further understanding and development of novel interventions for neuronal damage. Beyond considering the role of peripherally derived ammonia in glutamate-mediated neuronal damage, changes in the peripheral organ function may also contribute significantly to neuronal inflammation, metabolism, blood flow and signaling via both hormones and . Accordingly, altered peripheral physiology may represent a significant contributing factor to many neuronal disorders, as well as a target for the treatment of these disorders.

94

Taken together, these findings support that peripherally-derived ammonia contributes to the neurotoxicity of METH through increases in extracellular glutamate and excitotoxicity. Overall, these findings identify a role for ammonia and peripheral organ damage in the neurotoxicity of METH and provide further evidence that peripheral organ damage can play a significant role in neuropsychiatric disorders.

95

References:

Abekawa T, Ohmori T, Koyama T (1994) Effects of repeated administration of a high dose of methamphetamine on dopamine and glutamate release in rat striatum and nucleus accumbens. Brain Res 643:276-281.

Adeva MM, Souto G, Blanco N, Donapetry C (2012) Ammonium metabolism in humans. Metabolism 61:1495-1511.

Ago M, Ago K, Hara K, Kashimura S, Ogata M (2006) Toxicological and histopathological analysis of a patient who died nine days after a single intravenous dose of methamphetamine: a case report. Leg Med (Tokyo) 8:235- 239.

Al Sibae MR, McGuire BM (2009) Current trends in the treatment of hepatic encephalopathy. Therapeutics and clinical risk management 5:617-626.

Albers DS, Sonsalla PK (1995) Methamphetamine-induced hyperthermia and dopaminergic neurotoxicity in mice: pharmacological profile of protective and nonprotective agents. The Journal of pharmacology and experimental therapeutics 275:1104-1114.

Arai S, Takuma K, Mizoguchi H, Ibi D, Nagai T, Takahashi K, Kamei H, Nabeshima T, Yamada K (2008) Involvement of pallidotegmental neurons in methamphetamine- and MK-801-induced impairment of prepulse inhibition of the acoustic startle reflex in mice: reversal by GABAB receptor baclofen. 33:3164-3175.

Bao YP, Liu ZM, Lu L (2010) Review of HIV and HCV infection among drug users in China. Current opinion in psychiatry 23:187-194.

Barbour B, Brew H, Attwell D (1988) Electrogenic glutamate uptake in glial cells is activated by intracellular potassium. Nature 335:433-435.

96

Batshaw ML, Hyman SL, Mellits ED, Thomas GH, DeMuro R, Coyle JT (1986) Behavioral and neurotransmitter changes in the urease-infused rat: a model of congenital hyperammonemia. Pediatr Res 20:1310-1315.

Batt AM, Ferrari L (1995) Manifestations of chemically induced liver damage. Clin Chem 41:1882-1887.

Battaglia G, Fornai F, Busceti CL, Aloisi G, Cerrito F, De Blasi A, Melchiorri D, Nicoletti F (2002) Selective blockade of mGlu5 metabotropic glutamate receptors is protective against methamphetamine neurotoxicity. J Neurosci 22:2135-2141.

Bell DS (1973) The experimental reproduction of amphetamine psychosis. Archives of general psychiatry 29:35-40.

Benamar K, Geller EB, Adler MW (2008) A new brain area affected by 3,4- methylenedioxymethamphetamine: A microdialysis-biotelemetry study. European journal of pharmacology 596:84-88.

Benjamin AM, Quastel JH (1975) Metabolism of amino acids and ammonia in rat brain cortex slices in vitro: a possible role of ammonia in brain function. J Neurochem 25:197-206.

Berridge CW (2006) Neural substrates of psychostimulant-induced arousal. Neuropsychopharmacology 31:2332-2340.

Betarbet R, Greenamyre JT (1999) Differential expression of glutamate receptors by the dopaminergic neurons of the primate striatum. Experimental neurology 159:401- 408.

Bianchi L, Ohnacker H, Beck K, Zimmerli-Ning M (1972) Liver damage in heatstroke and its regression. A biopsy study. Hum Pathol 3:237-248.

Billger M, Wallin M, Karlsson JO (1988) Proteolysis of tubulin and microtubule- associated proteins 1 and 2 by calpain I and II. Difference in sensitivity of assembled and disassembled microtubules. Cell Calcium 9:33-44.

Bowyer JF, Ali S (2006) High doses of methamphetamine that cause disruption of the blood-brain barrier in limbic regions produce extensive neuronal degeneration in mouse hippocampus. Synapse 60:521-532.

Bowyer JF, Davies DL, Schmued L, Broening HW, Newport GD, Slikker W, Jr., Holson RR (1994) Further studies of the role of hyperthermia in methamphetamine neurotoxicity. J Pharmacol Exp Ther 268:1571-1580.

Bowyer JF, Gough B, Slikker W, Jr., Lipe GW, Newport GD, Holson RR (1993) Effects of a cold environment or age on methamphetamine-induced dopamine release in the caudate putamen of female rats. Pharmacol Biochem Behav 44:87-98.

97

Bowyer JF, Robinson B, Ali S, Schmued LC (2008) Neurotoxic-related changes in tyrosine hydroxylase, microglia, myelin, and the blood-brain barrier in the caudate-putamen from acute methamphetamine exposure. Synapse 62:193-204.

Bowyer JF, Tank AW, Newport GD, Slikker W, Jr., Ali SF, Holson RR (1992) The influence of environmental temperature on the transient effects of methamphetamine on dopamine levels and dopamine release in rat striatum. The Journal of pharmacology and experimental therapeutics 260:817-824.

Breier JM, Bankson MG, Yamamoto BK (2006) L-tyrosine contributes to (+)-3,4- methylenedioxymethamphetamine-induced serotonin depletions. J Neurosci 26:290-299.

Brown JM, Quinton MS, Yamamoto BK (2005) Methamphetamine-induced inhibition of mitochondrial complex II: roles of glutamate and peroxynitrite. Journal of neurochemistry 95:429-436.

Brown JM, Riddle EL, Sandoval V, Weston RK, Hanson JE, Crosby MJ, Ugarte YV, Gibb JW, Hanson GR, Fleckenstein AE (2002) A single methamphetamine administration rapidly decreases vesicular dopamine uptake. J Pharmacol Exp Ther 302:497-501.

Brown PL, Bae D, Kiyatkin EA (2007) Relationships between locomotor activation and alterations in brain temperature during selective blockade and stimulation of dopamine transmission. Neuroscience 145:335-343.

Burrows KB, Gudelsky G, Yamamoto BK (2000a) Rapid and transient inhibition of mitochondrial function following methamphetamine or 3,4- methylenedioxymethamphetamine administration. Eur J Pharmacol 398:11-18.

Burrows KB, Nixdorf WL, Yamamoto BK (2000b) Central administration of methamphetamine synergizes with metabolic inhibition to deplete striatal monoamines. J Pharmacol Exp Ther 292:853-860.

Cadet JL, Jayanthi S, Deng X (2005) Methamphetamine-induced neuronal apoptosis involves the activation of multiple death pathways. Review. Neurotoxicity research 8:199-206.

Caldwell J, Dring LG, Williams RT (1972) Metabolism of ( 14 C)methamphetamine in man, the guinea pig and the rat. Biochem J 129:11-22.

Carvalho F, Remiao F, Soares ME, Catarino R, Queiroz G, Bastos ML (1997) d- Amphetamine-induced hepatotoxicity: possible contribution of catecholamines and hyperthermia to the effect studied in isolated rat hepatocytes. Archives of toxicology 71:429-436.

98

Carvalho M, Carvalho F, Bastos ML (2001) Is hyperthermia the triggering factor for hepatotoxicity induced by 3,4-methylenedioxymethamphetamine (ecstasy)? An in vitro study using freshly isolated mouse hepatocytes. Archives of toxicology 74:789-793.

Cauli O, Rodrigo R, Piedrafita B, Boix J, Felipo V (2007) Inflammation and hepatic encephalopathy: ibuprofen restores learning ability in rats with portacaval shunts. Hepatology 46:514-519.

Chan H, Butterworth RF (1999) Evidence for an astrocytic glutamate transporter deficit in hepatic encephalopathy. Neurochem Res 24:1397-1401.

Chan H, Hazell AS, Desjardins P, Butterworth RF (2000) Effects of ammonia on glutamate transporter (GLAST) protein and mRNA in cultured rat cortical astrocytes. Neurochem Int 37:243-248.

Chan P, Di Monte DA, Luo JJ, DeLanney LE, Irwin I, Langston JW (1994) Rapid ATP loss caused by methamphetamine in the mouse striatum: relationship between energy impairment and dopaminergic neurotoxicity. Journal of neurochemistry 62:2484-2487.

Chan PH, Fishman RA, Longar S, Chen S, Yu A (1985) Cellular and molecular effects of polyunsaturated fatty acids in brain ischemia and injury. Prog Brain Res 63:227- 235.

Chen JP (2007) Methamphetamine-associated acute myocardial infarction and cardiogenic shock with normal coronary arteries: refractory global coronary microvascular spasm. J Invasive Cardiol 19:E89-92.

Choi DW (1988) Glutamate neurotoxicity and diseases of the nervous system. Neuron 1:623-634.

Choi DW (1994) Calcium and excitotoxic neuronal injury. Annals of the New York Academy of Sciences 747:162-171.

Cisneros IE, Ghorpade A (2012) HIV-1, methamphetamine and astrocyte glutamate regulation: combined excitotoxic implications for neuro-AIDS. Curr HIV Res 10:392-406.

Clark L, Robbins TW, Ersche KD, Sahakian BJ (2006) Reflection impulsivity in current and former substance users. Biological psychiatry 60:515-522.

Clemmesen JO, Larsen FS, Kondrup J, Hansen BA, Ott P (1999) Cerebral herniation in patients with acute liver failure is correlated with arterial ammonia concentration. Hepatology 29:648-653.

99

Commins DL, Vosmer G, Virus RM, Woolverton WL, Schuster CR, Seiden LS (1987) Biochemical and histological evidence that methylenedioxymethylamphetamine (MDMA) is toxic to neurons in the rat brain. J Pharmacol Exp Ther 241:338-345.

Conant K, Lim ST, Randall B, Maguire-Zeiss KA (2012) Matrix metalloproteinase dependent cleavage of cell adhesion molecules in the pathogenesis of CNS dysfunction with HIV and methamphetamine. Curr HIV Res 10:384-391.

Cruickshank CC, Dyer KR (2009) A review of the clinical pharmacology of methamphetamine. Addiction 104:1085-1099.

Danbolt NC (2001) Glutamate uptake. Progress in neurobiology 65:1-105.

Dawson VL, Dawson TM (1996) Nitric oxide neurotoxicity. J Chem Neuroanat 10:179- 190.

Deng X, Cadet JL (1999) Methamphetamine administration causes overexpression of nNOS in the mouse striatum. Brain research 851:254-257.

Doyle JR, Yamamoto BK (2010) Serotonin 2 receptor modulation of hyperthermia, corticosterone, and hippocampal serotonin depletions following serial exposure to chronic stress and methamphetamine. Psychoneuroendocrinology 35:629-633.

Dugan LL, Sensi SL, Canzoniero LM, Handran SD, Rothman SM, Lin TS, Goldberg MP, Choi DW (1995) Mitochondrial production of in cortical neurons following exposure to N-methyl-D-aspartate. The Journal of neuroscience : the official journal of the Society for Neuroscience 15:6377-6388.

Dumuis A, Sebben M, Haynes L, Pin JP, Bockaert J (1988) NMDA receptors activate the arachidonic acid cascade system in striatal neurons. Nature 336:68-70.

Dunah AW, Wang Y, Yasuda RP, Kameyama K, Huganir RL, Wolfe BB, Standaert DG (2000) Alterations in subunit expression, composition, and phosphorylation of striatal N-methyl-D-aspartate glutamate receptors in a rat 6-hydroxydopamine model of Parkinson's disease. Molecular pharmacology 57:342-352.

Escubedo E, Guitart L, Sureda FX, Jimenez A, Pubill D, Pallas M, Camins A, Camarasa J (1998) Microgliosis and down-regulation of adenosine transporter induced by methamphetamine in rats. Brain research 814:120-126.

Eugenin EA, Greco JM, Frases S, Nosanchuk JD, Martinez LR (2013) Methamphetamine Alters Blood Brain Barrier Protein Expression in Mice, Facilitating Central Nervous System Infection by Neurotropic Cryptococcus neoformans. J Infect Dis.

Eyerman DJ, Yamamoto BK (2007) A rapid oxidation and persistent decrease in the vesicular monoamine transporter 2 after methamphetamine. J Neurochem 103:1219-1227.

100

Fan P, Szerb JC (1993) Effects of ammonium ions on synaptic transmission and on responses to quisqualate and N-methyl-D-aspartate in hippocampal CA1 pyramidal neurons in vitro. Brain Res 632:225-231.

Fan XD, Li XM, Ashe PC, Juorio AV (1999) Lesion of the substantia nigra pars compacta downregulates striatal glutamate receptor subunit mRNA expression. Brain research 850:79-86.

Farfel GM, Vosmer GL, Seiden LS (1992) The N-methyl-D-aspartate antagonist MK-801 protects against serotonin depletions induced by methamphetamine, 3,4- methylenedioxymethamphetamine and p-chloroamphetamine. Brain Res 595:121- 127.

Felipo V, Butterworth RF (2002) Neurobiology of ammonia. Prog Neurobiol 67:259-279.

Flaum M, Schultz SK (1996) When does amphetamine-induced psychosis become schizophrenia? Am J Psychiatry 153:812-815.

Fleckenstein AE, Metzger RR, Gibb JW, Hanson GR (1997a) A rapid and reversible change in dopamine transporters induced by methamphetamine. Eur J Pharmacol 323:R9-10.

Fleckenstein AE, Metzger RR, Wilkins DG, Gibb JW, Hanson GR (1997b) Rapid and reversible effects of methamphetamine on dopamine transporters. J Pharmacol Exp Ther 282:834-838.

Fleckenstein AE, Wilkins DG, Gibb JW, Hanson GR (1997c) Interaction between hyperthermia and oxygen radical formation in the 5-hydroxytryptaminergic response to a single methamphetamine administration. The Journal of pharmacology and experimental therapeutics 283:281-285.

Fornstedt B, Brun A, Rosengren E, Carlsson A (1989) The apparent autoxidation rate of catechols in dopamine-rich regions of human increases with the degree of depigmentation of substantia nigra. J Neural Transm Park Dis Dement Sect 1:279- 295.

Fukami G, Hashimoto K, Koike K, Okamura N, Shimizu E, Iyo M (2004) Effect of antioxidant N-acetyl-L-cysteine on behavioral changes and neurotoxicity in rats after administration of methamphetamine. Brain Res 1016:90-95.

Gao B, Bataller R (2011) Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology 141:1572-1585.

Gluck MR, Moy LY, Jayatilleke E, Hogan KA, Manzino L, Sonsalla PK (2001) Parallel increases in lipid and protein oxidative markers in several mouse brain regions after methamphetamine treatment. J Neurochem 79:152-160.

101

Gonzales R, Mooney L, Rawson RA (2010) The methamphetamine problem in the United States. Annu Rev Public Health 31:385-398.

Gorg B, Morwinsky A, Keitel V, Qvartskhava N, Schror K, Haussinger D (2010) Ammonia triggers exocytotic release of L-glutamate from cultured rat astrocytes. 58:691-705.

Gorg B, Qvartskhava N, Voss P, Grune T, Haussinger D, Schliess F (2007) Reversible inhibition of mammalian glutamine synthetase by tyrosine nitration. FEBS Lett 581:84-90.

Gow AJ, Buerk DG, Ischiropoulos H (1997) A novel reaction mechanism for the formation of S-nitrosothiol in vivo. The Journal of biological chemistry 272:2841- 2845.

Gow AJ, Duran D, Malcolm S, Ischiropoulos H (1996) Effects of peroxynitrite-induced protein modifications on tyrosine phosphorylation and degradation. FEBS letters 385:63-66.

Graham DG (1978) Oxidative pathways for catecholamines in the genesis of neuromelanin and cytotoxic quinones. Mol Pharmacol 14:633-643.

Granado N, Ares-Santos S, O'Shea E, Vicario-Abejon C, Colado MI, Moratalla R (2010) Selective vulnerability in striosomes and in the nigrostriatal dopaminergic pathway after methamphetamine administration : early loss of TH in striosomes after methamphetamine. Neurotoxicity research 18:48-58.

Grewer C, Gameiro A, Zhang Z, Tao Z, Braams S, Rauen T (2008) Glutamate forward and reverse transport: from molecular mechanism to transporter-mediated release after ischemia. IUBMB Life 60:609-619.

Grippon P, Le Poncin Lafitte M, Boschat M, Wang S, Faure G, Dutertre D, Opolon P (1986) Evidence for the role of ammonia in the intracerebral transfer and metabolism of tryptophan. Hepatology 6:682-686.

Haberle J, Gorg B, Toutain A, Rutsch F, Benoist JF, Gelot A, Suc AL, Koch HG, Schliess F, Haussinger D (2006) Inborn error of amino acid synthesis: human glutamine synthetase deficiency. J Inherit Metab Dis 29:352-358.

Had-Aissouni L (2012) Toward a new role for plasma membrane sodium-dependent glutamate transporters of astrocytes: maintenance of antioxidant defenses beyond extracellular glutamate clearance. Amino Acids 42:181-197.

Hadamitzky M, Markou A, Kuczenski R (2011) Extended access to methamphetamine self-administration affects sensorimotor gating in rats. Behavioural brain research 217:386-390.

102

Haga N, Fujita N, Tsuruo T (2003) Mitochondrial aggregation precedes cytochrome c release from mitochondria during apoptosis. Oncogene 22:5579-5585.

Haga N, Fujita N, Tsuruo T (2005) Involvement of mitochondrial aggregation in arsenic trioxide (As2O3)-induced apoptosis in human glioblastoma cells. Sci 96:825-833.

Harris AS, Morrow JS (1988) Proteolytic processing of human brain alpha spectrin (fodrin): identification of a hypersensitive site. J Neurosci 8:2640-2651.

Harris D, Batki SL (2000) Stimulant psychosis: symptom profile and acute clinical course. Am J Addict 9:28-37.

Harris DS, Boxenbaum H, Everhart ET, Sequeira G, Mendelson JE, Jones RT (2003) The bioavailability of intranasal and smoked methamphetamine. Clinical pharmacology and therapeutics 74:475-486.

Haughey HM, Fleckenstein AE, Metzger RR, Hanson GR (2000) The effects of methamphetamine on serotonin transporter activity: role of dopamine and hyperthermia. Journal of neurochemistry 75:1608-1617.

Hawkins RA, Miller AL, Nielsen RC, Veech RL (1973) The acute action of ammonia on rat brain metabolism in vivo. Biochem J 134:1001-1008.

Hermens DF, Lubman DI, Ward PB, Naismith SL, Hickie IB (2009) Amphetamine psychosis: a model for studying the onset and course of psychosis. Med J Aust 190:S22-25.

Hernandez LF, Segovia G, Mora F (2003) Effects of activation of NMDA and AMPA glutamate receptors on the extracellular concentrations of dopamine, acetylcholine, and GABA in striatum of the awake rat: a microdialysis study. Neurochemical research 28:1819-1827.

Hess A, Desiderio C, McAuliffe WG (1990) Acute neuropathological changes in the caudate nucleus caused by MPTP and methamphetamine: immunohistochemical studies. Journal of neurocytology 19:338-342.

Hogan KA, Staal RG, Sonsalla PK (2000) Analysis of VMAT2 binding after methamphetamine or MPTP treatment: disparity between homogenates and vesicle preparations. J Neurochem 74:2217-2220.

Hollmann M, Hartley M, Heinemann S (1991) Ca2+ permeability of KA-AMPA--gated glutamate receptor channels depends on subunit composition. Science 252:851- 853.

Hong R, Matsuyama E, Nur K (1991) Cardiomyopathy associated with the smoking of crystal methamphetamine. JAMA 265:1152-1154.

103

Hotchkiss AJ, Gibb JW (1980) Long-term effects of multiple doses of methamphetamine on tryptophan hydroxylase and tyrosine hydroxylase activity in rat brain. J Pharmacol Exp Ther 214:257-262.

Huang YH, Tsai SJ, Su TW, Sim CB (1999) Effects of repeated high-dose methamphetamine on local cerebral utilization in rats. Neuropsychopharmacology 21:427-434.

Imam SZ, Islam F, Itzhak Y, Slikker W, Jr., Ali SF (2000) Prevention of dopaminergic neurotoxicity by targeting nitric oxide and peroxynitrite: implications for the prevention of methamphetamine-induced neurotoxic damage. Annals of the New York Academy of Sciences 914:157-171.

Jalan R, Wright G, Davies NA, Hodges SJ (2007) L-Ornithine phenylacetate (OP): a novel treatment for hyperammonemia and hepatic encephalopathy. Medical hypotheses 69:1064-1069.

Jayanthi S, Deng X, Bordelon M, McCoy MT, Cadet JL (2001) Methamphetamine causes differential regulation of pro-death and anti-death Bcl-2 genes in the mouse neocortex. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 15:1745-1752.

Jayanthi S, Deng X, Ladenheim B, McCoy MT, Cluster A, Cai NS, Cadet JL (2005) Calcineurin/NFAT-induced up-regulation of the Fas ligand/Fas death pathway is involved in methamphetamine-induced neuronal apoptosis. Proc Natl Acad Sci U S A 102:868-873.

Jayanthi S, Ladenheim B, Andrews AM, Cadet JL (1999) Overexpression of human copper/ superoxide dismutase in transgenic mice attenuates oxidative stress caused by methylenedioxymethamphetamine (Ecstasy). Neuroscience 91:1379- 1387.

Jayanthi S, Ladenheim B, Cadet JL (1998) Methamphetamine-induced changes in antioxidant enzymes and lipid peroxidation in copper/zinc-superoxide dismutase transgenic mice. Annals of the New York Academy of Sciences 844:92-102.

Jeng W, Ramkissoon A, Parman T, Wells PG (2006) Prostaglandin H synthase-catalyzed bioactivation of amphetamines to free radical intermediates that cause CNS regional DNA oxidation and nerve terminal degeneration. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 20:638-650.

Jia L, Zhang MH (2005) Comparison of probiotics and lactulose in the treatment of minimal hepatic encephalopathy in rats. World J Gastroenterol 11:908-911.

Johanson CE, Frey KA, Lundahl LH, Keenan P, Lockhart N, Roll J, Galloway GP, Koeppe RA, Kilbourn MR, Robbins T, Schuster CR (2006) Cognitive function 104

and nigrostriatal markers in abstinent methamphetamine abusers. Psychopharmacology (Berl) 185:327-338.

Johnson GV, Jope RS, Binder LI (1989) Proteolysis of tau by calpain. Biochemical and biophysical research communications 163:1505-1511.

Jones AL, Jarvie DR, McDermid G, Proudfoot AT (1994) Hepatocellular damage following amphetamine intoxication. J Toxicol Clin Toxicol 32:435-444.

Kadota T, Kadota K (2004) Neurotoxic morphological changes induced in the medial prefrontal cortex of rats behaviorally sensitized to methamphetamine. Archives of histology and cytology 67:241-251.

Kamijo Y, Soma K, Nishida M, Namera A, Ohwada T (2002) Acute liver failure following intravenous methamphetamine. Vet Hum Toxicol 44:216-217.

Katz NR (1989) Methods for the study of liver cell heterogeneity. Histochem J 21:517- 529.

Keefe KA, Zigmond MJ, Abercrombie ED (1993) In vivo regulation of extracellular dopamine in the neostriatum: influence of impulse activity and local excitatory amino acids. J Neural Transm Gen Sect 91:223-240.

Kew M, Bersohn I, Seftel H, Kent G (1970a) Liver damage in heatstroke. The American journal of medicine 49:192-202.

Kew M, Bersohn I, Seftel H, Kent G (1970b) Liver damage in heatstroke. Am J Med 49:192-202.

Kew MC, Minick OT, Bahu RM, Stein RJ, Kent G (1978) Ultrastructural changes in the liver in heatstroke. Am J Pathol 90:609-618.

Kim I, Oyler JM, Moolchan ET, Cone EJ, Huestis MA (2004) Urinary pharmacokinetics of methamphetamine and its metabolite, amphetamine following controlled oral administration to humans. Ther Drug Monit 26:664-672.

Kiyatkin EA, Sharma HS (2009) Acute methamphetamine intoxication brain hyperthermia, blood-brain barrier, brain edema, and morphological cell abnormalities. Int Rev Neurobiol 88:65-100.

Kosenko E, Kaminsky Y, Grau E, Minana MD, Grisolia S, Felipo V (1995) Nitroarginine, an inhibitor of nitric oxide synthetase, attenuates ammonia toxicity and ammonia-induced alterations in brain metabolism. Neurochem Res 20:451- 456.

105

Kosenko E, Kaminsky Y, Kaminsky A, Valencia M, Lee L, Hermenegildo C, Felipo V (1997) Superoxide production and antioxidant enzymes in ammonia intoxication in rats. Free Radic Res 27:637-644.

Kosenko E, Llansola M, Montoliu C, Monfort P, Rodrigo R, Hernandez-Viadel M, Erceg S, Sanchez-Perez AM, Felipo V (2003a) Glutamine synthetase activity and glutamine content in brain: modulation by NMDA receptors and nitric oxide. Neurochemistry international 43:493-499.

Kosenko E, Llansola M, Montoliu C, Monfort P, Rodrigo R, Hernandez-Viadel M, Erceg S, Sanchez-Perez AM, Felipo V (2003b) Glutamine synthetase activity and glutamine content in brain: modulation by NMDA receptors and nitric oxide. Neurochem Int 43:493-499.

Kousik SM, Graves SM, Napier TC, Zhao C, Carvey PM (2011) Methamphetamine- induced vascular changes lead to striatal hypoxia and dopamine reduction. Neuroreport 22:923-928.

Krasnova IN, Justinova Z, Ladenheim B, Jayanthi S, McCoy MT, Barnes C, Warner JE, Goldberg SR, Cadet JL (2010) Methamphetamine self-administration is associated with persistent biochemical alterations in striatal and cortical dopaminergic terminals in the rat. PLoS One 5:e8790.

Kuczenski R, Everall IP, Crews L, Adame A, Grant I, Masliah E (2007) Escalating dose- multiple binge methamphetamine exposure results in degeneration of the neocortex and limbic system in the rat. Experimental neurology 207:42-51.

Kuhn DM, Aretha CW, Geddes TJ (1999) Peroxynitrite inactivation of tyrosine hydroxylase: mediation by sulfhydryl oxidation, not tyrosine nitration. J Neurosci 19:10289-10294.

Kuhn DM, Francescutti-Verbeem DM, Thomas DM (2006) Dopamine quinones activate microglia and induce a neurotoxic gene expression profile: relationship to methamphetamine-induced nerve ending damage. Annals of the New York Academy of Sciences 1074:31-41.

Kuhn DM, Geddes TJ (1999) Peroxynitrite inactivates tryptophan hydroxylase via sulfhydryl oxidation. Coincident nitration of enzyme tyrosyl residues has minimal impact on catalytic activity. J Biol Chem 274:29726-29732.

Ladenheim B, Krasnova IN, Deng X, Oyler JM, Polettini A, Moran TH, Huestis MA, Cadet JL (2000) Methamphetamine-induced neurotoxicity is attenuated in transgenic mice with a null mutation for interleukin-6. Molecular pharmacology 58:1247-1256.

106

Lai SK, Tse YC, Yang MS, Wong CK, Chan YS, Yung KK (2003) Gene expression of glutamate receptors GluR1 and NR1 is differentially modulated in striatal neurons in rats after 6-hydroxydopamine lesion. Neurochemistry international 43:639-653.

LaVoie MJ, Hastings TG (1999a) Dopamine quinone formation and protein modification associated with the striatal neurotoxicity of methamphetamine: evidence against a role for extracellular dopamine. J Neurosci 19:1484-1491.

LaVoie MJ, Hastings TG (1999b) Peroxynitrite- and nitrite-induced oxidation of dopamine: implications for nitric oxide in dopaminergic cell loss. J Neurochem 73:2546-2554.

Lee KS, Frank S, Vanderklish P, Arai A, Lynch G (1991) Inhibition of proteolysis protects hippocampal neurons from ischemia. Proc Natl Acad Sci U S A 88:7233- 7237.

Lehre KP, Levy LM, Ottersen OP, Storm-Mathisen J, Danbolt NC (1995) Differential expression of two glial glutamate transporters in the rat brain: quantitative and immunocytochemical observations. The Journal of neuroscience : the official journal of the Society for Neuroscience 15:1835-1853.

Letelier ME, Lopez-Valladares M, Peredo-Silva L, Rojas-Sepulveda D, Aracena P (2011) Microsomal oxidative damage promoted by acetaminophen metabolism. Toxicol In Vitro 25:1310-1313.

Lin LY, Di Stefano EW, Schmitz DA, Hsu L, Ellis SW, Lennard MS, Tucker GT, Cho AK (1997) Oxidation of methamphetamine and methylenedioxymethamphetamine by CYP2D6. Drug Metab Dispos 25:1059- 1064.

Liu SJ, Zukin RS (2007) Ca2+-permeable AMPA receptors in synaptic plasticity and neuronal death. Trends in neurosciences 30:126-134.

Llansola M, Rodrigo R, Monfort P, Montoliu C, Kosenko E, Cauli O, Piedrafita B, El Mlili N, Felipo V (2007) NMDA receptors in hyperammonemia and hepatic encephalopathy. Metab Brain Dis 22:321-335.

Lorez H (1981) Fluorescence histochemistry indicates damage of striatal dopamine nerve terminals in rats after multiple doses of methamphetamine. Life Sci 28:911-916.

Magistretti PJ (2006) Neuron-glia metabolic coupling and plasticity. J Exp Biol 209:2304-2311.

Mark KA, Soghomonian JJ, Yamamoto BK (2004) High-dose methamphetamine acutely activates the striatonigral pathway to increase striatal glutamate and mediate long- term dopamine toxicity. J Neurosci 24:11449-11456.

107

Martin WR, Sloan JW, Sapira JD, Jasinski DR (1971) Physiologic, subjective, and behavioral effects of amphetamine, methamphetamine, ephedrine, phenmetrazine, and methylphenidate in man. Clinical pharmacology and therapeutics 12:245-258.

Martins T, Baptista S, Goncalves J, Leal E, Milhazes N, Borges F, Ribeiro CF, Quintela O, Lendoiro E, Lopez-Rivadulla M, Ambrosio AF, Silva AP (2011) Methamphetamine transiently increases the blood-brain barrier permeability in the hippocampus: role of tight junction proteins and matrix metalloproteinase-9. Brain research 1411:28-40.

Martins T, Burgoyne T, Kenny BA, Hudson N, Futter CE, Ambrosio AF, Silva AP, Greenwood J, Turowski P (2013) Methamphetamine-induced nitric oxide promotes vesicular transport in blood-brain barrier endothelial cells. Neuropharmacology 65:74-82.

McBean GJ, Doorty KB, Tipton KF, Kollegger H (1995) Alteration in the glial cell metabolism of glutamate by kainate and N-methyl-D-aspartate. Toxicon 33:569- 576.

McCandless DW, Schenker S (1981) Effect of acute ammonia intoxication on energy stores in the cerebral reticular activating system. Exp Brain Res 44:325-330.

McCann UD, Szabo Z, Seckin E, Rosenblatt P, Mathews WB, Ravert HT, Dannals RF, Ricaurte GA (2005) Quantitative PET studies of the serotonin transporter in MDMA users and controls using [11C]McN5652 and [11C]DASB. Neuropsychopharmacology 30:1741-1750.

McCann UD, Wong DF, Yokoi F, Villemagne V, Dannals RF, Ricaurte GA (1998) Reduced striatal dopamine transporter density in abstinent methamphetamine and methcathinone users: evidence from positron emission tomography studies with [11C]WIN-35,428. J Neurosci 18:8417-8422.

McFadden LM, Hadlock GC, Allen SC, Vieira-Brock PL, Stout KA, Ellis JD, Hoonakker AJ, Andrenyak DM, Nielsen SM, Wilkins DG, Hanson GR, Fleckenstein AE (2012a) Methamphetamine self-administration causes persistent striatal dopaminergic alterations and mitigates the deficits caused by a subsequent methamphetamine exposure. J Pharmacol Exp Ther 340:295-303.

McFadden LM, Hunt MM, Vieira-Brock PL, Muehle J, Nielsen SM, Allen SC, Hanson GR, Fleckenstein AE (2012b) Prior methamphetamine self-administration attenuates serotonergic deficits induced by subsequent high-dose methamphetamine administrations. Drug Alcohol Depend 126:87-94.

McGregor C, Srisurapanont M, Jittiwutikarn J, Laobhripatr S, Wongtan T, White JM (2005) The nature, time course and severity of methamphetamine withdrawal. Addiction 100:1320-1329.

108

McKetin R, Kelly E, McLaren J (2006a) The relationship between crystalline methamphetamine use and methamphetamine dependence. Drug and alcohol dependence 85:198-204.

McKetin R, McLaren J, Lubman DI, Hides L (2006b) The prevalence of psychotic symptoms among methamphetamine users. Addiction 101:1473-1478.

Mendelson J, Uemura N, Harris D, Nath RP, Fernandez E, Jacob P, 3rd, Everhart ET, Jones RT (2006) Human pharmacology of the methamphetamine stereoisomers. Clinical pharmacology and therapeutics 80:403-420.

Messeri S, Messerini L, Vizzutti F, Laffi G, Marra F (2012) Glycogenic hepatopathy associated with type 1 diabetes mellitus as a cause of recurrent liver damage. Annals of hepatology 11:554-558.

Mizoguchi H, Arai S, Koike H, Ibi D, Kamei H, Nabeshima T, Kim HC, Takuma K, Yamada K (2009) Therapeutic potential of nicotine for methamphetamine- induced impairment of sensorimotor gating: involvement of pallidotegmental neurons. Psychopharmacology (Berl) 207:235-243.

Moon KH, Upreti VV, Yu LR, Lee IJ, Ye X, Eddington ND, Veenstra TD, Song BJ (2008) Mechanism of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy)- mediated mitochondrial dysfunction in rat liver. Proteomics 8:3906-3918.

Moroni F, Lombardi G, Carla V, Lal S, Etienne P, Nair NP (1986) Increase in the content of quinolinic acid in cerebrospinal fluid and frontal cortex of patients with hepatic failure. J Neurochem 47:1667-1671.

Mousseau DD, Baker GB, Butterworth RF (1997) Increased density of catalytic sites and expression of brain monoamine oxidase A in humans with hepatic encephalopathy. J Neurochem 68:1200-1208.

Nakajima A, Yamada K, Nagai T, Uchiyama T, Miyamoto Y, Mamiya T, He J, Nitta A, Mizuno M, Tran MH, Seto A, Yoshimura M, Kitaichi K, Hasegawa T, Saito K, Yamada Y, Seishima M, Sekikawa K, Kim HC, Nabeshima T (2004) Role of tumor necrosis factor-alpha in methamphetamine-induced drug dependence and neurotoxicity. The Journal of neuroscience : the official journal of the Society for Neuroscience 24:2212-2225.

Nash JF, Yamamoto BK (1992) Methamphetamine neurotoxicity and striatal glutamate release: comparison to 3,4-methylenedioxymethamphetamine. Brain Res 581:237- 243.

Nicaise C, Prozzi D, Viaene E, Moreno C, Gustot T, Quertinmont E, Demetter P, Suain V, Goffin P, Deviere J, Hols P (2008) Control of acute, chronic, and constitutive hyperammonemia by wild-type and genetically engineered Lactobacillus plantarum in rodents. Hepatology 48:1184-1192. 109

Northrop NA, Yamamoto BK (2012) Persistent neuroinflammatory effects of serial exposure to stress and methamphetamine on the blood-brain barrier. Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology 7:951-968.

O'Callaghan JP, Miller DB (1994) Neurotoxicity profiles of substituted amphetamines in the C57BL/6J mouse. J Pharmacol Exp Ther 270:741-751.

Ohta K, Araki N, Shibata M, Komatsumoto S, Shimazu K, Fukuuchi Y (1994) Presynaptic ionotropic glutamate receptors modulate in vivo release and metabolism of striatal dopamine, noradrenaline, and 5-hydroxytryptamine: involvement of both NMDA and AMPA/kainate subtypes. Neurosci Res 21:83- 89.

Ozer J, Ratner M, Shaw M, Bailey W, Schomaker S (2008) The current state of serum biomarkers of hepatotoxicity. Toxicology 245:194-205.

Parikh N, Nonnemacher MR, Pirrone V, Block T, Mehta A, Wigdahl B (2012) Substance abuse, HIV-1 and hepatitis. Current HIV research 10:557-571.

Parkes JL, Grieninger G (1985) Insulin, not glucose, controls hepatocellular glycogen deposition. A re-evaluation of the role of both agents in cultured liver cells. The Journal of biological chemistry 260:8090-8097.

Paulus MP, Tapert SF, Schuckit MA (2005) Neural activation patterns of methamphetamine-dependent subjects during decision making predict relapse. Archives of general psychiatry 62:761-768.

Perez-Reyes M, White WR, McDonald SA, Hicks RE, Jeffcoat AR, Hill JM, Cook CE (1991) Clinical effects of daily methamphetamine administration. Clinical neuropharmacology 14:352-358.

Pontieri FE, Crane AM, Seiden LS, Kleven MS, Porrino LJ (1990) Metabolic mapping of the effects of intravenous methamphetamine administration in freely moving rats. Psychopharmacology (Berl) 102:175-182.

Pourahmad J, Eskandari MR, Nosrati M, Kobarfard F, Khajeamiri AR (2010) Involvement of mitochondrial/lysosomal toxic cross-talk in ecstasy induced liver toxicity under hyperthermic condition. Eur J Pharmacol 643:162-169.

Ramirez SH, Potula R, Fan S, Eidem T, Papugani A, Reichenbach N, Dykstra H, Weksler BB, Romero IA, Couraud PO, Persidsky Y (2009) Methamphetamine disrupts blood-brain barrier function by induction of oxidative stress in brain endothelial cells. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 29:1933-1945.

110

Rasmussen N (2008) America's first amphetamine epidemic 1929-1971: a quantitative and qualitative retrospective with implications for the present. Am J Public Health 98:974-985.

Rautou PE, Cazals-Hatem D, Moreau R, Francoz C, Feldmann G, Lebrec D, Ogier-Denis E, Bedossa P, Valla D, Durand F (2008) Acute liver cell damage in patients with anorexia nervosa: a possible role of starvation-induced hepatocyte autophagy. Gastroenterology 135:840-848, 848 e841-843.

Ricaurte G, Bryan G, Strauss L, Seiden L, Schuster C (1985) Hallucinogenic amphetamine selectively destroys brain serotonin nerve terminals. Science 229:986-988.

Ricaurte GA, Guillery RW, Seiden LS, Schuster CR, Moore RY (1982) Dopamine nerve terminal degeneration produced by high doses of methylamphetamine in the rat brain. Brain Res 235:93-103.

Ricaurte GA, Schuster CR, Seiden LS (1980) Long-term effects of repeated methylamphetamine administration on dopamine and serotonin neurons in the rat brain: a regional study. Brain Res 193:153-163.

Richards JR, Derlet RW, Duncan DR (1997) Methamphetamine toxicity: treatment with a benzodiazepine versus a butyrophenone. Eur J Emerg Med 4:130-135.

Rogers RD, Everitt BJ, Baldacchino A, Blackshaw AJ, Swainson R, Wynne K, Baker NB, Hunter J, Carthy T, Booker E, London M, Deakin JF, Sahakian BJ, Robbins TW (1999) Dissociable deficits in the decision-making cognition of chronic amphetamine abusers, opiate abusers, patients with focal damage to prefrontal cortex, and tryptophan-depleted normal volunteers: evidence for monoaminergic mechanisms. Neuropsychopharmacology 20:322-339.

Rose C (2002) Increased extracellular brain glutamate in acute liver failure: decreased uptake or increased release? Metabolic brain disease 17:251-261.

Rothman RB, Baumann MH (2003) Monoamine transporters and psychostimulant drugs. Eur J Pharmacol 479:23-40.

Saez R, Llansola M, Felipo V (1999) Chronic exposure to ammonia alters pathways modulating phosphorylation of microtubule-associated protein 2 in cerebellar neurons in culture. J Neurochem 73:2555-2562.

SAMSHA (2012a) Drug Abuse Warning Network, 2010: National Estimates of Drug Related Emergency Department Visits: HHS Publication No. (SMA) 12-4733 DAWN Series D-38. Rockville, MD:Substance Abuse and Mental Health Services Adminstration.

111

SAMSHA (2012b) Results from the 2011 National Survey on Drug Use ahd Health: Detailed Tables: NSDUH Series H-44, HHS Publication No. (SMA) 12-4713. Rockville, MD: Substance Abuse and Mental Health Services Administration.

Santello M, Volterra A (2009) Synaptic modulation by astrocytes via Ca2+-dependent glutamate release. Neuroscience 158:253-259.

Sato H, Tamba M, Ishii T, Bannai S (1999) Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. The Journal of biological chemistry 274:11455-11458.

Saul R, Ghidoni JJ, Molyneux RJ, Elbein AD (1985) Castanospermine inhibits alpha- glucosidase activities and alters glycogen distribution in animals. Proc Natl Acad Sci U S A 82:93-97.

Saunders NR, Ek CJ, Habgood MD, Dziegielewska KM (2008) Barriers in the brain: a renaissance? Trends in neurosciences 31:279-286.

Schlaepfer WW, Lee C, Lee VM, Zimmerman UJ (1985) An immunoblot study of neurofilament degradation in situ and during calcium-activated proteolysis. Journal of neurochemistry 44:502-509.

Schmidt CJ, Ritter JK, Sonsalla PK, Hanson GR, Gibb JW (1985a) Role of dopamine in the neurotoxic effects of methamphetamine. J Pharmacol Exp Ther 233:539-544.

Schmidt CJ, Ritter JK, Sonsalla PK, Hanson GR, Gibb JW (1985b) Role of dopamine in the neurotoxic effects of methamphetamine. J Pharmacol Exp Ther 233:539-544.

Segovia G, Del Arco A, Mora F (1997) Endogenous glutamate increases extracellular concentrations of dopamine, GABA, and through NMDA and AMPA/kainate receptors in striatum of the freely moving rat: a microdialysis study. Journal of neurochemistry 69:1476-1483.

Seiden LS, Commins DL, Vosmer G, Axt K, Marek G (1988) Neurotoxicity in dopamine and 5-hydroxytryptamine terminal fields: a regional analysis in nigrostriatal and mesolimbic projections. Ann N Y Acad Sci 537:161-172.

Shappell SA, Kearns GL, Valentine JL, Neri DF, DeJohn CA (1996) Chronopharmacokinetics and chronopharmacodynamics of dextromethamphetamine in man. Journal of clinical pharmacology 36:1051-1063.

Sharma HS, Ali SF (2006) Alterations in blood-brain barrier function by morphine and methamphetamine. Annals of the New York Academy of Sciences 1074:198-224.

Sharma HS, Kiyatkin EA (2009) Rapid morphological brain abnormalities during acute methamphetamine intoxication in the rat: an experimental study using light and electron microscopy. J Chem Neuroanat 37:18-32.

112

Silva AP, Martins T, Baptista S, Goncalves J, Agasse F, Malva JO (2010) Brain injury associated with widely abused amphetamines: neuroinflammation, neurogenesis and blood-brain barrier. Curr Drug Abuse Rev 3:239-254.

Siman R, Noszek JC, Kegerise C (1989) Calpain I activation is specifically related to excitatory amino acid induction of hippocampal damage. The Journal of neuroscience : the official journal of the Society for Neuroscience 9:1579-1590.

Simon SL, Domier CP, Sim T, Richardson K, Rawson RA, Ling W (2002) Cognitive performance of current methamphetamine and cocaine abusers. Journal of addictive diseases 21:61-74.

Skibba JL, Powers RH, Stadnicka A, Cullinane DW, Almagro UA, Kalbfleisch JH (1991) Oxidative stress as a precursor to the irreversible hepatocellular injury caused by hyperthermia. Int J Hyperthermia 7:749-761.

Skibba JL, Stadnicka A, Kalbfleisch JH (1989) Hyperthermic liver toxicity: a role for oxidative stress. J Surg Oncol 42:103-112.

Smith DE, Fischer CM (1970) An analysis of 310 cases of acute high-dose methamphetamine toxicity in Haight-Ashbury. Clin Toxicol 3:117-124.

Sprague JE, Yang X, Sommers J, Gilman TL, Mills EM (2007) Roles of norepinephrine, free Fatty acids, thyroid status, and skeletal muscle uncoupling protein 3 expression in sympathomimetic-induced thermogenesis. The Journal of pharmacology and experimental therapeutics 320:274-280.

Squier MK, Miller AC, Malkinson AM, Cohen JJ (1994) Calpain activation in apoptosis. J Cell Physiol 159:229-237.

Sriram K, Miller DB, O'Callaghan JP (2006) Minocycline attenuates microglial activation but fails to mitigate striatal dopaminergic neurotoxicity: role of tumor necrosis factor-alpha. Journal of neurochemistry 96:706-718.

Stadlin A, Lau JW, Szeto YK (1998) A selective regional response of cultured astrocytes to methamphetamine. Ann N Y Acad Sci 844:108-121.

Stamler JS, Hausladen A (1998) Oxidative modifications in nitrosative stress. Nat Struct Biol 5:247-249.

Staszewski RD, Yamamoto BK (2006) Methamphetamine-induced spectrin proteolysis in the rat striatum. J Neurochem 96:1267-1276.

Stephans SE, Whittingham TS, Douglas AJ, Lust WD, Yamamoto BK (1998) Substrates of energy metabolism attenuate methamphetamine-induced neurotoxicity in striatum. Journal of neurochemistry 71:613-621.

113

Stephans SE, Yamamoto BK (1994) Methamphetamine-induced neurotoxicity: roles for glutamate and dopamine efflux. Synapse 17:203-209.

Sulzer D, Chen TK, Lau YY, Kristensen H, Rayport S, Ewing A (1995) Amphetamine redistributes dopamine from synaptic vesicles to the cytosol and promotes reverse transport. J Neurosci 15:4102-4108.

Sulzer D, Rayport S (1990) Amphetamine and other psychostimulants reduce pH gradients in midbrain dopaminergic neurons and chromaffin granules: a mechanism of action. Neuron 5:797-808.

Swain M, Butterworth RF, Blei AT (1992) Ammonia and related amino acids in the pathogenesis of brain edema in acute ischemic liver failure in rats. Hepatology 15:449-453.

Szatkowski M, Barbour B, Attwell D (1990) Non-vesicular release of glutamate from glial cells by reversed electrogenic glutamate uptake. Nature 348:443-446.

Tata DA, Yamamoto BK (2007) Interactions between methamphetamine and environmental stress: role of oxidative stress, glutamate and mitochondrial dysfunction. Addiction 102 Suppl 1:49-60.

Thomas DM, Dowgiert J, Geddes TJ, Francescutti-Verbeem D, Liu X, Kuhn DM (2004a) Microglial activation is a pharmacologically specific marker for the neurotoxic amphetamines. Neuroscience letters 367:349-354.

Thomas DM, Kuhn DM (2005a) Cyclooxygenase-2 is an obligatory factor in methamphetamine-induced neurotoxicity. The Journal of pharmacology and experimental therapeutics 313:870-876.

Thomas DM, Kuhn DM (2005b) MK-801 and dextromethorphan block microglial activation and protect against methamphetamine-induced neurotoxicity. Brain research 1050:190-198.

Thomas DM, Walker PD, Benjamins JA, Geddes TJ, Kuhn DM (2004b) Methamphetamine neurotoxicity in dopamine nerve endings of the striatum is associated with microglial activation. The Journal of pharmacology and experimental therapeutics 311:1-7.

UNODC (2012) World Drug Report. Vienna, Austria.

Valencia F, Bubar MJ, Milligan G, Cunningham KA, Bourne N (2012) Influence of methamphetamine on genital herpes simplex virus type 2 infection in a mouse model. Sexually transmitted diseases 39:720-725.

Vearrier D, Greenberg MI, Miller SN, Okaneku JT, Haggerty DA (2012) Methamphetamine: history, pathophysiology, adverse health effects, current

114

trends, and hazards associated with the clandestine manufacture of methamphetamine. Dis Mon 58:38-89.

Villa PG, Henzel WJ, Sensenbrenner M, Henderson CE, Pettmann B (1998) Calpain inhibitors, but not caspase inhibitors, prevent actin proteolysis and DNA fragmentation during apoptosis. J Cell Sci 111 ( Pt 6):713-722.

Volkow ND, Chang L, Wang GJ, Fowler JS, Franceschi D, Sedler M, Gatley SJ, Miller E, Hitzemann R, Ding YS, Logan J (2001a) Loss of dopamine transporters in methamphetamine abusers recovers with protracted abstinence. J Neurosci 21:9414-9418.

Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R, Ding YS, Logan J, Wong C, Miller EN (2001b) Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. The American journal of psychiatry 158:377-382.

Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R, Ding YS, Logan J, Wong C, Miller EN (2001c) Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. Am J Psychiatry 158:377-382.

Volkow ND, Fowler JS, Wang GJ, Shumay E, Telang F, Thanos PK, Alexoff D (2010) Distribution and pharmacokinetics of methamphetamine in the human body: clinical implications. PLoS One 5:e15269.

Wagner GC, Ricaurte GA, Seiden LS, Schuster CR, Miller RJ, Westley J (1980) Long- lasting depletions of striatal dopamine and loss of dopamine uptake sites following repeated administration of methamphetamine. Brain Res 181:151-160.

Wang AM, Suojanen JN, Colucci VM, Rumbaugh CL, Hollenberg NK (1990) Cocaine- and methamphetamine-induced acute cerebral vasospasm: an angiographic study in rabbits. AJNR Am J Neuroradiol 11:1141-1146.

Wang GJ, Smith L, Volkow ND, Telang F, Logan J, Tomasi D, Wong CT, Hoffman W, Jayne M, Alia-Klein N, Thanos P, Fowler JS (2012) Decreased dopamine activity predicts relapse in methamphetamine abusers. Molecular psychiatry 17:918-925.

Webb JT, Brown GW (1976) Some properties and occurrence of glutamine synthetase in fish. Comparative biochemistry and physiology B, Comparative biochemistry 54:171-175.

Weigand K, Riediger C, Stremmel W, Flechtenmacher C, Encke J (2007) Are heat stroke and physical exhaustion underestimated causes of acute hepatic failure? World journal of gastroenterology : WJG 13:306-309.

115

Wijetunga M, Seto T, Lindsay J, Schatz I (2003) Crystal methamphetamine-associated cardiomyopathy: tip of the iceberg? J Toxicol Clin Toxicol 41:981-986.

Wills EJ, Findlay JM, McManus JP (1976a) Effects of hyperthermia therapy on the liver. II. Morphological observations. J Clin Pathol 29:1-10.

Wills EJ, Findlay JM, McManus JP (1976b) Effects of hyperthermia therapy on the liver. II. Morphological observations. Journal of clinical pathology 29:1-10.

Woolfolk CA, Shapiro B, Stadtman ER (1966) Regulation of glutamine synthetase. I. Purification and properties of glutamine synthetase from Escherichia coli. Arch Biochem Biophys 116:177-192.

Woolverton WL, Ricaurte GA, Forno LS, Seiden LS (1989) Long-term effects of chronic methamphetamine administration in rhesus monkeys. Brain Res 486:73-78.

Xie T, McCann UD, Kim S, Yuan J, Ricaurte GA (2000) Effect of temperature on dopamine transporter function and intracellular accumulation of methamphetamine: implications for methamphetamine-induced dopaminergic neurotoxicity. J Neurosci 20:7838-7845.

Yamamoto BK, Pehek EA (1990) A neurochemical heterogeneity of the rat striatum as measured by in vivo electrochemistry and microdialysis. Brain Res 506:236-242.

Yamamoto BK, Raudensky J (2008) The role of oxidative stress, metabolic compromise, and inflammation in neuronal injury produced by amphetamine-related drugs of abuse. J Neuroimmune Pharmacol 3:203-217.

Yamamoto BK, Zhu W (1998) The effects of methamphetamine on the production of free radicals and oxidative stress. J Pharmacol Exp Ther 287:107-114.

Yarmolenko PS, Moon EJ, Landon C, Manzoor A, Hochman DW, Viglianti BL, Dewhirst MW (2011) Thresholds for thermal damage to normal tissues: an update. International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group 27:320-343.

Zhang X, Dong F, Mayer GE, Bruch DC, Ren J, Culver B (2007) Selective inhibition of cyclooxygenase-2 exacerbates methamphetamine-induced dopamine depletion in the striatum in rats. Neuroscience 150:950-958.

Zhu JP, Xu W, Angulo JA (2006a) Methamphetamine-induced cell death: selective vulnerability in neuronal subpopulations of the striatum in mice. Neuroscience 140:607-622.

116

Zhu JP, Xu W, Angulo N, Angulo JA (2006b) Methamphetamine-induced striatal apoptosis in the mouse brain: comparison of a binge to an acute bolus drug administration. Neurotoxicology 27:131-136.

117