77
University of North Georgia Nighthawks Open Institutional Repository Honors eses Honors Program Spring 2017 In Vivo Effects of Chronic Methamphetamine Exposure: A Study of Elemental and Inflammatory Responses and Potential Mechanisms for Zinc Accumulation in the Midbrain. Alyssa ompson University of North Georgia, [email protected] Follow this and additional works at: hps://digitalcommons.northgeorgia.edu/honors_theses Part of the Biology Commons is Honors esis is brought to you for free and open access by the Honors Program at Nighthawks Open Institutional Repository. It has been accepted for inclusion in Honors eses by an authorized administrator of Nighthawks Open Institutional Repository. Recommended Citation ompson, Alyssa, "In Vivo Effects of Chronic Methamphetamine Exposure: A Study of Elemental and Inflammatory Responses and Potential Mechanisms for Zinc Accumulation in the Midbrain." (2017). Honors eses. 16. hps://digitalcommons.northgeorgia.edu/honors_theses/16

In Vivo Effects of Chronic Methamphetamine Exposure: A

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: In Vivo Effects of Chronic Methamphetamine Exposure: A

University of North GeorgiaNighthawks Open Institutional Repository

Honors Theses Honors Program

Spring 2017

In Vivo Effects of Chronic MethamphetamineExposure: A Study of Elemental and InflammatoryResponses and Potential Mechanisms for ZincAccumulation in the Midbrain.Alyssa ThompsonUniversity of North Georgia, [email protected]

Follow this and additional works at: https://digitalcommons.northgeorgia.edu/honors_theses

Part of the Biology Commons

This Honors Thesis is brought to you for free and open access by the Honors Program at Nighthawks Open Institutional Repository. It has beenaccepted for inclusion in Honors Theses by an authorized administrator of Nighthawks Open Institutional Repository.

Recommended CitationThompson, Alyssa, "In Vivo Effects of Chronic Methamphetamine Exposure: A Study of Elemental and Inflammatory Responses andPotential Mechanisms for Zinc Accumulation in the Midbrain." (2017). Honors Theses. 16.https://digitalcommons.northgeorgia.edu/honors_theses/16

Page 2: In Vivo Effects of Chronic Methamphetamine Exposure: A

In Vivo Effects of Chronic Methamphetamine Exposure: A Study of Elemental and Inflammatory Responses and

Potential Mechanisms for Zinc Accumulation in the Midbrain

A Thesis Submitted to the Faculty of the University of North Georgia

In Partial Fulfillment Of the Requirements for the Degree

Bachelor of Science in Biology With Honors

Alyssa G. Thompson Spring 2017

Page 3: In Vivo Effects of Chronic Methamphetamine Exposure: A
Page 4: In Vivo Effects of Chronic Methamphetamine Exposure: A

II

ACKNOWLEDGEMENTS

Throughout the three years I have been conducting my thesis research, I have been helped by many mentors, family members, and friends. To each of them, I owe special thanks.

To my family for always supporting me, and encouraging me when I needed it the most.

To my friends for helping me in my journey, (at least pretending) to be interested as I continually obsessed over zinc, and always understanding why “I can’t, I have to be in lab” is a completely valid excuse.

To those I mentored for allowing me an opportunity to teach. I hope you maintain your enthusiasm and curiosity, no matter what field of study.

To the Biology and Chemistry departments for use of their equipment and research space.

To the Biology department and CURCA for funding for my research and opportunities to present at conferences.

To Dr. Dansby-Sparks for giving me direction when I first began my work in the Chemistry department, and spending many long afternoons with us over a burning crucible in the analytical lab.

To Dr. Leyba for his assistance with and permission to use the X-Ray Fluorescence Spectrometer, without which much of my research would not have been possible.

To Dr. Shanks and Dr. Lloyd for trusting me to take over my first research project, and for always being around to answer my millions of questions.

To Dr. Gomolak for… Absolutely everything else. Thank you for never accepting anything but the best from me. I have learned so much under your direction the past two years. I could never have picked a better research mentor, and I find myself extremely fortunate to have worked with you.

Page 5: In Vivo Effects of Chronic Methamphetamine Exposure: A

III

ABBREVIATIONS

BBB – Blood-Brain Barrier

Br – Bromine

BSA – Bovine Serum Albumin

Ca – Calcium

Cl – Chlorine

CNS – Central Nervous System

CT – Critical Threshold

Cu – Copper

CuZnSOD – Copper-Zinc Superoxide Dismutase

DA – Dopamine

EB – Evans Blue

Fe – Iron

IL-1β – Interleukin 1 Beta

IP – Intraperitoneal

K – Potassium

LD50 – Lethal Dose for 50% of Subjects

LLD – Lower Limit of Detection

MB – Midbrain

METH – Methamphetamine

MT – Metallothionein

MT3 – Metallothionein 3

mm – millimeter

Na – Sodium

NADPH - Nicotinamide Adenine Dinucleotide Phosphate

Ni – Nickel

NO – Nitric Oxide

Page 6: In Vivo Effects of Chronic Methamphetamine Exposure: A

IV

nNOS – Neuronal Nitric Oxide Synthase

PCR – Polymerase Chain Reaction

PFC – Prefrontal Cortex

qPCR – Real-time Polymerase Chain Reaction

Rb – Rubidium

RNA – Ribonucleic Acid

RNS – Reactive Nitrogen Species

ROS – Reactive Oxygen Species

s – seconds

SE – Standard Error

SN – Substantia Nigra

SOD – Superoxide Dismutase

STR – Striatum

Ti – Titanium

TNF-α – Tumor Necrosis Factor Alpha

T-PER – Total Protein Extraction Reagent

TXRF – Total-Reflection X-Ray Fluorescence Spectrometer

VTA – Ventral Tegmental Area

WST – Water-Soluble Tetrazolium Salt

Y – Yttrium

Zn – Zinc

18S – 18S Ribosomal Subunit

Page 7: In Vivo Effects of Chronic Methamphetamine Exposure: A

1

TABLE OF CONTENTS

BACKGROUND ................................................................................................................4

METHODS .......................................................................................................................12

Animal Care ..................................................................................................................13

Treatment Injections......................................................................................................13

Animal Sacrifice............................................................................................................13

Brain Harvest ................................................................................................................14

Brain Microdissection ...................................................................................................14

Brain Tissue X-Ray Fluorescence Spectroscopy ..........................................................15

Plasma X-Ray Fluorescence Spectroscopy ...................................................................15

Protein Purification .......................................................................................................16

Protein Quantification ...................................................................................................16

Superoxide Dismutase Activity Assay ..........................................................................16

Evans Blue Protocol ......................................................................................................17

Evans Blue Histology ....................................................................................................17

Evans Blue Microscopy and Analysis...........................................................................18

RNA Isolation ...............................................................................................................18

RNA Quantification ......................................................................................................19

Real-time Polymerase Chain Reaction .........................................................................19

PCR Analysis ................................................................................................................20

Statistical Testing ..........................................................................................................21

RESULTS .........................................................................................................................34

Significant Difference in Metal Concentrations in Midbrain .......................................35

Elemental Analysis of Mouse Plasma ...........................................................................35

Superoxide Dismutase Activity in Brain Homogenate .................................................36

Evidence of Blood-Brain Barrier Permeability via Evans Blue Assay .........................37

Significant Changes in DNA Transcription via qPCR Analysis ...................................37

DISCUSSION ...................................................................................................................56

Page 8: In Vivo Effects of Chronic Methamphetamine Exposure: A

2

CONCLUSION ................................................................................................................64

REFERENCES .................................................................................................................67

EQUATIONS

Equation 1. Determination of SOD activity by determination of percent inhibition of enzyme .......................................................................................................................................22

TABLES

Table 1. Superoxide Dismutase Reaction Well Setup ..................................................23

Table 2. Master Mix Preparation for qPCR ..................................................................24

Table 3. Optimized Primer Annealing Temperatures for qPCR ...................................35

Table 4. Total Reflection X-Ray Fluorescence Spectroscopy values of murine prefrontal cortex .............................................................................................................................38

Table 5. Total Reflection X-Ray Fluorescence Spectroscopy values of murine striatum .......................................................................................................................................39

Table 6. Total Reflection X-Ray Fluorescence Spectroscopy values of murine midbrain .......................................................................................................................................40

Table 7. Elemental concentrations in mouse plasma analyzed by TXRF .....................41

Table 8. Counts per pixel measurements of Evans Blue permeability in Saline and METH-treated mice ...................................................................................................................42

Table 9. Fold Change Values for IL-1β and MT3 mRNA Expression Determined via PCR .......................................................................................................................................43

FIGURES

Figure 1. Brief initial synopsis of the METH-induced neurodegenerative process ......11

Figure 2. Macro- and microdissection regions of murine brain ....................................26

Figure 3. TXRF Spectrum of murine plasma sample ...................................................27

Figure 4. TXRF Spectrum of murine midbrain sample ................................................28

Figure 5. Representative brain slices for Evans Blue Staining analysis of PFC ...........29

Figure 6. Representative brain slices for Evans Blue Staining analysis of STR...........30

Figure 7. Representative brain slices for Evans Blue Staining analysis of MB ............31

Figure 8. IL-1β primer optimization and melting curve ...............................................32

Figure 9. MT3 primer optimization and melting curve ................................................33

Page 9: In Vivo Effects of Chronic Methamphetamine Exposure: A

3

Figure 10. Elemental changes measured by TXRF in PFC of saline and METH treated mice .......................................................................................................................................44

Figure 11. Elemental changes measured by TXRF in STR of saline and METH treated mice .......................................................................................................................................45

Figure 12. Significant element increases in the midbrains of METH treated mice ......46

Figure 13. Elemental concentrations in mouse plasma .................................................47

Figure 14. Iron concentrations in mouse plasma ..........................................................48

Figure 15. Percent inhibition of Copper-Zinc Superoxide Dismutase in mouse MB samples .......................................................................................................................................49

Figure 16. Protocol optimization of Copper-Zinc Superoxide Dismutase Assay .........50

Figure 17. Whole brains after Evans Blue treatment ....................................................51

Figure 18. Determination of Blood-Brain Barrier Permeability by Evans Blue Assay 52

Figure 19. qPCR Data of IL-1β and MT3 from murine PFC ........................................53

Figure 20. qPCR Data of IL-1β and MT3 from murine STR .......................................54

Figure 21. qPCR Data of IL-1β and MT3 from murine MB .........................................55

Figure 22. Summation of known and experimentally derived data ..............................63

Page 10: In Vivo Effects of Chronic Methamphetamine Exposure: A

4

BACKGROUND

Page 11: In Vivo Effects of Chronic Methamphetamine Exposure: A

5

Methamphetamine (METH) is a widely used drug of abuse in the United States (NIDH 2013). The

characteristic “high” associated with METH intoxication comes from the excessive release of

dopamine (DA) into the neuronal synapse. The oxidative damage due to DA release is amplified

by blocked reuptake of the neurotransmitter (Ajjimaporn et al., 2005; Ajjimaporn et al., 2008;

Cadet et al., 1994; Mirecki, 2004). During the increased time in the synapse, DA causes irreparable

damage to the neuron terminals leaving lasting effects on the brain (Cadet et al., 1994; Riddle et

al., 2006). In the laboratory, the damaging effects of METH are used to model neurodegenerative

disease states that affect the dopaminergic pathways. Three major dopaminergic pathways exist in

the brain. The mesocortical pathway begins in the ventral tegmental area (VTA) and extends to

the prefrontal cortex (PFC). The mesolimbic pathway projects from the VTA to the striatum

(STR). The nigrostriatal pathway projects from the substantia nigra (SN) to the STR. Due to their

close proximity, for the purpose of this thesis, the SN and VTA are paired together and collectively

referred to as the midbrain (MB). Dopaminergic cell bodies reside in the MB and project their

axons to the PFC and STR. Therefore, all three regions (PFC, STR, and MB) are key in studying

the neurotoxic effects of METH.

A mouse model (adult C57BL/6J males) was chosen for their wild type characteristics and normal

neurological function, making a suitable model for studying the effects of drug treatment and

addiction. The doses of METH used in the literature range from subacute doses at 0.5 mg/kg

(Shanks et al., 2015) to large doses reaching 30 mg/kg (Ares-Santos et al., 2014; Carmena et al.,

2016). Higher doses may be used to model acute administration of METH and bring about

extensive neurodegeneration (Bowyer et al., 2008). Our dose of 5 mg/kg METH is moderate,

Page 12: In Vivo Effects of Chronic Methamphetamine Exposure: A

6

insuring a physiological effect without causing widespread death of dopaminergic neurons. For

our studies, a chronic 10-day drug treatment was to model METH abuse in humans.

Though it is the most widely cited effect of METH, DA release is not the only detrimental result

of METH use. Blood-brain barrier leakage, hyperthermia, potassium influx, edema, mitochondrial

dysfunction, and inflammation may all result from METH intoxication (Aizenman et al., 2010;

Ajjimaporn et al., 2005; Riddle et al., 2006). These consequences of METH treatment often work

in parallel, making it difficult to separate cause and effect.

The blood-brain barrier (BBB) functions to protect the brain from harmful substances in peripheral

circulation. Tight junctions between astrocytes and pericytes surrounding blood vessels in the brain

work to prevent large or charged molecules from passing through (Luissiant et al., 2012). Due to

its lipophilicity, METH is able to easily pass through the BBB. High doses of METH can cause a

breakdown of the BBB and may allow ions or proteins to pass through into the brain (Bowyer et

al., 2008; Northrop & Yamamoto, 2015; Turowski & Kenny, 2015). This unregulated passage of

potentially damaging materials may cause brain edema and can lead to CNS damage and cell death

(Turowski & Kenny, 2015). A potential mechanism for the BBB disruption is METH-induced

hyperthermia. Hyperthermia is a state of elevated body temperatures, and in this case, is most

noticeable in the brain (Brown et al., 2002). Studies have shown that hyperthermia is necessary for

BBB leakage, though the exact relationship between METH and BBB leakage is not currently

known (Northrop & Yamamoto, 2015; Turowski & Kenny, 2015). Commonly, questions of BBB

permeability are answered by Evans Blue (EB) assay (Manaenko et al., 2011). EB dye is a large

Page 13: In Vivo Effects of Chronic Methamphetamine Exposure: A

7

molecule that binds to plasma albumin, which may only cross into the brain if the BBB is disrupted

in some way. If EB dye is allowed to pass into the brain, the compromised brain region will

accumulate the dye and, in extreme cases, may even appear blue.

Brain edema following METH treatment is well documented and intertwined with both

hyperthermia and BBB disruption (Sharma & Hoopes, 2003). A study by Kiyatkin et al. 2007

presented hyperthermia, BBB disruption, and increased ion concentrations as major effects of

METH treatment. Increases in Chlorine (Cl), Sodium (Na), and Potassium (K) are associated with

increased water influx, called edema, and were all noted after treatment of rats with a 9 mg/kg

dose of METH (Kiyatkin et al., 2007). Ion influx is likely a precursor to brain edema, as net flow

of water tends to be towards hyperosmotic tissues. The ion imbalances and influx of water

characteristic of edema may be a fatal symptom of METH treatment. Brain edema may be

responsible for cases of increased mortality after dosing of METH well below the lethal dose for

50% of participants (LD50) (9 mg/kg; Kiyatkin et al., 2007).

An increase in K+ in the brain may be due to mobilized microglia as part of the inflammatory

response (Knoch et al., 2008). Subsequently, a study by Aizenman et al. 2010 found that the

damage associated with METH intoxication was increased when microglia were present, leading

to the belief that inflammation plays a key role in the damaging effects of METH. Microglia are

part of the immune response in the brain. In the event of CNS damage, microglia are responsible

for releasing proinflammatory cytokines, like interleukin 1 beta (IL-1β) and tumor necrosis factor

alpha (TNF-α), nitric oxide (NO), superoxide species, and other signaling molecules (Loane &

Page 14: In Vivo Effects of Chronic Methamphetamine Exposure: A

8

Byrnes, 2010; Shanks et al., 2012). The formation of radical oxygen species (ROS) and nitrogen

species (RNS) by microglia are fully dependent on the increase in K+ current and Zn2+ release

(Knock et al., 2008). Likewise, production of ROS and RNS from local microglia may be brought

about by METH (Ajjimaporn et al., 2005). Therefore, it is reasonable to assert that METH

treatment causes CNS damage adequate to mobilize microglia and begin an inflammatory response

(Fernandes et al., 2016). This inflammatory response is crucial in the dysregulation of homeostatic

ion and metal concentrations in the brain.

Iron serves a number of varied functions in the body from mitochondrial functioning to oxygen

transport (Ward et al., 2014). Initially, our studies focused on accumulation of iron in the spleen.

In previous studies by Shanks et al (unpublished), dark spots were noticed on the spleens of

METH-treated mice. These dark spots were thought to be excess heme, as the spleen functions to

filter and recycle damaged red blood cells. Excess iron in peripheral organs like the spleen and

liver may cause organ failure, which is a serious health risk (Badria et al., 2015). Imbalances in

metal concentration in the brain are often found as a characteristic of neurological dysfunction,

like Alzheimer’s disease (Alimonti et al., 2007; Andrasi et al., 2000). Neuroinflammation due to

disorders like Alzheimer’s and Parkinson’s disease, mediated by cytokines like IL-1β and TNF-α,

may cause an accumulation of iron in the brain (Li et al., 2016; Ji et al., 2016). Iron cannot be

excreted in humans, and concentrations are monitored by changing the rate of absorption from

food (Badria et al., 2015). With potential excess iron accumulation in peripheral organs, like the

spleen, my thesis focuses on studying potential mechanism and effects of metal accumulation,

including iron, zinc, and rubidium following neuroinflammation caused by METH treatment.

Page 15: In Vivo Effects of Chronic Methamphetamine Exposure: A

9

Zinc is an essential element for both plants and animals. Nearly three-quarters of zinc is transported

throughout the body bound to plasma albumin (Roohani et al., 2013). Zinc plays a key role in the

inflammatory pathway. Supplementation with zinc may decrease damaging effects of METH,

while chelating the ion may increase damage caused by METH within the brain (Aizenman et al.,

2010; Bao et al., 2010). As noted previously, the inflammatory response includes production of

ROS and RNS by microglia. Under normal circumstances, ROS are metabolized by several

mechanisms within the body. Within normal ranges, zinc may act as an antioxidizing agent through

superoxide dismutase (SOD) and metallothionein (MT) proteins (Tyszka-Gochara et al., 2014).

Copper-zinc superoxide dismutase (CuZnSOD) is a member of the SOD family of enzymes and

uses Cu and Zn as cofactors. Some amount of oxygen free radicals are formed as a byproduct of

normal cell metabolic processes. CuZnSOD plays a role in neutralizing the free radicals formed

before they have a chance to exert toxic effects on proteins or nucleic acids within the cell.

CuZnSOD has shown an increase after METH treatment in some, but not all cases (Mirecki et al.,

2004). Metallothionein proteins are one of many zinc-transporting proteins found within cell

membranes. MTs act to transport metals in or out of cells, also acting as a reservoir to maintain

metal homeostasis by binding or releasing bound metal ions as necessary. Metallothionein-3

(MT3) is specific to brain tissues, and may have free-radical neutralizing properties due to the

presence of bound divalent metals, including Zn2+. These two proteins, as well as the homeostasis

of intracellular zinc levels, are extremely important to the health of cells. Decreases in zinc levels

may be linked to depression (Doboszewska et al., 2016) while excess zinc levels cause

mitochondrial dysfunction (Ajjimaporn et al., 2005; Sensi et al., 2009; Weiss et al., 2014). Cell

death may occur as excess zinc may activate proapoptotic cascades (Sensi et al., 2009; Pochwat et

Page 16: In Vivo Effects of Chronic Methamphetamine Exposure: A

10

al., 2015). Excess zinc exerts mitochondrial toxicity by entering the organelle and disrupting the

membrane potential (Tyszka-Gochara et al., 2014). ROS and RNS created following excessive

zinc levels are likely due to the mitochondrial dysfunction (Weiss et al., 2014; Emoto et al., 2015),

but may relate to other pathways, including nicotinamide adenine dinucleotide phosphate

(NADPH) oxidase, as well (Sensi et al., 2009).

In the preliminary studies for this thesis, we determined that excess zinc was sequestered in the

midbrains of METH-treated mice when compared to Saline-treated controls. Using that

information, our studies of the relationship between METH intoxication and Zn2+ broaden the

understanding of the damaging effects of this drug and metal accumulation in the brain. A wild-

type mouse model (male C57BL6J mice) was used to study the in vivo neurodegenerative process.

Brain regions (PFC, STR, and MB) were chosen based on previously optimized lab protocols, due

to the high prevalence of dopaminergic neurons in the reward pathway in those regions. A brief

synopsis of this process may be found in figure 1.

Page 17: In Vivo Effects of Chronic Methamphetamine Exposure: A

11

Figure 1. Brief initial synopsis of the METH-induced neurodegenerative process. During the initial phase of this thesis research, an increase in zinc concentration in the midbrain was noted in METH-treated mice as compared to Saline-treated controls. Further studies consisted of the exploration of this phenomenon and search for potential mechanisms.

Page 18: In Vivo Effects of Chronic Methamphetamine Exposure: A

12

METHODS

Page 19: In Vivo Effects of Chronic Methamphetamine Exposure: A

13

Animal Care

Adult (n=66; average age=112.4 days ± 3.22) male C57BL/6J mice (Jackson Laboratory, Bar

Harbor, ME) were housed in plastic cages on a 12-hour light/dark cycle. They were allowed food

and water ad libitum. All procedures were performed in accordance to the Guide for the Care and

Use of Laboratory Animals and established university laboratory procedures (Shanks et al. 2015).

Treatment Injections

C57BL/6J mice were weighed daily to determine dosing of drug treatment. Mice were given one

intraperitoneal (IP) injection daily of either 5 mg/kg METH or 0.1 mL/10 g body mass of 0.9 %

sterile saline (Saline). Treatment injections were administered each morning (0800) for 10 days.

Animal Sacrifice

On Day 10, 4 hours after the final treatment injection, animals were given an IP injection of

Avertin. Avertin was prepared in accordance to established protocols by the Division of

Laboratory Animal Resources at the University of Kentucky Medical Center. A timepoint of 4

hours was chosen because it allowed most of the treatment drug to be metabolized, as may be

noted by a return to normal ambulation levels in METH-treated mice. Once sufficiently

anesthetized, the animals were decapitated and the brains were harvested.

Page 20: In Vivo Effects of Chronic Methamphetamine Exposure: A

14

Brain Harvest

The brain was removed from the skull by first cutting the fur from the base of the skull to the nose.

The skin was peeled back to reveal the skull beneath. The skull was cut in small sections and

peeled away from the brain, starting from the hindbrain region and moving forward to the olfactory

bulbs. Once the majority of the skull was removed, the ear bones were removed, the frontal bone

between eye sockets was cut, and the brain was removed. Whole brains were either frozen at

-80°C, or further microdissected and then frozen until use.

Brain Microdissection

After removal, the brain was placed in a chilled metal brain matrix. 2 mm slices containing the

prefrontal cortex (PFC), striatum (STR), and midbrain (MB) were taken. Macro-incisions were

made as shown in Figure 2A: Olfactory bulbs (0-2mm), prefrontal cortex (2-3 mm), striatum (4-5

mm), and midbrain (9-10 mm). The center of the PFC was determined as 1.94 bregma, STR at

0.50 bregma, and MB at -3.28 bregma, as shown in Figure 2B-D. PFC, STR, and MB 2 mm slices

were further dissected under a dissection scope, and the regions of interest were either processed

immediately or stored in labeled Eppendorf tubes at -80°C. Brain regions (PFC, STR, and MB)

were chosen based on previously optimized lab protocols and the presence of many dopaminergic

neurons in the reward pathway.

Page 21: In Vivo Effects of Chronic Methamphetamine Exposure: A

15

Brain Tissue X-Ray Fluorescence Spectroscopy

Mice were injected with 5 mg/kg METH or 0.9% Saline for 10 days before sacrifice and brain

dissection, as described previously. PFC, STR, and MB were homogenized by sonication. 100 μL

of homogenized tissue was transferred to a separate Eppendorf tube and 10 μL of internal standard

(Yttrium) was added. The sample was vortexed to mix components thoroughly. 10 μL of the new

sample was transferred to a disposable acrylic disc and allowed to dry on a hot plate. The sample

disc was analyzed by Bruker S2 PICOFOX Total-Reflection X-Ray Fluorescence spectrometer

(TXRF). Each brain sample was run in triplicate for 1000 seconds (s) and turned clockwise 1/3

each time, to account for any uneven pipetting of sample homogenate on the carrier disc (Figure

3).

Plasma X-Ray Fluorescence Spectroscopy

After decapitation, blood was collected and centrifuged (12,000 RPM for 5 min) to pellet the red

blood cells. Plasma was isolated and transferred to a labeled Eppendorf tube. 10 μL of internal

standard (Yttrium) was added to 100 μL of plasma and the tube was vortexed. 10 μL of the

prepared plasma sample was transferred to a disposable acrylic disc and allowed to dry on a hot

plate. The sample discs were transferred to the TXRF for analysis. Plasma samples were run once

each for 500 s (Figure 4).

Page 22: In Vivo Effects of Chronic Methamphetamine Exposure: A

16

Protein Purification

Mice were injected with 5 mg/kg METH or 0.9% Saline for 10 days before sacrifice and brain

dissection, as described previously. Microdissected brain tissue samples were sonicated in 1 mL

Tissue Protein Extraction reagent (T-PER) per 0.05 g of tissue, as per supplier (Thermo

Scientific®) recommendations. Homogenized samples were centrifuged at 10,000 rpm for 5

minutes to pellet debris. Supernatant was pipetted into a labeled Eppendorf tube and stored at -

80°C.

Protein Quantification

Protein concentrations were determined by Bio-Rad Protein assay. The assay was conducted as

per manufacturer (Bio-Rad®) instructions, although further dilutions were required to achieve

measurable concentrations. Two-fold serial dilutions of bovine serum albumin (BSA) were created

to make the standard curve. 10 μL of each BSA standard and protein sample was pipetted in

triplicate into a 96-well plate. 200 μL of dye reagent was added to each well, and the plate was

briefly centrifuged to remove any bubbles. The plate was allowed to incubate at room temperature

for approximately 5 minutes before the absorbance was measured in a 96-well plate reader at 595

nm.

Page 23: In Vivo Effects of Chronic Methamphetamine Exposure: A

17

Superoxide Dismutase Activity Assay

Reagents were prepared as per manufacturer (Abcam) instructions. Briefly, 1 mL Water-soluble

Tetrazolium Salt (WST) solution was diluted with 19 mL of SOD Assay Buffer. SOD Enzyme

solution was centrifuged briefly and mixed by pipet. 15 μL of enzyme solution was diluted with

2.5 mL of Dilution buffer. Materials and reagents were equilibrated to room temperature before

assay was run. Reaction wells were prepared as shown in Table 1, with Enzyme Working Solution

added last.

Immediately after the enzyme working solution was added, the plate was incubated for 20 minutes

at 37°C, then measured on a microplate reader at OD450 nm. Absorbance values for each sample

(run in triplicate) were averaged. SOD activity for each sample was calculated based on equation

1.

Evans Blue Protocol

Protocol was developed based on studies by Manaenko et al. 2010. Mice were injected with 5

mg/kg METH or 0.9% Saline for 10 days, as described previously. On Day 10, 2.5 hours after the

final treatment injection, an injection of 2% Evans Blue dye equal to the volume of treatment

administered (0.1 mL/10 g body mass) was given IP. Evans Blue dye was allowed to circulate for

1.5 hours before the animals were anesthetized by IP Avertin injection and brains were removed.

Whole brains were immediately transferred to -80°C freezer.

Page 24: In Vivo Effects of Chronic Methamphetamine Exposure: A

18

Evans Blue Histology

Frozen brains were removed from -80°C storage and placed into a chilled metal brain matrix.

Brains were allowed to thaw for 2 minutes before being sliced into 1 mm sections, then returned

to the freezer. Brain slices were removed from the brain matrix and transferred to microscope

slides, returning to the freezer for 2 minutes whenever the tissue became too thawed to easily

handle. When not in use, brain slices were stored at -80°C. PFC, STR, and MB slices were

examined under a dissecting scope at 12x magnification.

Evans Blue Microscopy and Analysis

Microscope pictures were taken using OMAX® 0.5x Reduction Lens (A3RDF50). Pictures of

magnified PFC, STR, and MB sections were taken at 12x magnification. Pictures of whole brains

were taken at 7.5x magnification. Brain sections were grouped based on location in the atlas

(Figures 5, 6, 7). Photographs of brain regions were converted from JPEG to 8-bit format to better

differentiate individual colors. Pictures were analyzed by histogram for blue pixel concentration

using ImageJ. Blue pixel values were recorded by a blind observer and later analyzed for

significance.

RNA Isolation

Mice were injected with 5 mg/kg METH or 0.9% Saline for 10 days before sacrifice and brain

dissection, as described previously. Microdissected PFC, STR, and MB samples were sonicated in

Page 25: In Vivo Effects of Chronic Methamphetamine Exposure: A

19

600 mL of lysis solution and transferred to RNase-free Eppendorf tubes. Tubes were centrifuged

for 2 minutes at 12,000 RPM and the supernatant (approximately 550 μL) was transferred to

another tube. An equal volume of 70% ethanol was added to the supernatant and the tubes were

vortexed to mix. If loose debris was present in the tubes, they were centrifuged again at 12,000

RPM for 1 minute. The RNA collection column was assembled with the collection tube.

Approximately 600 μL of lysate and ethanol solution was applied to the column and it was

centrifuged at 12,000 RPM for 1 minute. The flow-through was discarded, and the step is repeated

with the remaining lysate and ethanol solution, again, discarding the remaining flow-through. 400

μL of column wash solution was applied to the column and centrifuged for 1 minute, repeated once

under identical conditions, then repeated again with 400 μL of wash solution but centrifuged for

two minutes. Flow-through was discarded after each RNA wash. An elution tube was assembled

to the column and 50 μL of elution buffer was added to the column. The column was centrifuged

for 2 minutes at 1500 RPM, then for 1 minute at 12,000 RPM. The final flow-through containing

purified RNA was placed on the column again and centrifuged once more to maximize the amount

of RNA isolated.

RNA Quantification

Purified RNA concentrations for each sample were assessed by NanoDrop 1000

spectrophotometry (Thermo Scientific®). RNA was diluted to 1 ng/mL concentrations before use.

Both stock RNA and diluted RNA aliquots were stored at -20°C when not in use.

Page 26: In Vivo Effects of Chronic Methamphetamine Exposure: A

20

Real-time Polymerase Chain Reaction

Primers were diluted to 1 μM solutions using 1 μL of stock primer solution (concentration 100

μM) and 99 μL molecular grade H2O. Primer sequences are as follows:

MT3 – 5’ CTGCTGGACTGGATATGGAC 3’ and 5’ CTCACTGGCAGCAGCTGCAT 3’ (Chen

et al., 2004).

IL-1β – 5’ CAACCAACAAGTGATATTCTCCAT 3’ and 5’ GATCCACACTCTCCAGCTGCA

3’ (Linares et al., 2006).

18S – 5’ CTGAGAAACGGCTACCACATC 3’ and 5’ CGCTCCCAAGATCCAACTAC 3’

(Abbas et al., 2011).

Purified RNA was isolated, as described above, and used for replication. Verso SYBR Green qPCR

master mix was prepared according to manufacturers’ (Thermo Scientific) suggestion and

previously established lab protocol (Table 2). Plate layout was designed using Real-time

Polymerase Chain Reaction (qPCR) software with each sample run in triplicate.

qPCR Program was set to run as follows: 50°C for 15 minutes, 95°C for 15 minutes, 95°C for 15

seconds, 55°C* for 30 seconds, 72°C for 30 seconds (*temperature varied by primer). Melting

curves were added at the end of each thermal cycling program, to ensure the primers were

functioning properly. Initial primer optimization included test plates of escalating RNA

concentrations run at different temperatures until the optimum annealing temperature for each

primer was found. Optimization was based on melting curves and primer efficiency calculations

(Table 3; Figure 8-9).

Page 27: In Vivo Effects of Chronic Methamphetamine Exposure: A

21

PCR Analysis

qPCR plates were analyzed for critical threshold (CT) value. CT value is determined as the point

where each sample crosses the fluorescence threshold. First, CT values from each treatment gene

were compared to CT values from the housekeeping gene (18S). This is to ensure standard amounts

of RNA were being compared within treatment groups and gives the ΔCT value. To determine if

significant changes in gene expression existed, statistics were run comparing ΔCT values for

METH and Saline treatment groups. METH and Saline treatment groups were compared to

determine the fold change in gene expression, giving the ΔΔCT value.

Statistical Testing

Group sizes and animal numbers were determined based on a number of factors, including previous

experimental designs, information regarding variability, and power analysis. Paired t-tests for

single comparisons and unpaired t-tests for comparison of single groups were performed when

applicable. All data are expressed as means±SE. Statistical significance was accepted at p<0.05.

Error bars represent standard error (SE).

Page 28: In Vivo Effects of Chronic Methamphetamine Exposure: A

22

𝑆𝑆𝑆𝑆𝑆𝑆 𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴 (𝐴𝐴𝑖𝑖ℎ𝐴𝐴𝑖𝑖𝐴𝐴𝐴𝐴𝐴𝐴𝑖𝑖𝑖𝑖 𝑟𝑟𝑟𝑟𝐴𝐴𝑟𝑟 %) = (𝐴𝐴𝑖𝑖𝐴𝐴𝑟𝑟𝑖𝑖𝐴𝐴1 – 𝐴𝐴𝑖𝑖𝐴𝐴𝑟𝑟𝑖𝑖𝐴𝐴3) – (𝐴𝐴𝐴𝐴𝑟𝑟𝐴𝐴𝐴𝐴𝐴𝐴𝑟𝑟 – 𝐴𝐴𝑖𝑖𝐴𝐴𝑟𝑟𝑖𝑖𝐴𝐴2)

(𝐴𝐴𝑖𝑖𝐴𝐴𝑟𝑟𝑖𝑖𝐴𝐴1 − 𝐴𝐴𝑖𝑖𝐴𝐴𝑟𝑟𝑖𝑖𝐴𝐴3) ∗ 100

Equation 1. Determination of SOD activity by determination of percent inhibition of enzyme. Absorbance values from blank wells were compared to absorbance values from each sample well. These values were then converted to a percent and reported as percent inhibition of enzyme.

Page 29: In Vivo Effects of Chronic Methamphetamine Exposure: A

23

Table 1. Superoxide Dismutase Reaction Well Setup. Setup was proposed by Abcam in accordance with their Superoxide Dismutase Activity Colorimetric Assay kit (ab65354). Assay was performed in a standard colorimetric 96-well plate.

Page 30: In Vivo Effects of Chronic Methamphetamine Exposure: A

24

Table 2. Master Mix Preparation for qPCR. Master mix was prepared for each primer in accordance with kit protocols and standard lab procedure.

Page 31: In Vivo Effects of Chronic Methamphetamine Exposure: A

25

Table 3. Optimized Primer Annealing Temperatures for qPCR. Annealing temperature optimization is crucial to ensure primers anneal to the proper target and not a different, non-specific sequence. **18S served as our housekeeping gene and had been previously optimized by the lab of Dr. Shanks and Dr. Lloyd.

Page 32: In Vivo Effects of Chronic Methamphetamine Exposure: A

26

A. B.

C. D.

Figure 2. Macro- and microdissection regions of murine brain. A. Macrodissection landmarks for whole murine brain as may be shown in metal brain matrix: PFC 2-3 mm, STR 4-5 mm, and MB 9-10 mm. B. Microdissection region of murine PFC; approx. 1.94 bregma. C. Microdissection region of murine STR; approx. 0.50 bregma. D. Microdissection region of murine MB; approx. -3.28 bregma.

Page 33: In Vivo Effects of Chronic Methamphetamine Exposure: A

27

Figure 3. TXRF Spectrum of murine plasma sample. Blood was extracted from decapitated mice and centrifuged for 5 minutes at 12,000 RPM to separate out plasma. 10 μL of Yttrium (Y) was added to 100 μL of plasma, and 10 μL of the mixture was pipetted onto an acrylic carrier disc. The disc was analyzed for 500 s. Main peaks detected (from right to left) are Molybdenum (Mo), Yttrium (Y), Rubidium (Rb), Bromine (Br), Zinc (Zn), Copper (Cu), Iron (Fe), Calcium (Ca), Potassium (K), Argon (Ar), Chlorine (Cl). Molybdenum detected inside the instrument; Argon detected from the air. keV – energy signature of elements. Pulse – intensity of secondary x-rays emitted by sample. The height of each peak shows the prevalence of each element in the sample.

Page 34: In Vivo Effects of Chronic Methamphetamine Exposure: A

28

Figure 4. TXRF Spectrum of murine midbrain sample. Mice treated with either 5 mg/kg METH or 0.9% Saline for 10 days. On Day 10, the mice were sacrificed and their brains removed. Brains were microdissected into PFC, STR, and MB. Microdissected brain segments were sonicated in RNase free water. 10 μL of Y was added to 100 μL of brain homogenate, and 10 μL of the mixture was pipetted onto an acrylic carrier disc. Samples were run three times (1000 s each), turned 1/3 each time, for a total run time of 3000 s. Main peaks detected (from right to left) are Molybdenum (Mo), Yttrium (Y), Rubidium (Rb), Bromine (Br), Zinc (Zn), Copper (Cu), Nickel (Ni), Iron (Fe), Titanium (Ti), Potassium (K), Argon (Ar), Chlorine (Cl). Molybdenum detected inside the instrument; Argon detected from the air. keV – energy signature of each element. Pulse – number of secondary x-rays emitted per primary x-ray emitted by the instrument. Height of each peak relates to the concentration of each element within the sample.

Page 35: In Vivo Effects of Chronic Methamphetamine Exposure: A

29

A. B.

C.

Figure 5. Representative brain slices for Evans Blue Staining analysis of PFC. Bregma is determined as the point on the skull where coronal and sagittal sutures meet. Millimeter determinations anterior or posterior to bregma are given for each region. A. First PFC slice, approximately 2.80 mm anterior to bregma. B. Second PFC slice, approximately 1.94 mm anterior to bregma. C. Third PFC slice, approximately 1.10 mm anterior to bregma. Saline n=4; METH n=5. Yellow arrows indicate areas of blue staining within cortical blood vessels.

Page 36: In Vivo Effects of Chronic Methamphetamine Exposure: A

30

A. B.

Figure 6. Representative brain slices for Evans Blue Staining analysis of STR. Bregma is determined as the point on the skull where coronal and sagittal sutures meet. A. First STR slice, approximately 0.38 mm anterior to bregma. B. Second STR slice, approximately 1.34 mm posterior to bregma. Saline n=4; METH n=6. Yellow arrows indicate areas of blue staining within ventricles.

Page 37: In Vivo Effects of Chronic Methamphetamine Exposure: A

31

A. B

C.

Figure 7. Representative brain slices for Evans Blue Staining analysis of MB. Bregma is determined as the point on the skull where coronal and sagittal sutures meet. A. First MB slice, approximately 2.80 mm posterior to bregma. B. Second MB slice, approximately 4.60 mm posterior to bregma. C. Third MB slice, approximately 5.68 mm posterior to bregma. Saline n=4; METH n=6. Yellow arrows indicate areas of blue staining.

Page 38: In Vivo Effects of Chronic Methamphetamine Exposure: A

32

A.

B.

Figure 8. IL-1β primer optimization and melting curve. Primer concentrations for optimization were established at 10 ng, 5 ng, 1 ng, 0.5 ng, 0.1 ng. qPCR cycling program setup: 50°C for 15 minutes, 95°C for 15 minutes, 95°C for 15 seconds, 60°C for 30 seconds, 72°C for 30 seconds. A. PCR Product replication curves. Demonstrates that primers worked efficiently. B. Melting Curve. Demonstrates that primers were specific to the sequence of interest and only one product was created. Saline n=5; METH n=5.

Page 39: In Vivo Effects of Chronic Methamphetamine Exposure: A

33

A.

B.

Figure 9. MT3 primer optimization and melting curve. Primer concentrations for optimization were established at 10 ng, 5 ng, 1 ng, 0.5 ng, 0.1 ng. qPCR cycling program setup: 50°C for 15 minutes, 95°C for 15 minutes, 95°C for 15 seconds, 55°C for 30 seconds, 72°C for 30 seconds. A. PCR Product replication curves. Demonstrates that primers worked efficiently. B. Melting Curve. Demonstrates that primers were specific to the sequence of interest and only one product was created. Saline n=5; METH n=5.

Page 40: In Vivo Effects of Chronic Methamphetamine Exposure: A

34

RESULTS

Page 41: In Vivo Effects of Chronic Methamphetamine Exposure: A

35

Significant Difference in Metal Concentrations in Midbrain

PFC, STR, and MB microdissected from both Saline and METH-treated animals were analyzed

by TXRF and data was recorded the following elements: Bromine (Br), Chlorine (Cl), Copper

(Cu), Iron (Fe), Potassium (K), Nickel (Ni), Rubidium (Rb), Titanium (Ti), and Zinc (Zn) (Tables

4-6). Three measurements were taken for each brain sample, to account for any uneven pipetting

of brain homogenate. The three measurements for each sample were averaged to achieve a final

value. The element concentrations for Saline and METH treatments were compared by a one-tailed

two-sample t-test (α=0.05). Significant increases in the MB for the following elements were

observed in METH-treated mice: Cl (Saline 2079.4 ± 502.04 µg/l; METH 4734.9 ± 1061.54 µg/l;

p=0.03), K (Saline 44010 ± 5490.29 µg/l; METH 64724 ± 4085.13 µg/l; p=0.01), Zn (Saline 144.7

± 16.44 µg/l; METH 210.3 ± 17.28 µg/l; p=0.01), and Rb (Saline 14.5 ± 2.21 µg/l; METH 25.2 ±

3.44 µg/l; p=0.02) (Figures 10-12). When compared to Saline-treated mice, METH-treated mice

had an increased elemental response to the drug. The increase in element concentrations was

localized to the MB and did not appear in the PFC or STR.

Elemental Analysis of Mouse Plasma

Plasma was analyzed by TXRF and data was recorded for the following elements: Bromine (Br),

Calcium (Ca), Chlorine (Cl), Copper (Cu), Iron (Fe), Potassium (K), Rubidium (Rb), Zinc (Zn)

(Table 7). Elemental concentrations were analyzed by one-tailed two-sample t-test (α=0.05).

Unlike what was seen in the brain, no significant changes in Cl (Saline 2138673 ± 470153 μg/l;

METH 1424580 ± 416307 μg/l; p=0.14), K (Saline 207501 ± 56142.5 μg/l; METH 147498 ±

48870.1 μg/l; p=0.22), Rb (Saline below lower limit of detection (LLD); METH (n=2) 46.59 ±

Page 42: In Vivo Effects of Chronic Methamphetamine Exposure: A

36

7.87 μg/l), or Zn (Saline 591.35 ± 47.12 μg/l; METH 532.53 ± 46.21 μg/l; p=0.19) were found

between Saline-treated and METH-treated mice (Figure 13). Interestingly, a statistically

significant decrease (p=0.04) in Fe concentration was found in METH-treated mice (2142.43 ±

305.1 μg/l) as compared to Saline-treated controls (3060.89 ± 380.32 μg/l; figure 14). Rubidium

concentrations were unable to be measured as they fell below the lower limit of detection (LLD;

average LLD 45.31 ± 3.92 μg/l). Two Rb concentrations were above the LLD and quantified by

the TXRF. Both samples were taken from METH-treated mice (38.73 and 54.46 μg/l).

Superoxide Dismutase Activity in Brain Homogenate

SOD inhibition rates were calculated based on measured absorbance values. Absorbance values

give a comparative measure of the amount of SOD present in the sample. Based on the assay setup,

percent inhibition of SOD and the initial amount of SOD present have an inverse relationship. The

greater the amount of SOD inhibition, the less SOD is present in the initial sample. No significant

difference was found in the percent SOD inhibition between Saline (89.34 ± 2.81%) and METH

(92.01 ± 1.68%) treated groups in any region of the brain (Figure 15). Additional experiments

examining the effects of protein amount and incubation time were run, to verify manufacturer

protocols (Figure 16). The lack of difference in percent inhibition of SOD between Saline and

METH-treated groups leads to the conclusion there was also no significant difference in the

amount of SOD present between the samples.

Page 43: In Vivo Effects of Chronic Methamphetamine Exposure: A

37

Evidence of Blood-Brain Barrier Permeability via Evans Blue Assay

Dissection of brain tissue and analysis of 8-bit color images revealed significant differences in

Evans Blue permeability between Saline and METH-treated animals (Table 8). EB dye was most

concentrated in blood vessels, as well as ventricles within the brain, as would be expected based

on the circulation of EB. Within the tissue, no obvious large blue spots may be noted. However,

densitometry analysis revealed significant increases in EB permeability occurred in the STR

(p=0.03) and MB (p=0.01) of METH-treated animals when compared to Saline controls (Figure

18). PFC data were nonsignificant (p=0.44), with much larger counts per pixel values than the

other two brain regions. The lack of difference in PFC is likely due to the notable accumulation of

EB in cortical blood vessels near the upper border of the PFC tissue samples, which would have

artificially increased the counts per pixel values.

Significant Changes in DNA Transcription via qPCR Analysis

qPCR plates were analyzed for CT value, ΔCT value, and ΔΔCT value, as described previously.

Two-tailed two-sample t-tests (α=0.05) were run to compare each ΔCT value for IL-1β and MT3

from PFC, STR, and MB regions. A significant decrease in IL-1β was found in the midbrain of

METH-treated mice as compared to Saline (p<0.05). No other significant changes in IL-1β were

found in other brain regions, nor were significant changes of MT3 found in any brain region (Table

9). Relative expression (fold change) of IL-1β and MT3 was determined by the ΔΔCT value and

data is shown in Figures 19-21.

Page 44: In Vivo Effects of Chronic Methamphetamine Exposure: A

38

Table 4. Total Reflection X-Ray Fluorescence Spectroscopy values of murine prefrontal cortex. Brain samples taken from C57BL/6J mice treated with 5 mg/kg METH or 0.9% Saline by IP injection for 10 days. No significant concentration differences were determined between Saline and METH-treated mice. Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05.

Page 45: In Vivo Effects of Chronic Methamphetamine Exposure: A

39

Table 5. Total Reflection X-Ray Fluorescence Spectroscopy values of murine striatum. Brain samples taken from C57BL/6J mice treated with 5 mg/kg METH or 0.9% Saline by IP injection for 10 days. No significant concentration differences were determined between Saline and METH-treated mice. Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05.

Page 46: In Vivo Effects of Chronic Methamphetamine Exposure: A

40

Table 6. Total Reflection X-Ray Fluorescence Spectroscopy values of murine midbrain. Brain samples taken from C57BL/6J mice treated with 5 mg/kg METH or 0.9% Saline by IP injection for 10 days. Significant increases in Zn, Cl, K, and Rb were found in the midbrain of METH-treated mice as compared to Saline controls. Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05. *p<0.05

Page 47: In Vivo Effects of Chronic Methamphetamine Exposure: A

41

Table 7. Elemental concentrations in mouse plasma analyzed by TXRF. Plasma samples taken from C57BL6J mice treated with either 5 mg/kg METH or 0.9% Saline for 10 days were analyzed for elemental concentration. A significant decrease (p=0.04) in serum iron concentration was found in METH-treated mice as compared to Saline. Many of the sample levels of Rb fell below the lower limit of detection and therefore could not be measured. Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05. *p<0.05 **Rb METH treatment group: n=2.

Page 48: In Vivo Effects of Chronic Methamphetamine Exposure: A

42

Table 8. Counts per pixel measurements of Evans Blue permeability in Saline and METH-treated mice. Nine brains (Saline n=4; METH n=5) were analyzed for EB leakage. One mm brain slices were grouped according to location in brain atlas. If any dissected brain region did not fit into a group or if the tissue was not able to be properly analyzed, it was excluded. Selected brain regions were dissected and photographed. 8-bit color images were analyzed for blue pigment concentration in ImageJ. Concentrations were averaged and treatment groups were analyzed for significance (α=0.05) by two-tailed two-sample t-test. *p<0.05

Page 49: In Vivo Effects of Chronic Methamphetamine Exposure: A

43

A.

B.

Table 9. Fold Change Values for IL-1β and MT3 mRNA Expression Determined via PCR. PCR was performed to amplify sequences for IL-1β (A) and MT3 (B) in purified mRNA from murine PFC, STR, and MB samples. ΔCT values were calculated to compare CT values from the housekeeping gene and gene of interest within each sample. ΔΔCT values were calculated to compare ΔCT values between METH and Saline treatment groups. Fold change was calculated using ΔΔCT values and is reported for IL-1β and MT3. Saline n=5; METH n=5. Two-tailed two-sample t-tests were performed on ΔCT values; α=0.05. *p<0.05.

Page 50: In Vivo Effects of Chronic Methamphetamine Exposure: A

44

A. B.

C. D.

Figure 10. Elemental changes measured by TXRF in PFC of Saline and METH-treated mice. A. Chlorine concentrations in prefrontal cortex homogenate (p=0.22). B. Potassium concentrations in prefrontal cortex homogenate (p=0.46). C. Rubidium concentration in prefrontal cortex homogenate (p=0.32). D. Zinc concentration in prefrontal cortex homogenate (p=0.45). Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05.

Page 51: In Vivo Effects of Chronic Methamphetamine Exposure: A

45

A. B.

C. D.

Figure 11. Elemental changes measured by TXRF in STR of Saline and METH-treated mice. A. Chlorine concentration in striatum homogenate (p=0.40). B. Potassium concentration in striatum homogenate (p=0.43). C. Rubidium concentration in striatum homogenate (p=0.46). D. Zinc concentration in striatum homogenate (p=0.22). Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05.

Page 52: In Vivo Effects of Chronic Methamphetamine Exposure: A

46

A. B.

C. D.

Figure 12. Significant element increases in the midbrains of METH-treated mice. Zn, Cl, K, and Rb were increased after 10 days of treatment, as compared to Saline-treated mice. A) Chlorine concentration in midbrain homogenate (p=0.03). B) Potassium concentration in midbrain homogenate (p=0.01). C) Rubidium concentration in midbrain homogenate (p=0.02). D) Zinc concentration in midbrain homogenate (p=0.01). Saline n=6; METH n=6. One-tailed two-sample t-test; α=0.05. *p<0.05

* *

* *

Page 53: In Vivo Effects of Chronic Methamphetamine Exposure: A

47

A. B.

C. D.

Figure 13. Elemental concentrations in mouse plasma. After 10 days of treatment with either Saline or 5 mg/kg METH, mice were decapitated and trunk blood was collected. Plasma was isolated from blood and analyzed by TXRF. Saline n=9; METH n=9. A. Chlorine concentration from plasma sample (p=0.14). B. Potassium concentration from plasma sample (p=0.22). C. Rubidium concentration from plasma sample (n=2; 38.73 and 54.46 μg/l). D. Zinc concentration in plasma (p=0.19). One-tailed two-sample t-test (α=0.05).

Page 54: In Vivo Effects of Chronic Methamphetamine Exposure: A

48

Figure 14. Iron concentrations in mouse plasma. After 10 days of treatment with either Saline or 5 mg/kg METH, mice were decapitated and trunk blood was collected. Plasma was isolated from blood and analyzed by TXRF. Saline n=9; METH n=9. Iron concentration in plasma (Saline 3060.89 ± 380.32 μg/l; METH 2142.43 ± 503.01 μg/l; p=0.04). One-tailed two-sample t-test (α=0.05); *p<0.05.

*

Page 55: In Vivo Effects of Chronic Methamphetamine Exposure: A

49

Figure 15. Percent inhibition of Copper-Zinc Superoxide Dismutase in mouse MB samples. Colorimetric assay (Abcam assay ab65354) was performed to determine percent inhibition of SOD enzyme in Saline and METH-treated samples. No significant difference was determined between Saline (89.34 ± 2.81%) and METH-treated (92.01 ± 1.68%) MB samples (p=0.21). Other brain regions also showed no significant differences between Saline and METH treatment groups. Saline n=6; METH n=7. One-tailed two-sample t-test; α=0.05.

Page 56: In Vivo Effects of Chronic Methamphetamine Exposure: A

50

A. B.

Figure 16. Protocol optimization of Copper-Zinc Superoxide Dismutase Assay. A) Percent Inhibition as a function of protein amount (g). Saline n=3; METH n=3. B) Percent inhibition as a function of incubation time (min). Saline n=6; METH n=7. One-tailed two-sample t-test; α=0.05.

Page 57: In Vivo Effects of Chronic Methamphetamine Exposure: A

51

A. B.

C. D.

E. F.

Figure 17. Whole brains after Evans Blue treatment. A & B. Brain from Saline-treated mouse (A. Dorsal surface; B. Ventral surface); frozen prior to analysis. Brain displayed a large amount of cortical EB staining. C & D. Brain from METH-treated mouse (C. Dorsal surface; D. Ventral surface); frozen prior to analysis. Brain displayed a moderate amount of cortical staining. E & F. Freshly dissected brain (E. Dorsal surface; F. Ventral surface); not previously frozen. Brain displayed a minimal amount of cortical staining. Brain frozen before analysis. Scale bar = 1 mm; 7.5x magnification.

Page 58: In Vivo Effects of Chronic Methamphetamine Exposure: A

52

A. B.

C.

Figure 18. Determination of Blood-Brain Barrier Permeability by Evans Blue Assay. 8-bit images of 1 mm PFC, STR, and MB slices were analyzed by the Histogram feature in ImageJ. Blue pixel counts were summed and divided to achieve standardized counts/pixel measurements. (A) PFC; p=0.442. (B) STR; p=0.032. (C) MB; p=0.012. Saline n=4; METH n=5. Two-tailed two-sample t-test; α=0.05. *p<0.05

Page 59: In Vivo Effects of Chronic Methamphetamine Exposure: A

53

A. B.

C. D.

Figure 19. qPCR Data of IL-1β and MT3 from murine PFC. A. ΔCT values of IL-1B PFC. A significant increase in METH ΔCT value was determined as compared to Saline control (p=0.017). B. Fold Change from IL-1B PFC. Relative expression of IL-1β in METH-treated mice is downregulated two-fold of gene expression in Saline control mice. C. ΔCT values from MT3 PFC. D. Fold Change from MT3 PFC. No significant differences exist between the treatment groups in regard to relative MT3 gene expression. Saline n=5; METH n=5. Two-tailed two-sample t-test; α=0.05. *p<0.05.

Page 60: In Vivo Effects of Chronic Methamphetamine Exposure: A

54

A. B.

C. D.

Figure 20. qPCR Data of IL-1β and MT3 from murine STR. A. ΔCT value for IL-1B STR. B. Fold Change for IL-1B STR. C. ΔCT value for MT3 STR. D. Fold Change for MT3 STR. Saline n=5; METH n=5. Two-tailed two-sample t-test; α=0.05.

Page 61: In Vivo Effects of Chronic Methamphetamine Exposure: A

55

A. B.

C. D.

Figure 21. qPCR Data of IL-1β and MT3 from murine MB. A. ΔCT value for IL-1B MB. A significant increase in ΔCT value was determined for METH-treated mice as compared to Saline-treated mice (p=0.049). B. Fold Change for IL-1B MB. IL-β expression in METH-treated mice is approximately half of the expression found in Saline-treated mice. C. ΔCT value for MT3 MB. D. Fold Change for MT3 MB. Saline n=5; METH n=5. Two-tailed two-sample t-test; α=0.05. *p<0.05.

Page 62: In Vivo Effects of Chronic Methamphetamine Exposure: A

56

DISCUSSION

Page 63: In Vivo Effects of Chronic Methamphetamine Exposure: A

57

Although some studies cite the use of Zn in attenuating the deleterious effects of METH in the

brain (Aizenman et al., 2010; Ajjimaporn et al., 2005), our findings of both Zn and Rb

accumulation in the MB following METH exposure appear to be novel. For the purposes of this

thesis, our studies centered on Zn. Cl and K function as cell signaling ions with roles in neuronal

signal propagation and cell responses. Therefore, it is likely increases in Cl and K concentrations

are central to edema following METH treatment and the BBB disruption that typically precedes it

(Kiatkin et al., 2007). Rb is a trace metal found in body tissues, with no known biological function.

Due to their similarity in size and charge, K and Rb appear to have similar biological activities

(Relman, 1956). Rb cannot be substituted for K in biological processes, but its presence often

reflects the concentration of K in body tissues (Roberts et al., 2016). Recent studies present

decreased concentrations of both K and Rb as characteristics of Alzheimer’s disease (Roberts et

al., 2016). Though our studies did not include further investigation of the paired increase of both

Rb and K, the future examination of Rb as a biomarker may prove useful. Naturally occurring

radioactive isotopes of Rb may be examined by noninvasive scans. If scans show Rb is sequestered

in a certain area, it is likely, because of their similar activities, K is also present in significant

quantities. This may be useful in diagnosing edema related disorders, or other disorders that relate

to large quantities of K present. Zn and K concentration surges may both be related to microglial

activation and inflammatory stress (Knoch et al. 2008).

Studies of excess zinc levels link high metal concentrations in many scenarios to the increased

production of ROS, which leads to cell death (Ajjimaporn et al., 2005; Knoch et al., 2008; Sensi

et al., 2009). Through our studies, we analyzed the amount of two free radical scavenging agents

as possible mechanisms for the increase in zinc concentration. CuZnSOD is a free enzyme found

Page 64: In Vivo Effects of Chronic Methamphetamine Exposure: A

58

within cells that functions to neutralize free radicals in solution. It is well established that an

increase in free radical species occurs following METH treatment (Ajjimaporn et al., 2005; Cubells

et al., 1994; Friend et al., 2013). Therefore, we initially hypothesized that CuZnSOD enzymes are

in greater demand in the brain, and carry with them excess Zn. The activity assay correlating

inhibition of SOD to amount of SOD in solution determined there was no significant difference in

SOD amount between Saline and METH-treated groups. Therefore, although Zn is an important

cofactor in the activity of CuZnSOD, it does not appear to be the cause of the midbrain

accumulation of Zn. MT3 is a brain specific cell membrane protein that functions to transport Zn

in and out of cells, and possesses free radical scavenging properties when Zn ions are bound. We

hypothesized that an increase in the Zn transporter concentration could be responsible for the

excess deposition of zinc in the midbrain. However, qPCR analysis showed no significant

difference in the amount of MT3 mRNA transcript between METH and Saline-treated mice. The

lack of difference in mRNA produced gives no evidence for an increase in MT3 protein production.

Based on these results, both SOD and MT3 appear to be unrelated to and perhaps unaffected by

the increase in Zn in the midbrain after 10 days of METH treatment. Additional studies utilizing

Western Blots would detect changes in the amount of formed protein products between treatment

groups. Further experimentation examining changes in the concentration of other Zn-containing

proteins or in the strength of the inflammatory responses would be useful in elucidating another

mechanism of Zn entry.

METH-induced inflammation via microglial activation causes neurons to release zinc from

intracellular stores (Knoch et al., 2008). Also from the aforementioned study, microglia also cause

an increase in K+ channel signaling. Both K+ and Zn2+ play roles in apoptotic cascades after

Page 65: In Vivo Effects of Chronic Methamphetamine Exposure: A

59

neuronal inflammation (Knoch et al., 2008). This leads us to believe microglial-derived

inflammation plays a key role in METH toxicity. However, due to the decrease in cytokine IL-1β

mRNA transcript in METH-treated animals, our qPCR analysis suggests a decrease in

inflammation in the midbrain. Inflammation typically increases due to microglial activation with

METH treatment (Fernandes et al., 2016); however, reports of attenuation due to tolerance may

provide insight (Thomas & Kuhn, 2005). In the study by Thomas & Kuhn, pretreatment with

METH doses significantly reduces the activation of microglia following challenge doses, even

after a washout period of 7 days. Our data supports conclusions proposed by Thomas and Kuhn,

as our chronic 10-day treatment with 5 mg/kg METH falls outside of their studied acclimation

range of microglia. They do not propose a potential mechanism for the attenuation, only proposing

that the acclimation of microglia in the striatum noted in their study has a neuroprotective function

against further inflammatory damage following METH treatment. This decrease in inflammatory

markers may also help explain what was seen in CuZnSOD and MT3 studies. CuZnSOD and MT3

may act to neutralize free radicals as part of the immune response. However, if there is a lack of

inflammation occurring, there is no substantive reason for those mechanisms to be upregulated

inside of the cell. A decrease in TNF-α and IL-1β in cells under oxidative stress was noted after

zinc supplementation in a study of effective treatments of atherosclerosis by Bao et al. 2010.

Pretreatment with zinc often shows neuroprotective benefits against METH toxicity (Aizenman et

al., 2010; Bao et al., 2010; Ajjimaporn et al., 2005). Therefore, we hypothesize the accumulation

of zinc in vivo may have a neuroprotective effect against inflammation following METH treatment.

Further studies are required to determine the relationship between microglial attenuation, a

decrease in IL-1β cytokine mRNA, and zinc accumulation following METH treatment.

Page 66: In Vivo Effects of Chronic Methamphetamine Exposure: A

60

The transport of ions from the plasma into the brain under circumstances of METH intoxication

may be facilitated by the BBB breakdown following METH-induced hyperthermia. As has been

noted previously, we also observed statistically significant EB staining of METH-treated mice

when compared to Saline controls. Using photographs and ImageJ analysis, we were able to verify

the disruption of the BBB in two of the three areas directly affected by METH exposure, the STR

and MB. Our study of the PFC showed nonsignificant results, likely due to the accumulation of

EB in the cortical blood vessels. However, in future studies, perfusion of the animal with cold PBS

following EB treatment and anesthesia would likely resolve this error and allow for a proper study

of changes to PFC permeability.

Due to the lower zinc concentration in the brain of Saline-treated mice when compared to METH-

treated mice, we hypothesized a higher average plasma zinc concentration would appear in Saline-

treated mice as compared to METH-treated mice. Our data supported our hypothesis through a

modest decrease in zinc levels in plasma from METH-treated mice compared to Saline-treated

mice. However, due to variability within the treatment groups, the values did not approach

statistical significance (p=0.19). Iron concentrations in the plasma were significantly decreased

(p=0.04). The decrease in serum iron concentration may be related to the initial increase in splenic

iron. If red blood cells accumulate in the spleen following METH treatment, as we presume to be

causing the dark spots noted in our initial experiments, it would cause the drop in circulating iron

concentration, as we see experimentally. Instrumental limitations prevented us from measuring

levels of Rb in the plasma. The increase in the base LLD for Rb in plasma samples is likely due to

refractory noise from the Br peak nearby. The peak for Br was artificially increased in plasma

samples by IP injection of the anesthetic drug Avertin (2, 2, 2-tribromoethanol) before sacrifice.

Page 67: In Vivo Effects of Chronic Methamphetamine Exposure: A

61

Even though we were not able to analyze serum Rb concentrations to look for trends, the brain

increase in Rb concentration is promising. Based on the literature and our data, the increase in Rb

concentration in the brain after METH treatment may be a viable option for future use as a

biomarker for neurodegeneration.

METH intoxication has many deleterious effects, as supported by published literature (Carmena

et al., 2015; Cubells et al., 1994; Emoto et al., 2015; Friend et al., 2013; Thomas & Kuhn, 2005).

Elemental accumulation of Zn, K, Cl, and Rb in the midbrains of mice as a result of chronic METH

treatment was observed compared to Saline-treated controls. Ion concentrations were measured in

plasma, to understand if the values correlated to ion levels found in the brain. Although no

significant correlations were found in Zn, K, Cl, and Rb, as were found, a significant decrease in

Fe concentration was observed in METH-treated mice when compared to Saline. In the brain

tissue, no significant difference in SOD expression was found between METH and Saline-treated

mice. PCR analysis of genes coding for MT3 showed no significant difference between METH

and Saline-treated mice in any brain region. A significant decrease compared to Saline-treated

mice was found in genes for IL-1β, a cytokine marker for inflammatory activity, via PCR analysis

in the prefrontal cortex and midbrain of METH-treated mice. It is thought this significant decrease

compared to controls may be an effect of microglial tolerance to the drug over the chronic 10-day

treatment. A study of BBB permeability gave support to previous literature findings with

disruptions in METH-treated groups that were not shown to the same degree in Saline-treated

groups. An increase in permeability was seen between both STR and MB samples, but not in PCF,

as the remaining EB in cortical blood vessels likely washed out any potential measurable

difference. It is likely this disruption is linked to edema and influx of cell signaling ions and metals,

Page 68: In Vivo Effects of Chronic Methamphetamine Exposure: A

62

although the particular mechanism directly targeting the MB is currently unknown. A summary of

these processes may be found in figure 22.

Page 69: In Vivo Effects of Chronic Methamphetamine Exposure: A

63

Figure 22. Summation of known and experimentally derived data. This chart attempts to display the complex interplay of the studies of this thesis. It is known in the literature that METH causes BBB leakage, edema through ion influx, and mitochondrial dysfunction. Our studies support the increase in ion concentration, including an influx of Rb (likely linked to the K influx) and also Zn accumulation. Significant BBB disruption was noted in STR and MB samples, potentially giving a potential point of entry for the accumulation of metals. Interestingly, our studies showed a decrease in IL-1β (a cytokine released by microglia) gene product, rather than an increase when compared to controls as would be expected following METH treatment. This surprising response is likely due to microglial tolerance to our chronic 10-day treatment of METH, though further studies are necessary to confirm this hypothesis. From the literature, it has been noted that excess zinc may be sequestered in mitochondria, leading to a disruption of the electron transport chain and ROS production. Though ROS production specifically was not a focus of our studies, the accumulation of zinc after METH treatment as compared to Saline in our studies may give a potential cause for the ROS production typically noted after METH treatment.

Page 70: In Vivo Effects of Chronic Methamphetamine Exposure: A

64

CONCLUSION

Page 71: In Vivo Effects of Chronic Methamphetamine Exposure: A

65

Our investigation into the neurodegeneration caused by chronic METH treatment has offered many

valuable contributions to the field. C57BL/6J mice treated with chronic doses of METH show an

accumulation of Zn, K, Rb, and Cl in the MB, as compared to Saline controls. Mice treated with

METH for 10 days also show a decrease in inflammatory cytokines in both the PFC and MB,

contrary to what is typically understood following METH treatment. Our studies suggested that

the accumulation of zinc is not due to an increase in SOD enzymes present, nor an increase in the

production of MT3 Zn-transporters in the brain. However, these results are supported by the

decrease in inflammatory cytokine mRNA found via PCR. Both SOD and MT3 help neutralize

free radicals, which may be released during the immune response. A lack of inflammation would

not require the production of anti-inflammatory proteins, potentially giving us sound reasoning for

the lack of significant differences between treatment groups in those studies. Increases in K may

be linked to BBB leakage, and may be further studied via Rb biomarker studies, as the two

elements share common accumulation patterns. BBB leakage was noted in the STR and MB, two

regions directly activated by METH intoxication and studied here. No difference was noted in the

PFC, likely because of the excess dye present in cortical blood vessels. Perfusion of the animal

prior to brain dissection and analysis should remove this confound in future studies and allow for

a clear look at the permeability of the PFC. The decrease in IL-1β mRNA concentration may be

due to the tolerance of microglia for METH after a 10-day chronic treatment. Studies of varying

length regarding METH treatment may be useful in determining the exact development of the zinc

accumulation and elucidating its role in METH-induced neurodegeneration. The idea of microglial

tolerance to METH-induced inflammation warrants further study to determine the exact

mechanism and its possible relationship to Zn accumulation. Further studies of other inflammatory

sources, like neuronal nitric oxide synthase (nNOS) production which has been found in increase

Page 72: In Vivo Effects of Chronic Methamphetamine Exposure: A

66

as a result of Zn excess, may also help elucidate a different facet of the role of Zn in RNS

production and inflammation.

Our studies contribute to the knowledge of metal accumulation following neurodegeneration, as is

seen with other neurodegenerative disorders like Parkinson’s and Alzheimer’s disease. Though the

mechanism of Zn accumulation is not yet known, our study of two potential mechanisms, both of

which attenuate METH-induced damage when upregulated (Cadet et al., 1994; Imam et al., 2001;

Ajjimaporn et al., 2005), point towards the necessity of different active mechanisms following

METH intoxication. Although we have so far been unable to determine the mechanism directly

targeting the MB or affecting the projection areas in PFC and STR, our studies provide support

and valuable insight into the changes within the brain following METH exposure, and warrant

further study.

Page 73: In Vivo Effects of Chronic Methamphetamine Exposure: A

67

REFERENCES

Page 74: In Vivo Effects of Chronic Methamphetamine Exposure: A

68

Abbas K, Breton J, Planson AG, Bouton C, Bignon J, Seguin C, Riquier S, Toledano MB, Drapier JC (2011) Nitric oxide activates an Nrf2/sulfiredoxin antioxidant pathway in macrophages. Free Rad Bio & Med. 51:107-114.

Aizenman E, McCord MC, Saadi RA, Hartnet KA, He K (2010). Complex role of zinc in methamphetamine toxicity in vitro. Neurosci. 171:31-39.

Ajjimaporn A, Shavali S, Ebadi M, Govitrapong P (2008). Zinc rescues dopaminergic SK-H-SH cell lines from methamphetamine-induced toxicity. Brain Res Bull. 77:361-366.

Ajjimaporn A, Swinscoe J, Shavali S, Govitrapong P, Ebadi M (2005). Metallothionein provides zinc-mediated protective effects against methamphetamine toxicity in SK-N-SH cells. Brain Res Bull. 67:466-475.

Alimonti A, Ristori G, Giubilei F, Stazi MA, Pino A, Visconti A, Brescianini S, Monti MS, Forte G, Stanzione P, Bocca B, Bomboi G, D’Ippolito C, Annibali V, Salvetti M, Sancesario G (2007) Serum chemical elements and oxidative status in Alzheimer’s disease, Parkinson disease and multiple sclerosis. Neurotox. 28:450-456.

Andrasi E, Farkas E, Gawlik D, Rosick U, Bratter P (2000) Brain iron and zinc contents of German patients with Alzheimer disease. J Alzheimer’s Disease. 2:17-26.

Badria FA, Ibrahim AS, Badria AF, Elmarakby AA (2015) Curcumin attenuates iron accumulation and oxidative stress in the liver and spleen of chronic iron-overloaded rats. PLoS ONE. 10(7):1-13.

Bao B, Prasad AS, Beck FWJ, Fitzgerald JT, Snell D, Bao GW, Singh T, Cardozo LJ (2010). Zinc decreases C-reactive protein, lipid peroxidation, and inflammatory cytokines in elderly subjects: a potential implication of zinc as an atheroprotective agent. Am J Clin Nutr. 91:1634-1641.

Bowyer JF, Thomas M, Schmued LC, Ali SF (2008) Brain region-specific neurodegenerative profiles showing the relative importance of amphetamine dose, hyperthermia, seizures, and the blood-brain barrier. Annals of the New York Academy of Sciences. 1139:127-139.

Brown PL, Wise RA, Kiyatkin EA (2003) Brain hyperthermia is induced by methamphetamine and exacerbated by social interaction. J Neurosci. 23(9):3924-3929.

Cadet JL, Sheng P, Ali S, Rothman R, Carlson E, Epstein C (1994) Attenuation of methamphetamine-induced neurotoxicity in copper/zinc superoxide dismutase transgenic mice. J Neurochem. 62(1):380-383.

Carmena A, Granado N, Ares-Santos S, Alberquilla S, Tizabi Y, Moratalla R (2015) Methamphetamine-induced toxicity in indusium griseum of mice is associated with astro- and microgliosis. Neurotox Res. 27:209-216.

Chen L, Wu W, Dentchev T, Wong R, Dunaief JL (2004) Increased metallothionein in light damaged mouse retinas. Exp Eye Res. 79:287-293.

Page 75: In Vivo Effects of Chronic Methamphetamine Exposure: A

69

Cubells JF, Rayport S, Rajendran G, Sulzer D (1994) Methamphetamine neurotoxicity involves vacuolation of endocytic organelles and dopamine-dependent intracellular oxidative stress. J Neurosci. 14(4):2260-2271.

Doboszewska U, Szewczyk B, Sowa-Kucma M, Noworyta-Sokolowska K, Misztak P, Golebiowska J, Mlyniec K, Ostachowicz B, Krosniak M, Wojtanowska-Krosniak A, Golembiowska K, Lankosz M, Piekowszewski W, Nowak G (2015) Alterations of bio-elements, oxidative, and inflammatory status in the zinc deficiency model in rats. Neurotox Res. 29:143-154.

Emoto MC, Yamato M, Sato-Akaba S, Yamada K, Matsuoka Y, Fujii HG (2015) Brain imaging of methamphetamine-treated mice using a nitroxide contrast agent for EPR imaging of the redox status and a gadolinium contrast agent for MRI observation of blood-brain barrier function. Free Rad Res. 49(8):1038-1047.

Fernandes NC, Sriram U, Gofman L, Cenna JM, Ramirez SH, Potula R (2016) Methamphetamine alters microglial immune function through P2X7R signaling. J Neuroinflam.13(91):1-13.

Friend DM, Son JH, Keefe KA, Fricks-Gleason AN (2013) Expression and activity of nitric oxide synthase isoforms in methamphetamine-induced striatal dopamine toxicity. J Pharmacol Exp Ther. 344:511-521.

Imam SZ, Newport GD, Itzhak Y, Cadet JL, Islam F, Slikker Jr W, Ali SF (2001) Peroxynitrite plays a role in methamphetamine-induced dopaminergic neurotoxicity: evidence from mice lacking neuronal nitric oxide synthase gene or overexpressing copper-zinc superoxide dismutase. J Neurochem. 76:745-749.

Ji S, Zhang S, Mao Z, Han H, Yang Q, Zhu W, Zhu S, Xue Z (2016) Quantitative assessment of iron deposition in Parkinson’s disease using enhanced T2 star-weighted angiography. Neurol India. 64(3):428-435.

Kiyatkin EA, Brown PL, Sharma HS (2007) Brain edema and breakdown of the blood-brain barrier during methamphetamine intoxication: critical role of brain hyperthermia. Eur J Neurosci. 26:1242-1253.

Kiyatkin EA, Brown PL, Wise RA (2002) Brain temperature fluctuation: a reflection of functional neural activation. Eur J Neurosci. 16:164-168.

Knoch ME, Hartnett KA, Hara H, Kandler K, Aizenman E (2008) Microglia induce neurotoxicity via intraneuronal Zn2+ release and a K+ current surge. Glia. 56(1):89-96.

Li Y, Pan K, Chen L, Ning J, Li X, Yang T, Terrando N, Gu J, Tao G (2016) Deferoxamine regulates neuroinflammation and iron homeostasis in a mouse model of postoperative cognitive dysfunction. J Neuroinflam. 13(268):1-12.

Page 76: In Vivo Effects of Chronic Methamphetamine Exposure: A

70

Linares D, Taconis M, Mana P, Correcha M, Fordham S, Staykova M, Willenborg DO (2006) Neuronal nitric oxide synthase plays a key role in CNS demyelination. J Neurosci. 26(49)12672-12681.

Loane DJ, Byrnes KR (2010) Role of microglia in neurotrauma. J Am Soc Exp NeuroTherapeutics. 7:366-377.

Luissint AC, Artus C, Glacial F, Ganeshamoorthy K, Couraud PO (2012) Tight junctions at the blood brain barrier: physiological architecture and disease-associated dysregulation. Fluids and Barriers of the CNS. 9(23):1-12.

Manaenko A, Chen H, Kammer J, Zhang JH, Tang J (2010) Comparison evans blue injection routes: intravenous versus intraperitoneal, for measurement of blood-brain barrier in a mice hemorrhage model. J Neurosci Meth. 195:206-210.

Mirecki A, Fitzmaurice P, Ang L, Kalasinsky KS, Peretti FJ, Aiken SS, Wickham DJ, Sherwin A, Nobrega JN, Forman HJ, Kish SJ (2004) Brain antioxidant systems in human methamphetamine users. J Neurochem. 89:1396-1408.

NIDA (2013). Methamphetamine. https://www.drugabuse.gov/publications/research-reports/methamphetamine.

Northrop NA, Yamamoto BK (2015) Methamphetamine effects on blood-brain barrier structure and function. Front Neurosci. 9(69):1-11.

Pochwat B, Nowak G, Szewczyk B (2015) Relationship between zinc (Zn2+) and glutamate receptors in the processes underlying neurodegeneration. Neural Plasticity. Article ID 591563. 1-9.

Relman AS (1956) The physiological behavior of rubidium and cesium in relation to that of potassium. Yale J Bio Med. 29(3):248-262.

Riddle EL, Fleckenstein AE, Hanson GR (2006) Mechanisms of methamphetamine-induced dopaminergic neurotoxicity. The AAPS Journ. 8(2):E413-418.

Roberts BR, Doecke JD, Rembach A, Yévenes LF, Fowler CJ , McLean CA, Lind M, Volitakis I, Masters CL, Bush AI, Hare DJ, and the AIBL research group (2016) Rubidium and potassium levels are altered in Alzheimer’s disease brain and blood but not in cerebrospinal fluid. Acta Neuropathologica Communications. 4:119.

Roohani N, Hurrell R, Kelishadi R, Schulin R (2013) Zinc and its importance for human health: An integrative review. J Res Med Sci 18(2):144-157.

Shanks RA, Anderson JR, Taylor JR, Lloyd SA (2012) Amphetamine and methamphetamine have a direct and differential effect on BV2 microglia cells. Bull Exp Biol Med. 154:228-232.

Shanks RA, Ross JM, Doyle HH, Helton AK, Picou BN, Schulz J, Tavares C, Bryant S, Dawson BL, Lloyd SA (2015) Adolescent exposure to cocaine, amphetamine, and methylphenidate cross-

Page 77: In Vivo Effects of Chronic Methamphetamine Exposure: A

71

sensitizes adults to methamphetamine with drug- and sex-specific effects. Behav Brain Res. 281:116-124.

Sharma HS, Hoopes PJ (2003) Hyperthermia induced pathophysiology of the central nervous system. Int J Hyperthermia. 19(3):325-354.

Thomas DM, Kuhn DM (2005) Attenuated microglial activation mediates tolerance to the neurotoxic effects of methamphetamine. J. Neurochem. 92:790-797.

Turowski P, Kenny BA (2015) The blood-brain barrier and methamphetamine: open sesame? Front Neurosci. 9(156):1-7.

Tyszka-Czochara M, Grzywacz A, Gdula-Argasinska J, Librowski T, Wilinski B, Opoka W (2014) The role of zinc in the pathogenesis and treatment of central nervous system (CNS) diseases. Implications of zinc homeostasis for proper CNS function. Acta Poloniae Pharmaceutica – Drug Res 71(3):369-377.

Ward RJ, Zucca FA, Duyn JH, Crichtton RR, Zecca L (2014) The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 13:1045-1060