<<

University of South Florida Scholar Commons

Graduate Theses and Dissertations Graduate School

January 2015 Sigma Activation Mitigates Toxicity Evoked by the Convergence of Ischemia, Acidosis and Amyloid-beta Adam Alexander Behensky University of South Florida, [email protected]

Follow this and additional works at: http://scholarcommons.usf.edu/etd Part of the Neurosciences Commons, Pharmacology Commons, and the Physiology Commons

Scholar Commons Citation Behensky, Adam Alexander, " Activation Mitigates Toxicity Evoked by the Convergence of Ischemia, Acidosis and Amyloid-beta" (2015). Graduate Theses and Dissertations. http://scholarcommons.usf.edu/etd/5641

This Dissertation is brought to you for free and open access by the Graduate School at Scholar Commons. It has been accepted for inclusion in Graduate Theses and Dissertations by an authorized administrator of Scholar Commons. For more information, please contact [email protected].

Sigma Receptor Activation Mitigates Toxicity Evoked by the

Convergence of Ischemia, Acidosis and Amyloid-β

by

Adam A. Behensky

A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Medical Sciences Department of Molecular Pharmacology and Physiology College of Medicine University of South Florida

Major Professor: Javier Cuevas, Ph.D. Thomas E. Taylor-Clark, Ph.D. Jerome W. Breslin, Ph.D. Kay-Pong Yip, Ph.D. Mack H. Wu, MD

Date of Approval: June 11, 2015

Keywords: intracellular calcium, whole-cell currents, ASIC1a, fluorescent imaging, stroke, vascular dementia

© Copyright 2015, Adam A. Behensky

ACKNOWLEDGMENTS

First of all, I like to think my mentor, Dr. Javier Cuevas, for giving me the opportunity to become a scientist and future independent investigator. Through his support and mentorship I have thrived in the field of stroke research. I would also like to thank my committee members, Drs. Thomas Taylor-Clark, Jerome Breslin, Kay-Pong

Yip and Mack Wu for their advice and support through these years. I would also like to extend thanks to my external examiner Dr. Donna Wilcock, for accepting to chair my dissertation committee and for her constructive criticism and comments on my dissertation.

I would also like to thank Dr. Chris Katnik and Dr. Parmvir Bahia for providing advice and troubleshooting for various experiments and offer my thanks to Dr. Sarah

Yuan, the chair of the Department of Molecular Pharmacology and Physiology, for all of her support as well as our department staff, Bridget Shields, Stacy Reed, Suzanne

McMahon and Dr. Eric Bennett our department vice chair and PhD program director.

Lastly, I would like to thank the vivarium staff, Haley, Karen and Jill for providing assistance with our experimental animals.

This work was funded in part by a pre-doctoral fellowship from the American

Heart Association to AAB (14PRE19570010).

TABLE OF CONTENTS

LIST OF TABLES ...... iv

LIST OF FIGURES ...... v

ABSTRACT ...... ix

CHAPTER 1 Introduction ...... 1 Significance ...... 1 Background ...... 2 Pathobiology of Ischemic Stroke ...... 4 Amyloid-β ...... 7 Microglia ...... 9 Sigma Receptors ...... 11 Afobazole ...... 14

CHAPTER 2 In Vitro Evaluation of Guanidine Analogues as Sigma Receptor Ligands for Potential Anti-Stroke Therapeutics ...... 17 Introduction ...... 17 Materials and Methods ...... 19 Preparation of Cortical Neurons ...... 19 Calcium Imaging ...... 20 In Vitro Competition Binding Assays ...... 21 Primary Cultures of Microglia ...... 21 Microglia Migration ...... 22 Drugs and Chemicals...... 23 Data Analysis ...... 24 Results ...... 24 Discussion ...... 39

CHAPTER 3 Afobazole Activation of Sigma-1 Receptors Modulates Neuronal Responses to Amyloid-β25-35 ...... 45 Introduction ...... 45 Materials and Methods ...... 48 Preparation of Rat Cortical Neurons ...... 48 Calcium Imaging ...... 49 ROS and NO Imaging ...... 49 Cytotoxicity Assay ...... 50 Immunocytochemistry ...... 51

i

Compounds, Reagents and Antibodies...... 52 Data Analysis ...... 52 Results ...... 52 Discussion ...... 66

CHAPTER 4 Stimulation of Sigma Receptors With Afobazole Blocks Activation of Microglia and Reduces Toxicity Caused by Amyloid-β25-35 ...... 72 Introducation ...... 72 Materials and Methods ...... 74 Primary Cultures of Microglia ...... 74 Membrane Morphology and Migration Assay ...... 75 Calcium Imaging Measurements ...... 76 ROS and NO Imaging ...... 77 Cell Death Assay ...... 77 Immunocytochemistry ...... 78 Reagents and Antibodies ...... 79 Data Analysis ...... 79 Results ...... 79 Discussion ...... 94

CHAPTER 5 Activation of Sigma Receptors via Afobazole Modulates Microglial but not Neuronal Apoptosis-Related Gene Expression in Response to Long-Term Ischemia Exposure ...... 101 Introduction ...... 101 Materials and Methods ...... 103 Primary Cultures of Microglia ...... 103 Preparation of Cortical Neurons ...... 104 Live/Dead Cell Assay ...... 104 Immunocytochemistry ...... 105 Calcium Imaging ...... 106 Compounds, Reagents and Antibodies...... 107 Data Analysis ...... 107 Results ...... 107 Discussion ...... 119

CHAPTER 6 Afobazole Abolishes Amyloid-β25-35 Potentiation of Ischemia- Acidosis Evoked Increases in Neuronal Intracellular Calcium Levels ...... 124 Introduction ...... 124 Materials and Methods ...... 126 Preparation of Rat Cortical Neurons ...... 126 Calcium Imaging ...... 126 Electrical Recordings ...... 127 Compounds, Reagents and Peptides ...... 128 Data Analysis ...... 128 Results ...... 129 Discussion ...... 141 ii

CHAPTER 7 Discussion ...... 146 Conclusion ...... 146

LIST OF REFERENCES ...... 168

APPENDICES ...... 182 Permissions to Reprint Previously Published Materials ...... 182

iii

LIST OF TABLES

Table 2.1 o-DTG binds to both σ receptor subtypes with greater affinity than p-BrDPhG ...... 38

iv

LIST OF FIGURES

Figure 1.1 Pathoneurobiology of ischemic stroke ...... 5

Figure 2.1 Shifting the location of the methyl moiety produces o-DTG 2+ analogues that inhibit ischemia-induced increases in [Ca ]i similar to the parent compound ...... 25

Figure 2.2 Removal, but not migration, of the methyl moiety increases capacity of o-DTG analogues to inhibit acidosis-evoked 2+ elevations in [Ca ]i ...... 26

Figure 2.3 Fluoro substitution does not affect o-DTG analogue inhibition 2+ of ischemia- or acidosis-evoked increases in [Ca ]i ...... 28

Figure 2.4 Chloro substitution at the meta and para positions increase inhibition of intracellular calcium elevations ...... 30

Figure 2.5 Substitution of the methyl moiety with bromide at the meta 2+ and para positions enhances [Ca ]i inhibition ...... 32

2+ Figure 2.6 Inhibition of ischemia-evoked increases in [Ca ]i by p-BrDPhG is concentration-dependent ...... 33

2+ Figure 2.7 p-BrDPhG inhibits acidosis-evoked increases in [Ca ]i in a concentration-dependent manner ...... 35

Figure 2.8 σ-1 receptor antagonist BD-1063 attenuates p-BrDPhG inhibition 2+ of acidosis-evoked increases in [Ca ]i ...... 36

Figure 2.9 p-BrDPhG blocks the migration of microglia elicited by ATP ...... 37

Figure 3.1 Aβ25-35 promotes time-dependent elevations in neuronal intracellular calcium ...... 53

2+ Figure 3.2 Afobazole inhibits increases in [Ca ]i evoked by Aβ25-35 ...... 55

Figure 3.3 Afobazole does not inhibit Aβ25-35 evoked ROS production but does inhibit nitric oxide production in neurons ...... 57

v

Figure 3.4 Afobazole, but not DTG, reduces neuronal death produced by application of Aβ25-35 ...... 59

Figure 3.5 Afobazole prevents upregulation of the pro-apoptotic gene product, Bax, caused by application of Aβ25-35 in neurons ...... 61

Figure 3.6 Afobazole reduces expression of activated caspase-3 in response to application of Aβ25-35 in neurons ...... 63

Figure 3.7 Afobazole prevents Bcl-2 down-regulation following 48 hr exposure of neurons to Aβ25-35 ...... 65

Figure 4.1 Afobazole blocks membrane ruffling induced by ATP in microglial cells ...... 80

Figure 4.2 Afobazole blocks membrane ruffling induced by Aβ25-35 in microglial cells ...... 81

Figure 4.3 Afobazole blocks the migration of microglia elicited by Aβ25-35 ...... 84

Figure 4.4 Afobazole does not decrease microglial ROS or NO production induced by Aβ25-35 ...... 85

Figure 4.5 Afobazole decreases microglial cell death produced by application of Aβ25-35 ...... 87

Figure 4.6 Afobazole reduces Aβ25-35-evoked expression of the pro- apoptotic gene product Bax, in microglia ...... 88

Figure 4.7 Afobazole reduces expression of activated caspase-3 in response to application of Aβ25-35 in microglia ...... 90

Figure 4.8 Afobazole has minor effects on Bcl-2 levels in microglia following application of Aβ25-35 ...... 91

2+ Figure 4.9 Afobazole preserves [Ca ]i responses to ATP in microglial cells exposed to Aβ25-35 ...... 93

Figure 5.1 Afobazole reduces microglia death produced by application of ischemia ...... 108

Figure 5.2 Afobazole prevents upregulation of the pro-apoptotic gene, Bax, observed in response to ischemia in microglia ...... 110

Figure 5.3 Afobazole reduces expression of caspase-3 in response to ischemia in microglia ...... 111 vi

Figure 5.4 Afobazole enhances Bcl-2 expression under control conditions and following ischemia in microglia ...... 113

Figure 5.5 Afobazole reduces neuronal death induced under ischemic conditions ...... 114

Figure 5.6 Afobazole fails to prevent upregulation of the pro-apoptotic gene, Bax, observed in response to extended ischemia in neurons ...... 115

Figure 5.7 Afobazole fails to reduce expression of caspase-3 in response to prolonged ischemia in neurons ...... 116

Figure 5.8 Afobazole alone upregulates the anti-apoptotic gene, Bcl-2, but does not enhance Bcl-2 levels following 24 hr ischemic incubation in neurons ...... 117

Figure 5.9 Afobazole prolongs neuronal survival from extended exposure of cortical neurons to ischemia, but does not maintain functionality ...... 118

Figure 6.1 Incubation for 12 hr with amyloid-β25-35 does not potentiate intracellular calcium responses to focal ischemia, acidosis or ischemia + acidosis application...... 130

Figure 6.2 24 hr incubation with amyloid-β25-35 significantly potentiates the intracellular calcium responses to ischemia, acidosis and ischemia + acidosis application...... 131

Figure 6.3 Amyloid-β fragments 25-35 and 1-42 potentiate ischemia + 2+ acidosis induce [Ca ]i increases ...... 132

Figure 6.4 Oligomerized amyloid-β25-35 fails to potentiate ischemia + 2+ acidosis evoked increases in [Ca ]i following a 1 hr incubation ...... 133

2+ Figure 6.5 Afobazole inhibits increases in [Ca ]i evoked by ischemia + acidosis application following 24hr Aβ25-35 incubation ...... 135

Figure 6.6 Amyloid-β25-35 incubation potentiates proton-evoked currents in cortical neurons ...... 136

Figure 6.7 Aβ potentiated calcium influx involves reversal of the sodium/ calcium exchanger ...... 137

2+ Figure 6.8 Amyloid-β25-35 potentiation of [Ca ]i increases evoked by ischemia + acidosis is not dependent of the sodium/calcium potassium-dependent exchanger (NCKX) ...... 139 vii

2+ Figure 6.9 Amyloid-β25-35 potentiation of the net [Ca ]i increases evoked by ischemia + acidosis may involve intracellular protons ...... 140

viii

ABSTRACT

Stroke is the fifth leading cause of death in the United States and a major cause of long-term disability in industrialized countries. The core region of an ischemic stroke dies within minutes due to activation of necrotic pathways. Outside of this core region is the penumbral zone, where some perfusion is maintained via collateral arteries. Delayed cell death occurs in this area due to the triggering of apoptotic mechanisms, which expands the ischemic injury over time. The cellular and molecular events that produce the expansion of the ischemic core continue to be poorly understood. The increases in the amyloid precursor and pathogenic secretases lead to the increase in amyloid-

β (Aβ) production. The relatively small amount of research in this area has hampered development of stroke therapy designed to prevent neuronal and glial cell degeneration in the penumbra. Currently, there is a significant lack of therapeutic options for acute ischemic stroke, and no drug has been approved for treating patients at delayed time points (≥ 4.5 hr post-stroke).

Afobazole, an anxiolytic currently used clinically in Russia, has been shown to reduce neuronal and glial cell injury in vitro following ischemia, both of which have been shown to play important roles following an ischemic stroke. Treatment with afobazole decreased microglial activation in response to ATP and Aβ, as indicated by reduced membrane ruffling and cell migration. Prolonged exposure of microglia to ischemia or Aβ

conditions resulted in glial cell death that was associated with increased expression of

ix

the pro-apoptotic protein, Bax, the death protease, caspase-3 and a reduced expression in Bcl-2. Co-application of afobazole decreased the number of cells expressing both Bax and caspase-3, while increasing the cells expressing Bcl-2 resulting in a concomitant enhancement in cell survival. While afobazole inhibited activation of microglia cells by

Aβ25-35, it preserved normal functional responses in these cells following exposure to the

amyloid peptide. Intracellular calcium increases induced by ATP were depressed in

microglia after 24 hr exposure to Aβ25-35. However, co-incubation with afobazole returned

these responses to near control levels. Therefore, stimulation of σ-1 and σ-2 receptors

by afobazole prevents Aβ25-35 activation of microglia and inhibits Aβ25-35-associated cytotoxicity.

Examining the molecular mechanisms involved in the increased neuronal survival demonstrates that ischemia or Aβ results in an increased expression of the pro-apoptotic

protein Bax and the death protease caspase-3, while at the same time decreasing expression of the anti-apoptotic protein, Bcl-2. However, unlike observations made with microglia, afobazole was unable to modulate this ischemia-induced expression, but was able to modulate Aβ-induced expression of apoptotic while still rescuing

neurons from death. Additional experiments were carried out to understand this disparity

between the failures of afobazole to prevent the up-regulation of pro-apoptotic genes while retaining the ability to mitigate neuronal death. Although the neurons were still alive they were in a senescent state and were unresponsive to depolarization by high K+.

However, these findings are still positive due to the ability of afobazole to delay neuron death, thus minimalizing the toxic environment of the penumbra.

x

These comorbidities of ischemia and Aβ toxicity may lead to potentiated responses and increase the risk for various vascular dementias. It was of particular interest to study how the convergence of ischemia, acidosis and Aβ influence cellular activity and survival within core and penumbral regions. Application of Aβ increased the

2+ [Ca ]i overload produced by concurrent ischemia + acidosis application in isolated cortical neurons. We found that the acid-sensing ion channels 1a (ASIC1a) are involved

2+ in the potentiation of [Ca ]i overload induced by Aβ. Furthermore, afobazole (100 µM)

2+ abolished Aβ potentiation of ischemia + acidosis evoked [Ca ]i overload, which may represent a therapeutic strategy for mitigating injury produced by Aβ and stroke.

xi

CHAPTER 1

Introduction

Significance

Ischemic stroke often overlaps with diseases involving Aβ, such as Alzheimer’s disease (AD) and cerebral amyloid angiopathy. These comorbidities can lead to

potentiated responses that ultimately result in vascular dementia (Wilcock et al., 2009,

Nelson et al., 2013, Sudduth et al., 2014). The mechanisms contributing to this potentiation remain poorly understood. One key player in the pathobiology both of stroke

2+ and Aβ toxicity is the dysregulation of [Ca ]i homeostasis (Cuevas et al., 2011a, Cuevas

et al., 2011b, Behensky et al., 2013a, b). Previous studies have suggested that a link

may exist between cerebral ischemia and AD (Koistinaho and Koistinaho, 2005, Pluta et

al., 2013) due to the increased Aβ expression following an ischemic stroke (Selkoe,

2001, Shiota et al., 2013). The Aβ fragment, Aβ25-35, which has been found in AD brains,

has been proposed as a contributor to the pathogenesis of AD (Kaminsky et al., 2010).

This Aβ fragment has been shown to induce injury via various mechanisms including

production of reactive oxygen species (ROS) (Schubert et al., 1995), dysregulation in

intracellular Ca2+ (Joseph and Han, 1992), enhanced NMDA channel activity (Molnar et

al., 2004), and increased activation of caspase-3 (Marin et al., 2000). In addition, Aβ has been shown to initiate a sequence of events in microglia, including a pro-inflammatory

1

cascade and enhanced hemichannel formation, which ultimately results in neuronal and glial cell death (Orellana et al., 2011). Aβ toxicity is therefore of particular interest in lacunar stroke, where following a micro infarct, Aβ production increases thereby disrupting both the microvascular and the intracellular calcium levels in the neighboring neurons. This resulting Aβ toxicity may promote future infarcts with potentiated injury.

Studies have shown that in mice with increased amyloid precursor protein expression, there is an increased risk of ischemic stroke, but as of yet there are no studies that can elucidate the molecular mechanisms by which this injury occurs (Zhang et al., 1997).

Background

Stroke is the second leading cause of death globally and continues to be a major cause of serious long-term disability (Go et al., 2013). In 2013 the reported annual cost associated of this disease in the United States was approximately $35 billion. This economic strain has driven the search for potential treatments to alleviate the debilitating effects of this disease. Stroke is the sudden loss or interruption of blood circulation to the brain. There are two types of stroke. Hemorrhagic stroke, where a blood vessel ruptures and bleeds into the brain. Although hemorrhagic only accounts for a small percentage of all strokes the survival rate for these patients is low with 60% dying within the first year following the insult. An ischemic stroke in the most common type of stroke accounting for about 87% and occurs when blood flow to the brain is interrupted. The clot can occur locally by a thrombus or from an embolus forming elsewhere in the body and migrating into the brain. For the present study, when referring to a stroke we are denoting an ischemic stroke. The central region affected by the ischemic insult, or the core, is an

2

area with little to no blood perfusion and dies within minutes of stroke onset via necrosis.

Outside of this core region is the penumbral zone, where some perfusion is maintained

by collateral arteries. In this area, delayed cell death occurs due to the triggering of

apoptotic mechanisms, and over the course of hours to days the size of the lesion

expands. The cellular and molecular events that produce the expansion of the ischemic

core continue to be poorly understood, which has hampered development of stroke

therapies designed to prevent neuronal and glial cell degeneration in the penumbra. In

addition to large vessel ischemic strokes there are also small vessel ischemic strokes.

Known as lacunar infarcts, these strokes account for approximately 25% of all ischemic

insults (Samuelsson et al., 1996). They result from the blockage of small penetrating

arteries to the subcortical regions of the brain (Kang et al., 2012). While the injury from

these strokes were once thought to be negligible, lacunar infarcts can and do result in

death and long-term disability. This type infarct is a leading cause of cognitive

impairment and vascular dementia (Charidimou and Werring, 2012). While the risk

factors resulting in lacunar verse large artery infarcts differ, the resulting cellular cascade

promoting cerebral cell death most likely have overlapping mechanisms.

Treatments have been explored to provide neuroprotection in an effort to mitigate

neuronal injury following ischemic stroke. One main area of research had been to inhibit

N-methyl-D-aspartate (NMDA) and (±)-α-Amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors. While some in vitro studies were promising, the in vivo results were less assuring. One such drug was Selfotel, a synthetic NMDA antagonist, which showed significant improvements in an animal model of transient focal ischemia

(Finnegan et al., 1993). However, Selfotel needed to be administered 1hr prior to the 3

ischemic insult for best results, and showed no benefit if treatment was started >60 min post stroke (Smith et al., 2008). Furthermore, clinical trials were terminated early due to adverse effects (Kurokawa et al., 2011a). One potential reason for the failures of neuroprotective drugs that target a single ion channel is the fact that ischemic stroke is a complex syndrome acting on a multitude of ion channels and receptors. Furthermore, recent studies have shown that microglia, part of the immune/inflammatory response, migrate to the site of insult, become over-activated and contribute to the stroke injury

(Eichhoff et al., 2011).

The only treatment currently approved by the FDA for ischemic stroke is recombinant tissue plasminogen activator (tPA), a thrombolytic agent used to restore blood-flow interrupted by a blood clot (Weintraub, 2006). However, tPA therapy is time sensitive, and the drug must be administered < 4.5 hours post-infarct. In addition, aging patients presenting various comorbidities lead to an increase in tPA’s risk to benefit ratio, resulting in less than 4% of patients being able to use it (Armstead et al., 2010). The goal of future therapies would be to treat these patients at 24 to 48 hrs follow the insult.

However, for this to be accomplished the mechanisms of stroke must be further elucidated.

Pathobiology of Ischemic Stroke

To better understand neurodegeneration following an ischemic stroke, it is important to study the mechanisms underlying stroke. Figure 1.1 shows a simplified overview of the biochemical events that are involved in ischemic stroke. Following an ischemic insult, the core rapidly depletes its energy stores due to decreased blood flow.

4

Figure 1.1

Pathoneurobiology of Ischemic Stroke. Intracellular calcium plays an important role in the events during ischemia. Ischemia leads to acidosis in the core and penumbra regions, which contributes to further calcium dysregulation. Calcium toxicity results in the activation these secondary events that result in apoptosis and subsequent expansion of the core.

The susceptibility of neurons to ischemia is in part due to their high consumption of

oxygen and glucose. Penumbra cell death is delayed in part due to blood flow from

collateral arteries supplying minimal oxygen and glucose. This reduced supply, however, is not enough to maintain normal cellular respiration and neurons are forced to switch from oxidative metabolism to anaerobic glycolysis. This provides neurons with some energy in the hopes that blood flow will be restored. However, lactate builds up within the

5

cells reducing the surrounding pH (Munoz Maniega et al., 2008). This drop in tissue pH, along with synaptic proton release triggered in neurons by ischemia, activates the acid sensing ion channel 1a (ASIC1a) (Xiong et al., 2004, Mari et al., 2010). ASIC1a is permeable to both Na+ and Ca2+ (Xiong et al., 2004) and following activation contributes

+ 2+ to the intracellular Na loading and [Ca ]i dysregulation seen following an ischemic

2+ stroke. This convergence of ischemia and acidosis has been shown to potentiate [Ca ]i

dyshomeostasis (Mari et al., 2010).

Despite neuronal attempts to generate ATP, the reduction in cerebral blood flow

causes a massive energy shortage. Energy depletion triggers cell depolarization due to

the failure of Na+/K+ ATPase (Besancon et al., 2008) and results in cations, Na+ and

Ca2+, rushing into the cell. The increasing presence of calcium triggers the release of

glutamate from the extracellular stores, which then over-activate NMDA and AMPA

receptors (Mark et al., 2001, Lipton, 2004, Muir, 2006), thus resulting in wide spread

intracellular ionic imbalances. In addition, because the Na+/K+ ATPase cannot function

+ + there is a buildup of intracellular sodium ([Na ]i) and extracellular potassium ([K ]o). The

+ increase in [K ]o triggers neighboring neurons to depolarize. Whereas, sodium loading leads to edema and results in the reversal of the Na+/Ca2+ and Na+/Ca2+ K+-dependent

2+ exchangers, which contribute to [Ca ]i dysfunction (Kiedrowski, 2007). This calcium

overload triggers reactive oxygen species (ROS) and nitrite oxide (NO) production, which in turn breakdown proteins, lipids and nucleic acids (Mattson, 2000, Camello-Almaraz et

2+ al., 2006). Additionally, the buildup of [Ca ]i triggers the activation of secondary events

including the release of cytochrome c from the mitochondria initiating the expression of

pro-apoptotic genes and the release of pro-inflammatory mediators (Figure 1.1). This in 6

turn triggers the activation of microglia and reactive astrocytes, which are then capable of exacerbating neuronal injury and expanding the ischemic core (Takano et al., 2009).

Moreover, despite studies which have shown that following an ischemic stroke there is an increase the expression of amyloid precursor protein (APP) and both β and γ secretase, which lead to increased Aβ production, there have been little to no studies addressing the neuronal and glial injury induced by Aβ following ischemia (Popa-Wagner et al., 1998, Guglielmotto et al., 2009, Pluta et al., 2013).

Amyloid-β

Amyloid-β (Aβ) is a peptide found in many diseases and is best known with contributing to Alzheimer’s disease. Aβ is formed following cleavage of the amyloid precursor protein (APP) by secretases. It was once believed that cleavage of APP would enter one of two pathways which would either result in a physiological (secretases α and

γ) or pathological (secretases β and γ) peptide (Zhang et al., 2011). However, we now know this to be incorrect and that both pathways produce physiologic and pathologic peptides. The functions of Aβ have been largely debated to have no physiological endogenous function, only pathological. However, in recent years low picomolar (pM) concentrations of Aβ has been shown to promote platelet aggregation and be essential to synaptic plasticity, memory formation and fear conditioning (Shen et al., 2008,

Lawrence et al., 2014). It is only when concentrations of Aβ elevate ≥ 200 pM that it becomes toxic (Puzzo and Arancio, 2013). It was previously thought that amyloid precursor protein was acting as a signaling molecule rather than Aβ to promote synaptic plasticity (Octave et al., 2013). However, studies by (Puzzo and Arancio, 2013) revealed

7

that siRNA against APP could impaired synaptic plasticity, which was rescued with concurrent Aβ1-42 application. While there have been studies for (Nagele et al., 2002)

and against (Small et al., 2007) Aβ binding to α-7 nicotinic acetylcholine receptors

(α7nAChRs), it was only recently that studies from (Lawrence et al., 2014) showed that

Aβ (pM) worked as an agonist on presynaptic α7nAChRs to enhance long-term

potentiation (LTP). The controversy as to whether Aβ functions as agonist or antagonist

is dependent on which fragment is binding. Aβ1-40 has been shown to inhibit

transmitter release via the antagonism of α7nAChRs (Nery et al., 2013, Ju

et al., 2014, Salamone et al., 2014), whereas, the fragments 1-42 and 25-35 both bind to

α7nAChRs and enhance their synaptic transmission (Talantova et al., 2013, Lee et al.,

2014).

Amyloid-β accumulation is more commonly associated with diseases such as

Alzheimer’s (AD) and cerebral amyloid angiopathy. However, an ischemic insult has been shown to increase APP mRNA by >200% (Pluta et al., 2009). It been suggested that this increase in APP might be protective following an insult (Koike et al., 2012).

However, the additional up-regulation of the β and γ secretases lead to increased

cleavage of APP and the deposition of Aβ in the penumbral zone (Guglielmotto et al.,

2009, Pluta et al., 2013). These depositions were found to be significantly higher on day

4 following the ischemic episode with peak tissue levels at day 7 (Stephenson et al.,

1992, Jablonski et al., 2011). Aβ levels remained significantly high when checked during a one year follow up, which has been postulated as a potential risk factor for developing

AD (Jablonski et al., 2011). In addition to an increase Aβ, there is also an increase in the hyperphosphorylation of tau following a stroke, which is known to contribute to AD (Wen 8

et al., 2004). There is some evidence that a stroke or other cerebrovascular disease liken the chances of developing AD. It was found during the autopsies of AD patients that

approximately 40% had some form of cerebrovascular incident prior to developing AD.

With studies showing the increase in Aβ, many view Aβ as a potential target in the

prevention of AD. However, there are virtually no studies looking at the injury Aβ incurs

directly following a stroke. There are more than a few overlapping mechanisms between

ischemia and Aβ induced toxicity. Aβ has been shown to disrupt ionic homeostasis

through several pathways, which involve Aβ having a direct and indirect action on

neuronal and glial cells. Aβ can cause the release of astrocytic and microglial glutamate

that activates neuronal AMPA, NMDA or metabotropic glutamate receptors, as well as,

being able to directly bind to neuronal NMDA receptors, whereby increasing the influx of

Na+ and Ca2+ ions (Alberdi et al., 2010, Texido et al., 2011, Talantova et al., 2013). This

influx of Na+ and Ca2+ leads to an increase in Ca2+ dyshomeostasis, triggering

mitochondrial dysfunction and initiating apoptosis (Cha et al., 2012, Oseki et al., 2014,

2+ Ferreira et al., 2015). As previously mentioned, the dysregulation of [Ca ]i leads to

secondary responses including activation of reactive astrocytes and microglia (Figure

1.1).

Microglia

Microglia are the resident macrophages of the brain, and constitute nearly 10% of

the total cell population of this organ. The function of these cells in normal and

pathological conditions has been an area of significant research, and considerable

controversy surrounds the role of these cells in diseases of the CNS. It has been shown

9

that microglia transform from a resting to an activated state in response to various

stimuli, such as neuronal injury, release of ATP, or pathogen infiltration (Melani et al.,

2005, Hanisch and Kettenmann, 2007). Upon stimulation, these cells migrate to the site

of injury, phagocytose debris and release pro-inflammatory compounds (Hanisch and

Kettenmann, 2007). Such a microglial response has been reported to occur as a result of a cerebral ischemic insult, following which microglia are attracted to the site of injury and proliferate (Weinstein et al., 2010). In addition, microglial cells have been shown to be

attracted to, and are required for, the clearance of Aβ (Wilcock et al., 2004, Morgan,

2009). Once phagocytosed, Aβ is degraded by the endothelin-converting enzymes in the

endosomal/ lysosomal pathway (Pacheco-Quinto and Eckman, 2013).

Since it has been shown that Aβ concentrations increase following an ischemic

insult it has been suggested that these microglia then contribute to the

neuroinflammatory response that results in the demise of neurons (Rogove et al., 2002,

Streit et al., 2004, Dheen et al., 2007). It is plausible that in brains with abnormally high

concentrations of Aβ, the inflammatory injury normally caused by microglial migration

following ischemia increases. This dual insult of ischemia and Aβ results in the over-

activation of microglial cells and maintaining prolonged microglia activation leads to cell

death or senescence. The loss of functional microglia to clear the accumulation Aβ may

lead to various vasculature dementias including cerebral amyloid angiopathy and AD. As

of yet, there have been no successful therapies in the treatment of stroke or AD. These

failures are compounded by not addressing the multiple receptor and/or pathways to

injury, in addition to not accounting for the interactions between multiple cell types. One

10

potential target for the treatment of stroke are the sigma receptors, which have been

shown to interact with multiple receptors, pathways and cell types.

Sigma Receptors

Sigma (σ) receptors were first identified by(Martin et al., 1976) and classified as

an receptor following the binding actions observed with racemic .

However, it was later discovered that only the (+) enantiomer of benzomorphans bound

to σ receptors, which was found to be inaccessible to placing them in their

current class of “opioid-like receptors” (Walker et al., 1990). σ receptors have been

shown to bind to a wide variety of substances including , opiates and

to name a few. Studies by (Hellewell and Bowen, 1990) showed a

decreased affinity for (+)-benzomorphans in PC12 cells suggesting a second σ subtype.

Thus far there have been two σ receptor subtypes identified pharmacologically, termed

σ-1 and σ-2. Receptors that bind (+)-benzomorphans with high affinity are classified as

σ-1receptors. Whereas, those that have a high affinity for are labeled σ-2

receptors (Bowen, 2000, Vilner and Bowen, 2000). Moreover, some drugs have been

shown to be pan-selective for both sigma receptors. This includes the pan-agonist 1,3-di-

o-tolyguanidine, the pan-antagonist and the σ-1 antagonist and σ-2 agonist .

Thus far, only the σ-1 receptor has been cloned. Consisting of 223 amino acids, this receptor shows no homology with any other mammalian protein (Seth et al., 1998,

Mei and Pasternak, 2001). Predictions from the early studies of the amino acid sequence suggested two different but possible transmembrane configurations. The first was a

11

single transmembrane domain with an extracellular N-terminus and intracellular C- terminus. Whereas, the second possibility retain the N- and C-termini intracellularly with two transmembrane spanning domains (Guitart et al., 2004), with more recent studies favoring the latter (Pal et al., 2007). Moreover, these studies predict that σ-1 receptors have two intracellular drug binding domains. In depth pharmacological studies by

(Glennon, 2005) describe the σ-1 receptor binding site as a figure eight configuration with a central amine moiety flanked by two hydrophobic regions, one of which consisting an aromatic ring. These studies were reaffirmed with the discovery of the σ receptors endogenous ligand N,N-dimethyltryptamine (DMT) (Fontanilla et al., 2009). Furthermore,

σ-1 receptors were shown to have a DMT concentration dependent action, which at low concentrations of DMT, σ-1 receptors would act as a chaperone between the rough endoplasmic reticulum (ER) and the mitochondria at the mitochondrion-associated ER membrane (MAM) to regulate intra-mitochondrial calcium concentrations. However, at high DMT concentrations, σ-1 receptors would migrate to the plasma membrane or subplasmalemmal ER to regulate various channels and receptors (Su et al., 2009, Tsai et al., 2009, Su et al., 2010). This makes σ-1 receptors highly attractive as a therapeutic target for numerous neurological diseases.

Despite years of study, the identity of σ-2 receptors is still unknown. There have been studies in recent years suggesting the σ-2 receptors might be either a histone binding protein (Colabufo et al., 2006, Abate et al., 2010) or the receptor membrane component-1 (PGRMC-1) (Xu et al., 2011, Abate et al., 2015). However, the molecular identity of σ-2 receptors has not been unequivocally established. In studies using human SK-N-SH neuroblastoma cells there is evidence to support that σ-2 12

receptors also localize with the ER, mitochondria, lysosomes and plasma membrane

(Vilner and Bowen, 2000, Zeng et al., 2007, Cassano et al., 2009). It has been

suggested that σ-2 receptors may be a splice variant of the σ-1 receptor (Monassier and

Bousquet, 2002). However, this hypothesis was refuted by the finding that σ-1 receptor

knockout mice still retained σ-2 receptor function (Langa et al., 2003).

The distribution of σ receptor in the mammalian body is quite ubiquitous being

expressed in the central nervous, liver, kidney, immune and cardiac cells (Dumont and

Lemaire, 1991, Wolfe and De Souza, 1993, Hellewell et al., 1994). Not much is known

about the functions of σ-2 receptors. However, what is known about their function varies

greatly depending on the cell type. In cancer cells, activation of σ-2 receptors causes the

toxic release of calcium from ER stores leading to apoptosis (Vilner and Bowen, 2000,

Cassano et al., 2009). Whereas, in immune cells σ-2 receptors are protective by

mitigating the activation and migration of microglia and T lymphocytes. Additional, σ-2

receptors have been shown to inhibit high-voltage-activated calcium channels in

intracardiac neurons (Zhang and Cuevas, 2002, Hall et al., 2009a, Iniguez et al., 2013).

In contrast to σ-2, σ-1 receptors have been identified as having extensive physiological

and pathophysiological functions. They are known to promote learning, memory and

synaptic plasticity in normal and in experimental stroke models (Ruscher et al., 2011,

Kourrich et al., 2012). Moreover, σ-1 receptors maintain intracellular ionic homeostasis

through regulation of 1,4,5-triphosphate (IP3) and ryanodine receptors at the

endoplasmic reticulum, as well as calcium signaling and the influx of Na+ and Ca2+ through voltage-gated, ionotropic and metabotropic receptors (Hayashi et al., 2000, Su and Hayashi, 2003, Tchedre et al., 2008, Mueller et al., 2013). 13

There are numerous studies showing the cytoprotective ability of σ-1 receptors, both in neurons and glial cells. σ-1 receptors have been shown to regulate intracellular calcium increases following in vitro models of ischemia, acidosis and ischemia-acidosis

(Katnik et al., 2006, Herrera et al., 2008, Mari et al., 2014). In addition, σ-1 receptor activation reduces oxidative and nitrosative stresses, while inhibiting apoptosis (Tchedre

and Yorio, 2008, Meunier and Hayashi, 2010, Pal et al., 2012). Following the in vivo

middle cerebral artery occlusion (MCAO) stroke model σ receptor activation was able to

reduce the injury of both white and grey matter when the agonist was administered 24

hours after the insult (Ajmo et al., 2006, Leonardo et al., 2010). Lastly, σ receptor

activation has been shown to be neuroprotective against Aβ-induced toxicity (Marrazzo et al., 2005, Behensky et al., 2013a, b), which as previously mentioned is up-regulated following an ischemic stroke. Therefore, σ receptor agonists offer great potential for the treatment of ischemic stroke and related Aβ toxicity. One such pan-selective σ receptor agonist which has shown promise is afobazole.

Afobazole

Afobazole (5-ethoxy-2-[2-(morpholino)-ethylthio]benzimidazole), is a drug currently used in Russia to treat anxiety and has shown the potential to be neuroprotective in both in vitro and in vivo models (Galaeva et al., 2005, Zenina et al.,

2005, Seredenin et al., 2008). In vitro and in vivo studies have shown that afobazole can

decrease neuronal death caused by oxidative stress and glutamate excitotoxicity and

increase survival rates, while decreasing neurological deficits in rats suffering from a

post-traumatic hematoma, respectively (Galaeva et al., 2005, Zenina et al., 2005).

14

Similarly, afobazole was shown to be neuroprotective in rats after bilateral local

photothrombosis of vessels in the prefrontal cortex and after global transient ischemia

(Seredenin et al., 2008, Baykova et al., 2011). Previous work in our laboratory has

shown that afobazole can reduce ischemic injury following a stroke. These studies

showed that afobazole affects cellular responses to ischemia via the stimulation of σ

receptors (Cuevas et al., 2011a, Cuevas et al., 2011b). Activation of σ-1 receptors by

afobazole blocks intracellular calcium overload produced by ischemia and acidosis and

reduces neuronal death after in vitro ischemia (Cuevas et al., 2011a). This activation of

σ-1 receptors results in the block of multiple ion channels that are functionally upregulated following ischemia and acidosis, including the acid-sensing ion channel 1a

(ASIC1a) (Cuevas et al., 2011a). In microglia, activation of both σ-1 and σ-2 receptors by afobazole reduces membrane ruffling and migration of these cells in response to ATP

(Cuevas et al., 2011b). Moreover, afobazole prevents microglial cell death following ischemia, even when the drug was applied after the ischemic insult (Cuevas et al.,

2011b). Lastly, in vivo studies using the middle cerebral artery occlusion (MCAO) stroke model showed that afobazole through σ receptor activation was able to reduce the total stroke injury, which resulted in and increased survival of both neuronal and glial cells even when administered 24-48 hrs post infarct (Katnik et al., 2014). The protection conveyed by afobazole translated to an increase in the animal’s behavior and cognition.

Therefore, studies suggest that afobazole, acting on σ receptors, can decrease ischemic brain injury as well as improving functional outcomes. This makes afobazole an attractive

15

drug, not only for the treatment of ischemic stroke but also for the treatment of Aβ- induced toxicity, which may contribute to secondary infarcts.

16

CHAPTER 2

In Vitro Evaluation of Guanidine Analogues as Sigma Receptor Ligands for Potential Anti-Stroke Therapeutics

Introduction

Treatments that provide neuroprotection have been explored in an effort to

mitigate neuronal injury following ischemic stroke. The main area of research has been

inhibition of N-methyl-D-aspartate (NMDA) and (±)-α-Amino-3-hydroxy-5- methylisoxazole-4-propionic acid (AMPA) receptors. While some in vitro studies were promising, the in vivo results were less assuring. For example, Selfotel, a synthetic

NMDA antagonist, showed significant improvements in an animal model of transient focal ischemia (Finnegan et al., 1993). However, Selfotel needed to be administered 1hr prior to the ischemic insult for best results, and showed no benefit if treatment was started >60 min post stroke (Smith et al., 2008). Furthermore, clinical trials were terminated early due to adverse effects (Kurokawa et al., 2011a).

One reason for the failure of neuroprotective drugs that target a single ion channel is the fact that ischemic stroke is a complex syndrome. For example, after an ischemic insult, neurons switch from oxidative metabolism to anaerobic glycolysis, leading to the

Reprinted with permission of the American Society for Pharmacology and Experimental Therapeutics. Copyright © 2013 by the American Society for Pharmacology and Experimental Therapeutics

17

buildup of lactate within the cells and a reduction in tissue pH (Munoz Maniega et al.,

2008).

This drop in tissue pH, along with synaptic proton release triggered in neurons by

ischemia, activates the acid sensing ion channel 1a (ASIC1a) (Xiong et al., 2004, Mari et

2+ al., 2010). These channels contribute directly and indirectly to [Ca ]i dysregulation in

neurons during ischemia (Herrera et al., 2008), and are responsible for significant NMDA channel-independent brain injury following an stroke (Xiong et al., 2004). In addition,

activated microglia migrate to the site of insult and are part of the immune/inflammatory

response that significantly contributes to stroke injury (Eichhoff et al., 2011).

Sigma (σ) receptors have been shown to be widely expressed in mammalian

brain tissue (Cobos et al., 2008) and perform a number of functions, including regulation

of plasma membrane ion channels and expression of anti-apoptotic transcription factors

(Zhang and Cuevas, 2002, 2005, Meunier and Hayashi, 2009, Kurokawa et al., 2011b).

Activation of σ receptors was shown to regulate multiple Ca2+ influx pathways known to be functionally upregulated during stroke, including ASIC1a (Katnik et al., 2006, Mari et al., 2010). In light of these findings and other reports in the literature, there has been much interest in exploring σ receptors as neuroprotective mediators during an ischemic stroke. One of the most exciting discoveries relating to σ receptors in stroke therapeutics is the potential of these receptors as viable targets for stroke therapy at delayed time points. Our group showed that activation of σ receptors using o-DTG reduced neuronal

injury in an animal model of ischemic stroke even when the drug was administered 24 hr

post-stroke (Ajmo et al., 2006).

18

o-DTG is a pan-selective agonist of σ receptors, binding to both σ-1 and σ-2 receptors at nanomolar concentrations, interacting with the σ-1 receptor via an amine moiety on the guanidine core (Glennon, 2005). From this parent molecule, novel compounds were designed and synthesized with the objective of developing σ ligands

2+ that inhibit increases in [Ca ]i with higher potency during in vitro experiments.

Experiments were conducted to compare the capacity of DTG analogues to affect three

important contributors to the demise of brain cells following ischemic stroke: intracellular

Ca2+ dysregulation produced in neurons by 1) acidosis and 2) ischemia, and 3) activation

and migration of microglial cells. While previous studies using guanidine substitutions

were made in an attempt to characterize the structure-affinity relationships (Reddy et al.,

1994, Schetz et al., 2007) our focus was to identify what affects steric hindrance,

electrostatic interactions, or increased lipid permeability had on the structure-activity

2+ relationship in mitigating increases in [Ca ]i. p-BrDPhG showed the greatest block in

2+ inhibiting increase in [Ca ]i evoked via ischemia or acidosis and in mitigating activation

and migration of microglial cells.

Materials and Methods

Preparation of Cortical Neurons

All experiments were carried out on cultured cortical neurons from mixed sex

embryonic day 18 (E18) Sprague-Dawley rats (Harlan, Indianapolis, IN). Methods used

here were identical to those previously reported for the isolation and culturing of these

cells (Katnik et al., 2006). Cortical neurons were used between 10-21 days in culture,

which permits synaptic contact formation and yields the most robust responses to 19

ischemia and acidosis. Animals were cared for in accordance with the regulations and

guidelines set forth by the University of South Florida’s College of Medicine Institution on

Animal Care and Use Committee.

Calcium Imaging

Intracellular Ca2+ concentration was measured in isolated cortical neurons using

ratiometric fluorometry as previously described (Katnik et al., 2006). Cells plated on poly-

L-lysine coated coverslips were incubated at room temperature for 1 hr in B27 supplemented Neurobasal medium (Invitrogen) containing 3 µg/ml of the acetoxymethylester form of fura-2 (fura-2 AM) in 0.3% DMSO. Prior to beginning the experiments, the coverslips were rinsed with physiological saline solution (PSS). For experiments in which cells were exposed to in vitro ischemia, the control PSS (PSS-1) consisted of (in mM): 140 NaCl, 3 KCl, 10 HEPES, 7.8 glucose, 2.5 CaCl2, and 1.2

MgCl2 (pH to 7.2 with NaOH). The control PSS (PSS-2) for acidosis experiments contained (in mM): 140 NaCl, 4.5 KCl, 25 HEPES, 20 glucose, 1.3 CaCl2, and 1.0 MgCl2

(pH to 7.4 with NaOH).

PSS was used as the control solution in all experiments. Solutions were delivered

onto the cells via a rapid application system identical to that described previously

(Cuevas and Berg, 1998). In vitro ischemia was achieved using glucose-free PSS-1

containing 4 mM sodium azide (NaN3) (Katnik et al., 2006) and acidosis was evoked via

application of PSS-2 with a pH of 6.0 (Herrera et al., 2008). Cells were only exposed to

three or fewer episodes of ischemia or acidosis separated by 10 min wash to prevent

rundown of cellular responses. For both ischemia and acidosis experiments, control

20

recordings were made prior to application of σ ligands. In experiments using the σ-1

inhibitor, BD-1063, a second control recording was made in the presence of the inhibitor

prior to application of p-BrDPhG. All test compounds were applied in PSS for 10 min

prior to ischemia or acidosis onset, and throughout the ischemic or acidic insult.

In Vitro Competition Binding Assays

Rat liver P2 membrane (~350 µg protein) was used for all σ binding assays.

Membrane homogenates were incubated for 120 min in 50 mM Tris-HCl buffer, pH 8.0,

with radioligand and various concentrations of test ligand in a total volume of 500 µl, at

25 C, in 96-well Unifilter GF/B filter plates (Perkin Elmer 50-905-1601). σ-1 receptors

were⁰ labeled with 5 nM [3H](+)-, and σ-2 receptors were labeled with 3 nM

[3H]o-DTG in the presence of 300 nM (+)-pentazocine to block σ-1 receptors. Ten concentrations of each test ligand (0.001-10,000 nM) were run in triplicate. Nonspecific binding was determined in the presence of 50 µM haloperidol, with filters pre-soaked in polyethyleneimine for 45 min. Following incubation and subsequent equilibrium, the samples were harvested, washed three times, and the bound radioactivity was counted.

Data from the competition binding studies were analyzed using GraphPad Prism (San

Diego, CA) and nonlinear regression to determine the concentration of test ligand that inhibits 50% of the specific binding of the radioligand (IC50 value). Ki values were

calculated using the Cheng-Prusoff equation.

Primary Cultures of Microglia

Primary cultures of microglia were prepared from Sprague-Dawley mixed sex rat

pups (post-natal day 2-3) as previously described by our laboratory (Hall et al., 2009a, 21

Cuevas et al., 2011b). The mixed glial cultures were incubated for 7-12 days at 37 o C

prior to experiments being carried out. Microglia were mechanically separated from the

cultures by brief shaking. Isolated microglia were resuspended in DMEM PLUS

containing: 500 mL Dulbecco’s Modified Eagle Medium with 4.5 g/L glucose, L-glutamine

and sodium pyruvate (DMEM) (Corning Cellgro, Manassas, VA), 40 mL horse serum

(Corning Cellgro), 12.5 mL heat-inactivated fetal bovine serum (Thermo Scientific

Hyclone, Logan, UT ), and 5 mL 10x antibiotic/antimycotic (Corning Cellgro). These cells

were used immediately for migration and cytotoxicity assays or plated on glass

coverslips for one day for imaging experiments.

Microglial Migration

The migration assays were carried out using a 48-well microchemotaxis chamber

(Neuro Probe, Inc., Gaithersburg, MD) as described previously (Cuevas et al., 2011b).

The bottom wells of the chamber were filled with DMEM containing the chemoattractant,

ATP (100 µM), and 0.1% DMSO. In the upper chamber, 1x106 freshly isolated microglia

were applied in DMEM. The test compounds, p-BrDPhG (30 µM) and o-DTG (30 µM), were applied both with the ATP and the microglia in the appropriate wells. These chambers were separated by a polycarbonate membrane containing 8 µm pores at a

density of 1 x 103 pores/mm2. Each well has an exposed filter area of 8 mm2. Prior to

experiments, the membrane was coated with fibronectin at 10 µg/ml in phosphate buffer

solution (PBS) for 1 hr at room temperature. The microglia were permitted to migrate for

o 2 hr in a CO2 incubator (5% CO2) at 37 C. The membrane was then removed and non- migrating microglia adhering to the top of the membrane were scraped off. The

22

membrane was then incubated in 4% paraformaldehyde in PBS for 15 min at room

temperature, washed 2 times in ice cold PBS, and washed once in distilled water. The

membrane was then cut to separate the wells and placed on microscope slides, bottom

of the membrane facing up. Once the membrane dried, Vectashield Hardset mounting

media containing 4'-6-diamidino-2-phenylindole (DAPI) (Vector Labs, Burlingame, CA)

was applied to the membrane and a coverslip affixed to the slide. Cells were illuminated

at 359 nm and visualized at 461 nm using a Zeiss Axioskop 2 outfitted with a 20X

objective. DAPI positive cells were identified and counted using Image-J in four random

fields/well and the average for the fields used as the value for the well. For each

experiment, a minimum of three wells were used, and the results of at least three

experiments were averaged together.

Drugs and Chemicals

The following drugs were used in this investigation: o-DTG and BD-1063 (Tocris

Biosciences, Ellisville, MO); adenosine 5′-triphosphate disodium salt hydrate (Sigma-

Aldrich, St. Louis, MO); and fura-2 acetoxymethyl ester (Invitrogen). The DTG analogues

were synthesized in our laboratory via copper-catalyzed-cross-coupling guanidinylation

as we have previously reported (Cortes-Salva et al., 2010). The structures of the DTG

analogues synthesized were subjected to verification via 13C-, 1H-NMR and mass

spectrometry. Vehicles for drugs used were either dimethyl sulfoxide (DMSO) or ethanol

(EtOH).

23

Data Analysis:

Fluorescence intensities were recorded from cortical neurons in the same manner

as previously described (Katnik et al., 2006, Herrera et al., 2008). Data were analyzed

using Sigma Plot/Sigma Stat 11(Systat Software, Inc., San Jose, CA). Data points

represent peak means ± standard error of the mean (S.E.M.). Paired and unpaired

Student’s t tests were used to determine significant differences within- and between- groups, respectively. Results were considered statistically significant if p < 0.05. One-

and two-way analysis of variance were used for determining significant differences for

multiple group comparisons, as appropriate, followed by post hoc analysis using either a

Tukey or Dunn’s test. Mean relative inhibitions were calculated using the following

equation: Mean Relative Inhibition = 𝐶𝐶−𝑋𝑋 𝐶𝐶 𝐶𝐶−𝐷𝐷𝐷𝐷𝐷𝐷 � 𝐶𝐶 � where C is the control response, X is the response in the presence of the test compound,

and DTG represents the response in the presence of 100 µM o-DTG. The Langmuir-Hill

equation was used to analyze concentration-response data.

Results

Experiments were conducted to determine the effects of o-DTG and derivatives

2+ on elevations in [Ca ]i induced in cultured cortical neurons by ischemia. Figure 2.1A

2+ shows representative traces of [Ca ]i as a function of time recorded from four neurons

during ischemia in the absence (Control) and presence of the indicated drugs (all at 100

µM). Similar inhibitions were observed for all of the compounds shown. Identical

experiments showed that while removal of the methyl moiety resulted in a compound

24

(DPhG) that was more potent than m-DTG and p-DTG, none of these compounds were statistically more potent than o-DTG in this assay (Figure. 2.1B).

Figure 2.1

Shifting the location of the methyl moiety produces o-DTG analogues that inhibit 2+ ischemic-induced increases in [Ca ]i similar to the parent compound. A, representative traces of intracellular calcium as a function of time recorded from four neurons during chemical ischemia in the absence (Control) and presence of the indicated compounds. 2+ B, mean change in peak relative inhibition of [Ca ]i (±S.E.M.) obtained in response to ischemia for the indicated compounds all at 100 µM. Experiments preformed are identical to those in (A) (n > 47). Dagger indicates significant difference between DPhG and m-, p-DTG (p < 0.05).

25

Recent studies have shown that activation of σ receptors inhibit acidosis-evoked

2+ [Ca ]i overload (Herrera et al., 2008, Cuevas et al., 2011a). Experiments were

conducted to determine whether migration or removal of the methyl moiety would alter

Figure 2.2

Removal, but not migration, of the methyl moiety increases capacity of o-DTG analogues 2+ 2+ to inhibit acidosis-evoked elevations in [Ca ]i. A, representative traces of [Ca ]i as a function of time recorded from four neurons during acidosis in the absence (Control) and presence of the indicated drug. B, bar graph of mean change in peak relative inhibition of 2+ acidosis-evoked [Ca ]i (±S.E.M.) for the indicated compounds, all at 100 µM; experiments are identical to (A). For each condition, n > 50. Asterisk denotes significant difference between o-DTG and DPhG (p < 0.05); Dagger represents significance between DPhG and m-DTG (p < 0.05).

26

2+ the ability of the o-DTG derivatives to inhibit acid-evoked elevations in [Ca ]i. Figure

2+ 2.2A shows representative traces of [Ca ]i recorded from four neurons during acidosis in

the absence (Control) and presence of the indicated DTG analogues. Data show that all

2+ of these compounds inhibit the acid-induced elevations in [Ca ]i. While m- and p-DTG showed similar potency relative to o-DTG, DPhG, which lacks the methyl moiety, produced a block statistically greater than the parent compound (p < 0.05) (Figure 2.2B)

Fluoro substitution of piperidine compounds has been shown to increase the lipophilicity of the molecules (de Candia et al., 2009) and several piperidine derivatives have long been used as σ receptor ligands (Nakazawa et al., 1998, Schetz et al., 2007).

Further experiments were conducted to evaluate whether methyl-to-fluoro substitution of

2+ o-DTG would result in increased drug potency. Representative traces of [Ca ]i as a

function of time recorded from three neurons are shown in Figure 2.3A. Cells were

exposed to ischemia in the absence (Control) and presence of fluoro substituted

2+ guanidine analogues in the ortho, meta, and para positions. Inhibitions of [Ca ]i

elevations were noted for all compounds. Identical experiments revealed that while these

compounds produced statistically significant inhibitions of ischemia evoked increases in

2+ [Ca ]i, none of the compounds were more potent than o-DTG (Figure 2.3B).

Furthermore, only p-FDPhG produced inhibition which was greater than that observed

for the compound with the methyl moiety at the same site (i.e. p-DTG). Figure 2.3C

2+ shows representative traces of [Ca ]i recorded from three neurons evoked by acidosis in

the absence (Control) and presence of the specified fluoro substituted analogues (all at

100 µM). In experiments identical to those previously shown, the fluoro substituted

guanidine analogues did not produce greater inhibitions relative to o-DTG or when 27

Figure 2.3

Fluoro substitution does not affect o-DTG analogue inhibition of ischemia- or acidosis- 2+ 2+ evoked increases in [Ca ]i. A, representative traces [Ca ]i as a function of time recorded from three neurons during chemical ischemia in the absence (Control) and 2+ presence of the indicated drug. B, mean change in peak relative inhibition of [Ca ]i (±S.E.M.) evoked by ischemia for the indicated compounds at 100 µM; measurements of experiments are identical to those in (A) (n > 58). Daggers indicate significance between o- and p-FDPhG from m-FDPhG (p < 0.05.). Pound symbol represents significant difference from relative inhibition produced by the methyl moiety in the equivalent position (p < 0.05). For comparison, block by the methyl compound is indicated by the 2+ line and arrow. C, representative traces of [Ca ]i as a function of time recorded from three neurons during acidosis in the absence (Control) and presence of the indicated 2+ drugs. D, bar graph of mean change in peak relative inhibition of [Ca ]i (±S.E.M.) obtained in response to acidosis for the indicated compounds at 100 µM (n > 67). There is no significant difference between o-DTG and the analogues.

28

compared to the compounds with a methyl moiety at the same site (i.e. m-DTG and p-

DTG; Figure 2.3C).

Halogen substitution is often used for enhancing permeability of a drug. It has been shown that replacing a single hydrogen residue with a chloro moiety in compounds with comparable size to DTG increased the hydrophobicity of a compound (Gerebtzoff et al., 2004). Experiments were carried out to assess how methyl-to-chloro substitution of

DTG in the ortho, meta, and para positions altered the ability of the compounds to affect

2+ [Ca ]i increases evoked by ischemia and acidosis. Figure 4A shows representative

2+ traces of [Ca ]i recorded from three neurons during episodes of ischemia in the absence

(Control) and presence of the chloro substituted compounds. Inhibition of ischemia-

2+ evoked elevations in [Ca ]i was observed for all compounds. However, only the chloro

moiety substituted at either the meta or the para position increased the potency of the

drug (Figure 2.4B). Both m- and p-ClDPhG yielded statistically significant increases in

2+ inhibition p < 0.05 and p < 0.001, respectively, of ischemia-induced elevations in [Ca ]i

relative to o-DTG. These compounds were also more potent than the compounds with a

methyl moiety at the equivalent position (Figure 2.4B). Figure 2.4C shows representative

2+ traces of [Ca ]i obtained from three neurons in response to acidosis in the absence

(Control) and presence of the indicated drugs. The pattern of inhibition of acidosis-

2+ evoked elevations in [Ca ]i produced by the chloro substituted compounds was similar

to that observed in the ischemia experiments. While the block produced by o-ClDPhG

did not show a significant difference when compared to o-DTG at the same

concentration, both m-ClDPhG and p-ClDPhG produced a statistically significant (p <

2+ 0.001) increase in the degree of block of acidosis-elicited [Ca ]i overload that was >2- 29

fold greater than that of o-DTG, and greater than that of the equivalent methyl

substitutions (Figure 2.4D).

Figure 2.4

Chloro substitution at the meta and para positions increase inhibition of intracellular 2+ calcium elevations. A, representative traces of [Ca ]i as a function of time recorded from three neurons during ischemia in the absence (Control) and presence of the drugs 2+ shown. B, bar graph of mean change in peak relative inhibition of [Ca ]i (±S.E.M.) induced by ischemia for the drugs indicated at 100 µM (n > 55). C, representative traces 2+ of [Ca ]i from three neurons recorded as a function of time induced by acidosis in the absence (Control) and presence of the drugs shown. D, bar graph of mean change in 2+ peak relative inhibition of [Ca ]i (± S.E.M.) evoked via acidosis for the drugs indicated at 100 µM (n > 59). For both (B) and (D), asterisks indicate significant differences between o-DTG and m- and p-ClDPhG groups (p < 0.001), daggers indicate statistical difference from o-ClDPhG (p < 0.001), and pound symbols represent significant differences from relative inhibition produced by the methyl moiety in the equivalent position (p < 0.001 for all). Lines and arrows in (B) and (D) mark the block by the methyl compound at each site, and are shown for comparison. 30

Experiments were also carried out to determine if the substitution of a bromo

moiety could likewise affect the potency of the compounds. Figure 2.5A shows

2+ representative traces of [Ca ]i recorded from three neurons responding to ischemia in

the absence (Control) and presence of o-, m-, or p-BrDPhG, with all compounds

producing some degree of block. The attenuation of ischemia-evoked elevations in

2+ [Ca ]i resulting when the ortho substituted compound (o-BrDPhG) was applied was

similar to that seen with o-DTG. However, both m-BrDPhG and p-BrDPhG blocked the

2+ [Ca ]i overload to a greater degree with the inhibition produced by each compound

being statistically greater than o-DTG, p < 0.001. The bromo substituted compounds also

2+ 2+ inhibited acidosis-evoked [Ca ]i increase. Representative traces of [Ca ]i recorded from

three neurons and demonstrating such inhibition by all of these compounds are shown in

Figure 2.5C. As with the ischemia assay, the m-BrDPhG and p-BrDPhG compounds

2+ were significantly more potent than o-DTG at inhibiting acidosis-induced [Ca ]i increases

p < 0.001, but o-BrDPhG was comparable in effectiveness to o-DTG (Figure 2.5D). In

both the ischemia and acidosis experiments, the m- and p-BrDPhG were more potent than the equivalent methyl substitutions (Figure 2.5B and 5D).

2+ The guanidine analogue that exhibited the most pronounced inhibition of [Ca ]i

overload in both assays was p-BrDPhG. Thus, further experiments were carried out to

precisely measure the concentration response relationship for p-BrDPhG inhibition of

2+ 2+ [Ca ]i overload produced by ischemia. Representative traces of [Ca ]i as a function of

time recorded from two neurons in response to ischemia in the absence (Control) and

presence of 10 µM and 100 µM o-DTG and p-BrDPhG are shown in Figure 2.6A and 6B,

2+ respectively. Inhibitions of the [Ca ]i elevations by both of these compounds showed a 31

Figure 2.5

Substitution of the methyl moiety with bromide at the meta and para positions enhances 2+ 2+ [Ca ]i inhibition. A, representative traces of [Ca ]i as a function of time recorded from three neurons induced via chemical ischemia in the absence (Control) and presence of the indicated bromo derivatives at 100 µM. B, mean relative inhibition of ischemia- 2+ induced change in peak [Ca ]i (±S.E.M.) evoked by bromo derivatives at 100 µM for 2+ each drug (n > 55). C, representative traces of [Ca ]i as a function of time recorded from three neurons during acidosis in the absence (Control) and presence of the drugs 2+ shown. D, bar graph of mean relative inhibition of change in peak [Ca ]i (±S.E.M.) induced via acidosis for the drugs indicated (all at 100 µM, n > 53). For both (B) and (D), asterisks indicate statistical differences from o-DTG (p < 0.001), daggers indicate significant difference from o-BrDPhG (p < 0.001), and pound symbols represents significant differences from the methyl moiety in that position (p < 0.001). For comparison, block by the methyl compounds in the equivalent site is indicated by the lines and arrows in (B) and (D).

32

2+ concentration dependence. Plots of the mean relative change in [Ca ]i as a function of

drug concentration for both o-DTG and p-BrDPhG are shown in Figure 2.6C. The data

were best fit by single-site Langmuir-Hill equations, and indicate a 34-fold greater

2+ potency for p-BrDPhG in inhibiting ischemia-evoked increases in [Ca ]i relative to o-

DTG.

Figure 2.6

2+ Inhibition of ischemia-evoked increases in [Ca ]i by p-BrDPhG is concentration- 2+ dependent. Representative traces of [Ca ]i recorded as a function of time from a single neuron during chemical ischemia in the absence (Control) and presence of 10 and 100 µM o-DTG (A) or in the absence (Control) and presence of 10 and 100 µM p-BrDPhG 2+ (B). C, concentration-response relationship for mean change in peak [Ca ]i (±S.E.M.) measured during chemical ischemia in the presence of o-DTG (open circle) and p- BrDPhG (closed circle). Drugs were normalized to their respective controls (ischemia in absence of drug for each cell). Lines were best fit to the data using single-site Langmuir- Hill equations with IC50 values and Hill coefficients of 74.7 µM and 0.58 (o-DTG), and 2.2 µM and 0.63 (p-BrDPhG), respectively. For each data point, n > 65. 33

Similar experiments were carried out to determine the concentration-response

2+ relationship for p-BrDPhG inhibition of elevations in [Ca ]i caused by acidosis. Figure

2+ 2.7A shows representative traces of [Ca ]i recorded as a function of time from a single

neuron during acidosis in the absence (Control) and presence of 10 and 100 µM p-

BrDPhG. Increasing the p-BrDPhG concentration resulted in further depression of

2+ acidosis-induced [Ca ]i overload. Figure 2.7B shows a plot of the mean change in peak

2+ [Ca ]i as a function of p-BrDPhG concentration obtained from multiple experiments.

These results show an 8-fold greater potency of p-BrDPhG in inhibiting increases in

2+ [Ca ]i induced via acidosis when compared to o-DTG. The efficacy of p-BrDPhG was similar to that of o-DTG in both the ischemia and the acidosis assay.

To determine if the o-DTG derivative p-BrDPhG retains the σ receptor binding properties of the parent compound, a binding assay was performed using o-DTG for comparison. Results of the binding study are presented in Table 2.1 and show that p-

BrDPhG binds both σ-1 and σ-2 receptors. However, the affinity for both receptors was decreased, with the affinity for σ-2 decreasing to a greater extent. The Ki for p-BrDPhG was statistically less than o-DTG at both σ-1 and σ-2 receptors, and the affinity of p-

BrDPhG for σ-2 receptors was significantly lower than for σ-1 receptors (for all, p <

0.001; Table 2.1. While the binding study suggests that p-BrDPhG can act via σ

2+ receptors to inhibit [Ca ]i dysregulation, functional experiments were carried out using

the σ-1 receptor antagonist, BD-1063, to confirm this possibility. Figures 2.8A and 8B

2+ show representative traces of [Ca ]i as a function of time recorded from two neurons in

response to acidosis in the absence (Control) and presence of 30 µM p-BrDPhG (Figure

2.8A) or p-BrDPhG with 10 nM BD-1063 (Figure 2.8B). p-BrDPhG produced a reduction 34

Figure 2.7

2+ p-BrDPhG inhibits acidosis-evoked increases in [Ca ]i in a concentration-dependent 2+ manner. A, representative traces of [Ca ]i recorded as a function of time from a single neuron during acidosis in the absence (Control) and presence of p-BrDPhG at the indicated concentrations. B, concentration-response relationship for mean change in 2+ peak [Ca ]i (±S.E.M.) recorded during acidosis in the absence and presence of p- BrDPhG. Responses obtained in the presence of p-BrDPhG were normalized to control (absence of drug) for each cell (n > 72). Black line represents best fit to the data p- BrDPhG using a Langmuir-Hill equation with an IC50 value of 13.5 µM and a Hill coefficient of 1.2. Presented for comparison was the best fit to the data obtained for o- 2+ DTG inhibition of acid-evoked elevations in [Ca ]i (gray line, IC50 = 109.3 µM; Hill coefficient, 0.9) (Herrera et al., 2008).

2+ in [Ca ]i in the absence and presence of BD-1063. However, less block by p-BrDPhG was noted when BD-1063 was included with the guanidine analogue. In identical

2+ experiments, a reduction in [Ca ]i was noted for both p-BrDPhG and p-BrDPhG + BD-

1063. However, the effects of p-BrDPhG were significantly reduced in the presence of

35

the σ-1 antagonist and this decrease was statistically significant (p < 0.001) (Figure

2+ 2.8C). BD-1063 alone had no effect on acidosis-induced [Ca ]i elevations (Figure 2.8C).

Figure 2.8

σ-1 receptor antagonist BD-1063 attenuates p-BrDPhG inhibition of acidosis-evoked 2+ 2+ increases in [Ca ]i. A, representative traces of [Ca ]i as a function of time recorded from a single neuron during acidosis in the absence (Control) and presence of 30 µM p- 2+ BrDPhG. B, representative traces of [Ca ]i as a function of time recorded from a single neuron during acidosis in the absence (Control) and presence of 10 nM BD-1063, or 10 nM BD-1063 + 30 µM p-BrDPhG (p-BrDPhG + BD-1063). C, Bar graph of relative mean 2+ change in peak [Ca ]i induced by acidosis (n > 50). Data were normalized to values obtained for each cell in the absence of σ ligands. Asterisks denote significant differences between p-BrDPhG and their respective DMEM (p > 0.001), and a dagger indicates a statistical difference between the p-BrDPhG groups of Control and BD-1063 (p > 0.001), pound denotes significance between p-BrDPhG with BD-1063 and the DMEM Control (p > 0.001). There was no difference between DMEM groups.

36

Figure 2.9

p-BrDPhG blocks the migration of microglia elicited by ATP. A, concentration-response relationship for p-BrDPhG (closed circles) and o-DTG (open circles) inhibition of ATP- induced (100 µM) migration of microglial cells. The black line represents the best fit to the o-DTG data using a Langmuir-Hill equation with an IC50 value and Hill coefficient of 23.33 µM and 0.67, respectively. The grey line represents the best fit to the p-BrDPhG data using a Langmuir-Hill equation with an IC50 value and Hill coefficient of 6.53 µM and 0.75, respectively. Points represent mean (±S.E.M.) and for all points, n > 9.

Recently, our laboratory showed that activation of σ receptors with pan-selective

ligands such as o-DTG or afobazole modulates microglial activation, and inhibits

microglial cell migration in response to chemoattractants such as ATP (Cuevas et al.,

2011b). This effect on microglial cell activation has been postulated to be a mechanism 37

by which σ receptor activation reduces ischemic stroke injury (Cuevas et al., 2011b). For

this reason, experiments were carried out to determine if p-BrDPhG is also more potent than o-DTG in preventing microglial cell migration. Figure 2.9 shows a plot of the mean number of migrating microglial cells as a function of p-BrDPhG concentration obtained from multiple experiments. Data from 30 µM p-BrDPhG were best fit using the Langmuir-

Hill equation with an IC50 value of 6.53 µM and a Hill coefficient of 0.53. Data from 30 µM

o-DTG were best fit using the Langmuir-Hill equation showed an IC50 = 23.33 µM and Hill

coefficient of 0.67. These results represent just under a 4-fold greater potency of p-

BrDPhG in inhibiting microglial migration induced via 100 µM ATP when compared to o-

DTG.

Table 2.1

o-DTG binds to both σ receptor subtypes with greater affinity than p-BrDPhG. Average Ki values ± S.E.M. determined from binding assays using membrane protein 3 extracts from rat livers. σ-1 receptors were labeled with 5 nM [ H](+)-pentazocine and σ- 2 receptors were labeled with 3 nM [3H] o-DTG in the presence of 300 nM (+)- pentazocine to block σ-1 receptors. Nonspecific binding was determined in the presence of 50 µM haloperidol. Asterisks indicate significant difference between p-BrDPhG and o- DTG binding at σ-1 (p < 0.01) and σ-2 (p < 0.001) receptors. Pound symbol denotes significant difference between p-BrDPhG affinity for σ-1 versus σ-2 sites.

Receptor Average Ki (nM) SEM

p-BrDPhG Sigma-1 296 46.01

Sigma-2 800.33 56.96

o-DTG Sigma-1 * 91.02 9.7

Sigma-2 * 60 8.76

38

Discussion

This study demonstrates three novel findings. First, methyl-to-chloro or methyl-to- bromo substitution of DTG in the meta and para positions results in new compounds with

2+ greater potency relative to o-DTG for inhibiting [Ca ]i overload induced by chemical ischemia or acidosis in neurons. Of the compounds tested, p-BrDPhG, is the most potent in these assays. Second, our data show that p-BrDPhG binds σ receptors with a reduced

2+ affinity relative to o-DTG, but that the inhibition of [Ca ]i overload continues to be dependent on σ-1 receptor activation. The fact that p-BrDPhG has reduced σ receptor binding but increased potency in the functional assay suggests that increased lipid permeability may account for the enhanced inhibition. Finally, p-BrDPhG is also more potent than o-DTG in preventing microglial migration in response to ATP, indicating that this novel compound can effectively reduce microglial activation at lower concentrations relative to o-DTG.

Our initial experiments involved shifting and removal of the methyl moiety of DTG to ascertain effects on in vitro functional assays used to predict therapeutic potential in ischemic stroke. Our findings with the DTG analogues show that rearrangement of the methyl moiety does not produce a compound which behaves differently from the parent

2+ compound in terms of modulating [Ca ]i increases induced via ischemia and acidosis.

Removal of the group entirely (DPhG) produces a significant 18.3 ± 0.05% increase over

2+ o-DTG in inhibiting elevations in [Ca ]i during acidosis. Similarly, DPhG show a 12.2 ±

0.04% greater inhibition than o-DTG in the ischemia assay, but this difference was not statistically significant. Previous studies have reported that the σ receptor binding affinities for o-, m-, and p-DTG are approximately 30, 50, and 530 nM, respectively; 39

whereas the affinity of DPhG for σ receptors is ~400 nM (Scherz et al., 1990, Reddy et al., 1994). Results from our functional studies, therefore, do not reflect the decrease in affinity observed for the p-DTG or DPhG. Calcium elevations evoked by ischemia and acidosis are in part due to activation of NMDA receptors (Katnik et al., 2006, Herrera et al., 2008). However, neither p-DTG nor DPhG shows increased affinity for NMDA receptors compared to o-DTG (Scherz et al., 1990, Reddy et al., 1994), and thus actions on that receptor cannot account for the discrepancy between σ receptor binding and functional effects. Since the earlier studies did not examine specifically σ receptor subtype binding affinity, the difference may be due to changes in affinity for one receptor subtype versus the other (i.e. σ-2 receptor affinity decreases significantly but σ-1 receptor affinity remains unchanged).

While migration and removal of the methyl moiety failed to alter functional properties of the molecule significantly, pronounced enhancement in potency were found with halide substitutions. Data presented here show that substituting a chloro for a methyl moiety in the meta and para positions of DTG produced compounds that had

~15% higher potency relative to the parent compound (o-DTG) at 100 µM concentration

2+ in the ischemia-induced increases in [Ca ]i assay. The enhancement in potency was

2+ even more pronounced in the acidosis-evoked [Ca ]i dysregulation assay. In this assay, m- and p-ClDPhG (100 µM) produced blocks of >100% greater than o-DTG. In contrast, substitution of a chloro moiety in the ortho position did not alter the functional effects of the parent compound in either the ischemia or acidosis assay. Similar observations were made using compounds with bromo substitutions (BrDPhG) of DTG in the meta and para

40

positions with an ~22% increase of inhibition during ischemia and an ~160% increase

during acidosis applications over o-DTG, respectively. The most potent compound tested

was p-BrDPhG, which had an IC50 indicating 8-fold and 34-fold greater potency over that

2+ 2+ of o-DTG in the acidosis [Ca ]i and ischemia [Ca ]i assays, respectively.

Functional assays indicated that p-BrDPhG was significantly more potent than o-

DTG, but a radioligand binding assay showed decreased affinity for both σ receptor

subtypes. Our experiments showed that o-DTG binds σ-1 and σ-2 receptors with an affinity in the range of 60-90 nM, which is similar to what was previously reported in the literature (Torrence-Campbell and Bowen, 1996). In contrast, p-BrDPhG was found to bind σ-1 and σ-2 receptors with 296 ± 46 nM and 800 ± 57 nM affinities, respectively, with the σ-2 receptor affinity being significantly lower. These values are similar to a previous report which indicates that p-BrDPhG binds guinea pig brain membrane extracts with an affinity of 540 ± 25 nM (Scherz et al., 1990). That study also demonstrated that p-BrDPhG has lower binding affinity for the NMDA receptor than the parent compound (o-DTG), and therefore, the increased functional potency cannot be explained by increased action at the NMDA receptor.

2+ Confirmation that p-BrDPhG mitigates [Ca ]i dysregulation via activation of σ-1

receptors was achieved via the use of the σ-1 receptor antagonist BD-1063. Using the

2+ acidosis [Ca ]i functional assay, BD-1063 was shown to inhibit the effects of p-BrDPhG

by >50% at a concentration of 10 nM, which is consistent with p-BrDPhG acting via σ-1

receptors (Matsumoto et al., 1995, Herrera et al., 2008). To date, only activation of the σ-

2+ 1 receptor has been shown to affect ischemia- and acidosis-evoked [Ca ]i dysregulation

41

(Katnik et al., 2006, Herrera et al., 2008), and thus effects by the σ-2 receptor were not tested in these assays.

The current study shows that p-BrDPhG also inhibits microglial activation and this

DTG analogue is 4-fold more potent than the parent compound and a second pan-

selective σ agonist, afobazole (Hall et al., 2009a, Cuevas et al., 2011b). Unlike ischemia-

2+ and acidosis-evoked [Ca ]i dysregulation, which appears to be exclusively affected by σ-

1 receptor activation, both σ-1 and σ-2 receptors can modulate microglial migration

(Cuevas et al., 2011b). This observation may explain why p-BrDPhG, which has even lower affinity for σ-2 than σ-1, is only 4-fold more potent than o-DTG in the microglial

2+ migration assay but 8- to 34-fold more potent than the parent compound in the [Ca ]i

dysregulation experiments. The observation that p-BrDPhG is more potent than o-DTG

in inhibiting microglial activation may have important therapeutic implications. Previous

studies have demonstrated that activation of microglia following an ischemic insult

results in the release of pro-inflammatory cytokines by these cells, which promotes

neuronal death (Lee et al., 2001).

One mechanism which may explain the discrepancy between decreased binding

affinity but increased functional potency of p-BrDPhG may be increased membrane

permeability of the compound. It has been shown that chloro and bromo substitution in

the para position on phenylalanine increases hydrophobicity and blood-brain barrier

permeability of a drug (Gentry et al., 1999). Likewise, halogenation of small molecules

has been demonstrated to increase membrane permeation (Gerebtzoff et al., 2004). It

has been shown that increasing membrane permeability of a σ ligand by deprotonation

42

increases potency of the compound, which was interpreted to suggest that the binding site for ligands such as BD-1063 are intracellular (Vilner and Bowen, 2000). Immuno- labeling studies have confirmed this hypothesis, showing that σ-1 receptors localize to the endoplasmic reticulum (Dussossoy et al., 1999, Seminerio et al., 2012). Moreover, the proposed binding site for drugs such as o-DTG are, in part, located in the transmembrane domain of the receptor (Pal et al., 2007). Thus, by increasing membrane permeability, chloro and bromo substitution more than compensate for the decreased binding affinity.

Our data, however, also suggest that factors other than membrane permeability are likely involved in the effects of the halogenation and methylation. Electronegativity and conformation of the molecule are also important. For example, fluoro substitution of a distal phenyl group of a piperidine greatly increased the lipophilicity of the compound

(de Candia et al., 2009). Despite the likely increased lipid permeability, our findings show

2+ that fluoro substitution does not result in increased inhibition of [Ca ]i dysregulation evoked via ischemia or acidosis relative to o-DTG. One possibility is that while the fluoro substitution may increase the drugs lipophilicity, the added increase in electronegativity caused by addition of a fluoro moiety may have disrupted binding of the compound to the

σ receptors. While fluorine has an electronegativity of 4.0, that of chlorine and bromine are ~ 3.0. Moreover, our data suggest that location of chloro and bromo substitutions on the phenyl ring for enhanced potency were limited to either the meta or para positions. A previous report on structure-activity relationship of DTG analogues suggested that the ortho position was the most tolerant to structural modifications, including addition of electronegative moiety (Scherz et al., 1990). However, that report did not distinguish 43

between σ-1 and σ-2 receptor binding, and thus the effects of those modifications on the

respective receptors remains to be determined.

In conclusion, these findings show that the chloro and bromo substituted

analogues of DTG have potential applications as anti-stroke therapeutics, showing a

greater potency for inhibiting intracellular calcium dysregulation over o-DTG. In addition, p-BrDPhG blocks microglial migration with greater potency than o-DTG, indicating that this DTG analogue will both provide neuroprotection and mitigate the neuroinflammatory response known to contribute to stroke injury. It is of significant interest to determine if the noted enhancement in potency of p-BrDPhG in vitro translates to superior in vivo outcomes.

44

CHAPTER 3

Afobazole Activation of Sigma-1 Receptors Modulates Neuronal Responses to Amyloid-β25-35

Introduction

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is the most common form of senile dementia in the United States (Gaugler et al., 2013). While the full pathophysiological underpinnings of AD remain elusive, one of the hallmarks of

AD is an increase in amyloid-β (Aβ) plaque formation in the brain (Selkoe, 2001). The Aβ fragment, Aβ25-35, which has been found in AD brains, has been proposed as a major

contributor to the pathogenesis of AD (Kaminsky et al., 2010). This Aβ fragment has

been shown to induce injury via various mechanisms including production of reactive

oxygen species (ROS) (Schubert et al., 1995), dysregulation in intracellular Ca2+ (Joseph

and Han, 1992), enhanced NMDA channel activity (Molnar et al., 2004), and increased

activation of caspase-3 (Marin et al., 2000).

Previous studies have shown that a possible link might exist between sigma

receptors and the etiology of AD. A loss of sigma binding sites has been observed in AD,

and specific σ-1 receptor polymorphisms have been associated with altered risk for this

Reprinted with permission of the American Society for Pharmacology and Experimental Therapeutics. Copyright © 2013 by the American Society for Pharmacology and Experimental Therapeutics

45

disease (Jansen et al., 1993, Uchida et al., 2005, Mishina et al., 2008, Feher et al.,

2012).

Sigma-1 selective agonists have also been shown to reduce memory loss produced by Aβ25-35 injections in mice (Maurice et al., 1998). Recently, tetrahydro-N, N- dimethyl-2, 2-diphenyl-3-furanmethanamine hydrochloride (ANAVEX2-73) was shown to reduce tau hyperphosphorylation and Aβ1-42 generation in a mouse model of Alzheimer’s disease via the activation of both σ-1 and muscarinic receptors (Lahmy et al., 2013).

However, it is unclear if other sigma receptor ligands, particularly ligands that do not affect muscarinic receptors, have similar neuroprotective properties. The molecular mechanism(s) by which σ-1 receptors decrease neuronal injury upon Aβ exposure have not been identified, and the effects of σ-2 receptor activation on neuronal responses to

Aβ need to be elucidated.

Sigma (σ) receptors, once thought to be a class of opioid receptors, are known to have widespread regulatory actions during neuropathophysiological states (Tsai et al.,

2009). The σ-1 receptor has been shown to be an inter-organelle chaperone localized to the mitochondrion-associated ER membrane (MAM) (Su et al., 2010). The σ-1 receptor-

MAM relationship plays important roles in cell signaling, mitochondrial function and

2+ 2+ maintaining intracellular Ca ([Ca ]i) homeostasis (Su et al., 2010). Furthermore, upon stimulation with high agonist concentrations, σ-1 receptors migrate to sub-plasmalemmal

ER or the plasma membrane and interact with various ion channels (Su et al., 2010). In

2+ response to ischemia or acidosis, activation of σ-1 receptors prevents [Ca ]i overload and concomitant cell death, likely via actions at multiple targets, including acid-sensing

46

ion channels and NMDA receptors (Katnik et al., 2006, Herrera et al., 2008). In addition

2+ to regulation of [Ca ]i homeostasis, other sigma receptor-mediated effects may

contribute to neuroprotection, including decreased production of ROS and reactive

nitrogen species (RNS), activation of the anti-apoptotic protein Bcl-2, and inhibition of the pro-apoptotic protein Bax (Tchedre and Yorio, 2008, Meunier and Hayashi, 2009,

Kourrich et al., 2012, Pal et al., 2012). It remains to be established if some or all of these effects are linked to σ receptor-mediated neuroprotection following neuronal ischemia, and if they contribute to neuroprotection following Aβ exposure.

Our laboratory recently showed that afobazole (5-ethoxy-2-[2-(morpholino)- ethylthio]benzimidazole), a drug currently used in Russia to treat anxiety and panic disorders, is both a σ-1 and σ-2 receptor agonist, and provides neuroprotection in an in vitro ischemia model (Cuevas et al., 2011a, Cuevas et al., 2011b). Unlike ANAVEX2-73,

afobazole does not interact with muscarinic receptors (Seredenin and Voronin, 2009).

Activation of σ-1 receptors by afobazole results in a decrease in ischemia-induced Ca2+

overload, which is due in part to inhibition of NMDA channel activation (Katnik et al.,

2006, Cuevas et al., 2011a). Previous studies have suggested other mechanisms for the

neuroprotective properties of afobazole, including decreased caspase-3 activation and

reduced oxidative stress (Zenina et al., 2005, Antipova et al., 2010). Thus, afobazole is

an excellent ligand for examining the relationship between sigma receptors and Aβ-

induced neurotoxicity, and is a potential candidate for the treatment of Alzheimer’s

disease.

47

Experiments were carried out to determine how afobazole affects neuronal

responses to the Aβ fragment, Aβ25-35. Afobazole mitigated Aβ25-35-evoked increases in

2+ [Ca ]i in neurons and these effects were blocked by inhibition of σ-1, but not σ-2 receptors. Afobazole was also found to lessen NO production in response to Aβ25-35 application, and reduced neuronal death. The decrease in cell death produced by afobazole was associated with reduced neuronal expression of the pro-apoptotic protein

Bax and the death protease caspase-3 and enhanced expression of the anti-apoptotic gene product, Bcl-2.

Materials and Methods

Preparation of Rat Cortical Neurons

All experiments were carried out on cultured cortical neurons from mixed sex

embryonic day 18 (E18) Sprague-Dawley rats (Harlan, Indianapolis, IN). Methods used

here were identical to those previously reported for the isolation and culturing of these

cells (Katnik et al., 2006). Cortical neurons were plated on poly-L-lysine coated

coverslips and cultured in B27 and L-glutamine supplemented Neurobasal medium

(Neurobasal Complete) (Life Technologies, Grand Island, NY). Cells were used for experiments after 10-21 days in culture, which permits synaptic contact formation and

yields robust responses to other pathophysiological conditions such as in vitro ischemia

and acidosis (Katnik et al., 2006, Herrera et al., 2008). Animals were cared for in accordance with the regulations and guidelines set forth by the University of South

Florida’s College of Medicine Institution on Animal Care and Use Committee.

48

Calcium Imaging

Intracellular Ca2+ concentrations were measured in isolated cortical neurons using

ratiometric fluorometry as previously described (Katnik et al., 2006). For most

2+ experiments, cultured neurons were incubated in 25 µM Aβ25-35 for 1-24 hr prior to [Ca ]i

being measured. Neurons were loaded with fura-2 by adding 3 µg/ml of the

acetoxymethylester form of fura-2 (fura-2 AM) in 0.3% DMSO to the media and incubating the cells at room temperature for 1 hr. Prior to beginning the experiments, the coverslips were rinsed with physiological saline solution (PSS) consisting of (in mM): 140

NaCl, 5.4 KCl, 25 HEPES, 20 glucose, 1.3 CaCl2, and 1.0 MgCl2 (pH to 7.4 with NaOH).

2+ For each cell, [Ca ]i was measured once a second for a 20 second period and the

2+ 2+ values averaged to determine baseline [Ca ]i. The Aβ-induced increases in [Ca ]i

2+ (∆[Ca ]i) in experiments using σ receptor antagonists were calculated by subtracting

2+ average [Ca ]i measured in control cells for each condition tested (i.e. absence or

presence of σ receptor ligand) from those recorded for the individual cells when Aβ was

added under the same conditions. In one series of experiments, the acute effects of

2+ Aβ25-35 on neuronal [Ca ]i were determined by first loading neurons with fura-2 and then

2+ administering Aβ25-35 via a rapid application system while measuring [Ca ]i.

ROS and NO Imaging

Intracellular ROS and NO concentrations were measured in isolated cortical

neurons using fluorometric digital microscopy (CoolSnap HQ2; Photometrics, Surrey,

BC, Canada). Cells were plated as described above and incubated for 24 hr at 37  C in

Neurobasal Complete containing 25 µM Aβ25-35 (± afobazole). Cultured cells were then 49

incubated at 37°C for 30 min in 10 µM DHE with 0.25% DMSO to measure ROS and for

1 hr in 5 µM DAF-FM with 0.1 % DMSO to measure NO, both in Neurobasal Complete

and with 25 µM Aβ25-35 (± afobazole). The fluorophores were then removed by rinsing

coverslips with physiological saline solution (PSS). DHE and DAF-FM loaded neurons were excited with light at 518 nm and 495 nm, respectively. ROS and NO concentrations were then measured as fluorescent emission intensities at 605 nm and

515 nm wavelengths, respectively. Image analysis was performed using Nikon Elements software (Nikon, Melville, NY, USA). No afobazole autofluorescence was observed at the wavelengths tested.

Cytotoxicity Assay

Neurons plated on poly-L-lysine coverslips were incubated at 37°C for 72 hr in

Neurobasal Complete with 25 µM Aβ25-35 in the absence (Control) and presence of 100

µM afobazole or 100 µM DTG. The coverslips were rinsed with PBS followed by 30 min

incubation at room temperature in PBS with 4 µL of a 2 mM ethidium homodimer-1

(EthD-1) dissolved in 1:4 DMSO:H2O solution. The coverslips were washed with PBS

and deionized water, dried, and mounted on a microscope slide with Vectashield Hardset

mounting media (Vector Labs, Burlingame, CA). EthD-1 loaded cells were illuminated

with light at 530 nm and visualized at 645 nm using a Zeiss Axioskop 2 equipped with a

20X objective. EthD-1-positive cells were identified and counted using Image-J in four

random fields per slide, and the average for the fields used as the value for the slide. For

each experiment, a minimum of three slides were used, and the results of at least three

experiments were averaged together for each condition tested.

50

Immunocytochemistry

Neurons plated on poly-L-lysine coverslips were incubated at 37°C for 24 hr in

Neurobasal Complete with 25 µM Aβ25-35 in the absence and presence of 100 µM afobazole. The coverslips were rinsed with phosphate-buffered saline (PBS) followed by an ethanol step-wise fixation. The cells were permeabilized with 0.1% Triton X in PBS for

15 min, then rehydrated with PBS and washed with 0.5% bovine serum albumin (BSA) in

PBS. Blocking was achieved with 45 min incubation in 2% BSA, followed by multiple washes with 0.5% BSA. Primary antibodies were diluted in PBS with 0.5% BSA and applied onto cells at 4°C for 24 hr. Primary antibody dilutions were as follows: Bax 1:20,

Caspase-3 1:25 and Bcl-2 1:100. The cells were then washed multiply times with 0.5%

BSA in PBS and then incubated in secondary antibodies for 60 min at room temperature.

Secondary antibodies were Alexa Fluor 488 conjugated anti-mouse or anti-rabbit, as appropriate, both diluted at a ratio of 1:300 in PBS with 0.5% BSA. Following incubation in secondary antibodies, cells were washed with 0.5% BSA in PBS and then with PBS alone. Coverslips were rinsed with deionized water, inverted and sealed onto a slide with

VectaShield containing DAPI. DAPI and the Alexa Fluor 488 conjugated secondary antibodies were illuminated at 359 and 485 nm and visualized at 461 and 530 nm, respectively, using a Zeiss Axioskop 2 outfitted with a 40X objective. Images of DAPI and Alexa Fluor 488 positive cells were counted and merged to demonstrate co- localization using Image-J software. Four random fields per slide were used to determine an average value for the slide. For each experiment, a minimum of three slides were used, and the results of at least three experiments were averaged together.

51

Compounds, Reagents and Antibodies

The following compounds and reagents were used in this investigation: Aβ25-35

(American Peptides, Sunnyvale, CA); DTG and BD-1047 (Tocris Biosciences, Ellisville,

MO); (Sigma-Aldrich, St. Louis, MO); fura-2 acetoxymethyl ester, DHE, DAF-

FM, Live/Dead Viability/Cytotoxicity Kit (Life Technologies). The following antibodies were used: Alexa Fluor 488 anti-mouse (A-11001) and anti-rabbit (A-11008) (Life

Technologies); anti-Bax (ab5714), anti-activated caspase-3 (ab32351) and anti-Bcl-2

(ab32370) (abcam, Cambridge, MA). Afobazole was generously provided by IBC

Generium (Moscow, Russian Federation). Vehicle for drugs and reagents used were either water (H2O), ethanol (EtOH) or dimethyl sulfoxide (DMSO), and appropriate

vehicle controls were carried out for each study.

Data Analysis

Data were analyzed using SigmaPlot 11 software (Systat Software, Inc., San

Jose, CA). Data points represent peak means ± standard error of the mean (S.E.M.).

One- and two-way analysis of variance were used for determining significant differences for multiple group comparisons, as appropriate, followed by post hoc analysis using the

Holm-Sidak test. Results were considered statistically significant if p < 0.05.

Results

2+ Experiments were first carried out to determine the effects of Aβ25-35 on [Ca ]i in

isolated cortical neurons from embryonic (E18) rats. Figure 3.1A shows representative

2+ traces of [Ca ]i measured in cortical neurons exposed to acute applications of 25 µM

Aβ25-35 (≤ 15 min). During this time period, the Aβ fragment did not significantly affect 52

2+ 2+ [Ca ]i in the neurons. To examine if extended incubation in Aβ25-35 alters [Ca ]i, neurons

were incubated from 1 hr to 24 hr in media contain 25 µM Aβ25-35. Figure 3.1B shows a

2+ bar graph of mean [Ca ]i recorded from different groups of neurons incubated in Aβ25-35

for the indicated time points. The Aβ fragment produced a statistically significant

2+ increase in [Ca ]i at all time points tested, with peak increases occurring 12 hr after

Aβ25-35 application.

Figure 3.1

Aβ25-35 promotes time-dependent elevations in neuronal intracellular calcium. A, representative traces of intracellular calcium as a function of time recorded from five neurons during acute application of 25 µM Aβ25-35. Line above traces indicates duration 2+ of Aβ application. B, Bar graph of mean (±SEM) [Ca ]i measured in neurons exposed to media (Control) or media containing 25 µM Aβ25-35 for the indicated times. Control value is compiled from measurements made at all time points. Asterisk denotes significant difference from Control (p < 0.05); for all time points, n > 1500 neurons.

Given that activation of σ receptors can inhibit increases in intracellular Ca2+

under various pathological conditions, we hypothesized that activation of σ receptors

53

with the pan-selective σ receptor agonists, afobazole or DTG, could affect the elevations

2+ in [Ca ]i triggered by incubation of cortical neurons in Aβ25-35. Cortical neurons were

incubated in 25 µM Aβ25-35 for 24 hr in the absence and presence of 100 µM afobazole or

100 µM DTG (Figure 3.2A). Following the 24 hr incubation with Aβ25-35, there was a

2+ statistically significant increase in [Ca ]i consistent with results shown in Figure 3.1B.

Incubation of neurons in media containing afobazole alone did not significantly affect

2+ neuronal [Ca ]i. However, incubation in afobazole blocked the Aβ25-35-evoked increases

2+ in [Ca ]i by 58 ± 12% (Figure 3.2A). In contrast, application of DTG (100 µM), failed to

2+ inhibit the Aβ25-35-evoked increase in [Ca ]i, and instead, potentiated the response by 67

± 21%.

The discrepancy between the results obtained for afobazole and DTG leave doubt

2+ as to the role of σ receptors in the regulation of Aβ25-35-induced increases in [Ca ]i by

afobazole. Therefore, to test if σ receptors are involved in these effects, selective

inhibitors of σ-1 (BD-1047) and σ-2 (rimcazole) receptors were used (Matsumoto et al.,

1995, Gilmore et al., 2004). Cortical neurons were incubated in 25 µM Aβ25-35 for 12 hr in

the absence and presence of 100 µM afobazole with and without BD-1047 or rimcazole

2+ (Figure 3.2B). Application of afobazole reduced elevations in [Ca ]i produced by Aβ25-35

by 77 ± 3%, which was statistically significant. While 10 µM BD-1047 alone lessened the

2+ effects of Aβ25-35 on [Ca ]i, the σ-1 receptor antagonist prevented any further reduction

by afobazole such that the block produced by the combination of afobazole + BD-1047

was almost identical to that produced by BD-1047 alone and 34 ± 4% less than that produced by afobazole alone. Unlike BD-1047, rimcazole (300 nM) alone did not

54

Figure 3.2

2+ Afobazole inhibits increases in [Ca ]i evoked by Aβ25-35. A, Bar graph of mean (±SEM) 2+ [Ca ]i measured in neurons incubated for 24 hr in media in the absence (Control) or presence of 25 µM Aβ25-35 (Aβ) with or without 100 µM Afobazole (Afob) or 100 µM DTG (DTG). Asterisks denote significant difference between Control and Aβ within the Media, Afob and DTG groups (p < 0.001 for Media and DTG groups, p < 0.05 for Afob group). Pound symbols denote significant difference from the Media group within the Aβ group (p < 0.001 for both). Dagger indicates significant difference between Afob and DTG groups within Aβ (p < 0.01). For all groups, n ≥ 125 neurons. B, Bar graph of mean (±SEM) Aβ- 2+ 2+ induced increase in [Ca ]i (∆[Ca ]i) measured in neurons incubated for 12 hr in 25 µM Aβ25-35 alone (Aβ) or in 25 µM Aβ25-35 + 100 µM afobazole (Aβ + Afob). Aβ or Aβ + Afob were applied in media alone (Media), in media with 10 µM BD-1047 (BD), or in media with 300 nM rimcazole (Rim 0.3) or 10 µM rimcazole (Rim 10). Asterisks denote significant difference between Aβ and Aβ + Afob within Media and rimcazole groups, respectively (for all p < 0.001). Pound symbols denote significant difference between BD- 55

1047 (BD) and all other groups within Aβ and between BD-1047 and the Media and 300 nM rimcazole (Rim 0.3) within Aβ + Afob, respectively (p < 0.01 for all). Dagger indicates significant difference between Rim 10 and Control groups within Aβ (p < 0.001). C, Bar 2+ graph of relative mean (±SEM) elevations in [Ca ]i evoked by Aβ25-35 in the presence of afobazole and in the absence (Control) or presence of 10 µM BD 1047 (BD), 300 nM rimcazole (Rim 0.3) and 10 µM rimcazole (Rim 10). Data were normalized to mean 2+ ∆[Ca ]i evoked by Aβ in the appropriate control (i.e. media or the corresponding σ receptor antagonist). Asterisks, pound symbols and dagger denote significant difference from Control, BD and Rim 0.3, respectively (for all p < 0.05). For all groups in B and C, n ≥ 168 neurons.

2+ significantly inhibit Aβ25-35-evoked increases in [Ca ]i. When afobazole was co-applied

with the low concentration of rimcazole (300 nM), which primarily affects σ-2 receptors,

2+ afobazole reduced Aβ25-35-evoked [Ca ]i elevations by 83 ± 5% (Figure 3.2B). This

decrease was similar to that seen with afobazole alone. Like BD-1047, a higher

concentration of rimcazole (10 µM), predicted to interact with σ-1 receptors (Husbands et

2+ al., 1999), reduced Aβ25-35-induced increases in [Ca ]i, (Figure 3.2B). However, Aβ25-35-

2+ evoked increases in [Ca ]i observed in the cells incubated in 10 µM rimcazole +

afobazole were not significantly different from those produced by the Aβ fragment in the

presence of afobazole (p = 0.179). In order to account for these direct effects of the

2+ 2+ antagonists on Aβ25-35 evoked elevations in [Ca ]i, we normalized the increases in [Ca ]i

observed when afobazole was added along with the σ receptor antagonist to those

observed when Aβ was added with the antagonist alone. The mean normalized

responses observed for the different conditions are shown in Figure 3.2C. The effects of

afobazole were blocked by both BD 1047 and the higher concentration of rimcazole (10

µM, Rim 10), but not by the lower concentrations of rimcazole (300 nM, Rim 0.3) (Figure

56

3.2C). Therefore, inhibition of σ-1, but not σ-2 receptors lessened the effects of afobazole.

Figure 3.3

Afobazole does not inhibit Aβ25-35 evoked ROS production but does inhibit nitric oxide production in neurons. A, Bar graph of mean (±SEM) DHE fluorescence intensity (ROS levels) measured in neurons incubated for 24 hr in the absence (Control) or presence of 25 µM Aβ25-35 (Aβ) without (Media) or with 100 µM Afobazole (Afob). Asterisks denote significant difference between Control and Aβ within Media and Afob groups (p < 0.05 for both). Pound symbols denote significant difference between Media and Afob groups within Control (p < 0.001) and Aβ (p < 0.05) groups. B, Bar graph of mean (±SEM) DAF- FM fluorescence intensity (NO levels) measured in neurons incubated for 24 hr in media alone (Control) or media containing 25 µM Aβ25-35 (Aβ), 100 µM Afobazole (Afob) or 25 µM Aβ25-35 and 100 µM Afobazole (Aβ + Afob). Asterisks denote significant difference between Control and Aβ within Media and Afob (p < 0.001 for both). Pound symbol denotes significant difference between Media and Afob groups within Aβ (p < 0.001). For all groups, (n > 108 neurons).

2+ Cortical neuron [Ca ]i overload has been shown to enhance production of

reactive oxygen species (ROS) and nitric oxide, with the latter molecule being converted

to reactive nitrogen species (RNS) in the presence of ROS. Activation of σ receptors has 57

been associated with changes in both ROS and nitric oxide levels. Thus, we evaluated if

2+ afobazole preservation of [Ca ]i homeostasis consequently affects ROS and/or nitric

oxide levels following exposure to Aβ25-35. Figure 3.3A shows a bar graph of the average

DHE fluorescence intensities measured in neurons in response to 24 hr incubation in

Aβ25-35 in the absence and presence of 100 µM afobazole. Greater DHE fluorescence

was observed in neurons exposed to Aβ25-35 relative to media alone, indicating an Aβ-

induced increase in ROS production (Figure 3.3A). Incubation of the neurons in

afobazole alone also produced a small but significant increase in neuronal ROS, and a

further increase in ROS was noted when Aβ25-35 was co-applied with afobazole (Figure

3.3A). NO levels were also measured in neurons exposed to Aβ25-35 for 24 hr. Incubation

of neurons in Aβ25-35 significantly enhanced neuronal NO levels. In contrast to the results obtained for ROS, afobazole alone had no effects on basal neuronal NO content.

However, the σ agonist significantly decreased Aβ25-35-evoked elevations in NO by 53 ±

5% (Figure 3.3B).

2+ Afobazole suppression of Aβ25-35-evoked [Ca ]i dysregulation and NO production

suggests that this compound may be neuroprotective in this injury model. Thus,

experiments were conducted to ascertain if afobazole can reduce Aβ25-35 cytotoxicity and enhance cell survival following exposure of cortical neurons to this Aβ fragment. For comparison, a second σ receptor agonist, DTG, which has been shown to augment

2+ survival in other neuronal injury models but does not block Aβ25-35-elicited [Ca ]i

increases was included. Neurons were incubated for 72 hr in 25 µM Aβ25-35 in the

absence and presence of 100 µM afobazole or 100 µM DTG and cell survival determined

58

Figure 3.4

Afobazole, but not DTG, reduces neuronal death produced by application of Aβ25-35. A, Photomicrographs showing EthD-1 labeling (red), a marker of cell death, observed following 72 hr incubation of neurons in media alone (i), or media containing 25 µM Aβ25- 35 (ii), 100 µM afobazole (iii), 100 µM DTG (v), 25 µM Aβ + 100 µM afobazole (iv), or 25 µM Aβ + 100 µM DTG (vi). Fields of view shown are representative regions within larger images used to calculate number of neurons labeled with EthD-1. B, Bar graph of relative neuronal death observed for the same conditions as in (A). Data were normalized to the average number of EthD-1-positive cells observed with media alone (i.e. no Aβ or σ ligand). Asterisks denote significant difference between Control and Aβ within Media and DTG groups, respectively (p < 0.001 for both). Pound symbol indicate significant difference between Afob and the other groups within Aβ (p < 0.001) and dagger indicates significant difference between DTG and the other groups within Control (p < 0.05). For all groups, n ≥ 12. Scale bar in (i) is 20 µm.

59

using EthD-1 to identify apoptotic cells. Figure 3.4A shows representative

photomicrographs of neurons labeled with EthD-1. While EthD-1 staining was noted in all groups, a higher number of cells were observed when Aβ25-35 was applied alone or in the

presence of DTG (Figure 3.4A ii & vi), but not when co-applied with afobazole (Figure

3.4A iv). In identical experiments, incubation in Aβ25-35 resulted in a 130 ± 6% increase in

the percentage of cells exhibiting positive labeling with EthD-1, relative to control (media alone, Figure 3.4B). When afobazole (100 µM) was applied alone, there was no change in the percentage of dead cells noted. However, application of afobazole along with the

Aβ25-35 resulted in an 88 ± 4% reduction in the percentage of EthD-1 positive cells

relative to the Aβ fragment alone group, which was statistically significant (Figure 3.4B).

In contrast, DTG alone increased the percentage of EthD-1 positive cells compared to

control (Figure 3.4B). Moreover, DTG failed to reduce the percentage of cells exhibiting

EthD-1 staining when co-incubated with Aβ25-35 such that the number of apoptotic cells

seen with Aβ25-35 alone and Aβ25-35 + DTG were similar (Figure 3.4B).

Experiments were next focused on identifying underlying molecular mechanisms

contributing to the enhanced survival of cortical neurons following afobazole application.

Previous studies have shown that σ-1 receptor activation by the selective agonist, (+)-

SKF10047, can protect RGC-5 cells from glutamate-induced apoptosis by reducing expression of the pro-apoptotic gene product Bax (Tchedre and Yorio, 2008). To determine if a similar pathway is involved in afobazole-mediated neuroprotection during

Aβ25-35 exposure, the cellular expression of this protein was determined. Representative

photomicrographs of cells co-labeled with DAPI and an anti-Bax antibody are shown in

Figure 3.5A. While Bax is detected in control neurons (Figure 3.5A i), there is a marked 60

Figure 3.5

Afobazole prevents upregulation of the pro-apoptotic gene product, Bax, caused by application of Aβ25-35 in neurons. A, Photomicrographs of merged images of cultured neurons exposed for 24 hr to media alone (Control) (i), or media containing 25 µM Aβ25- 35 (Aβ) (ii), 100 µM afobazole (Afob) (iii) or 25 µM Aβ25-35 + 100 µM afobazole (Aβ + Afob) (iv). Neurons were double labeled with DAPI (blue) and anti-Bax antibody (green). Fields of view shown are representative regions within larger images used to calculate Bax expression. B, Bar graph of mean percent of Bax-positive neurons observed in multiple experiments in which cells were exposed to the same conditions described in (A). Data are expressed as mean percent (±SEM) of the total number of neurons staining positive for DAPI. Asterisks denote significant difference between Control and Aβ within Media and Afob groups, respectively (p < 0.001). Pound symbols indicate significant difference between Afob and Media groups within Control (p < 0.05) and Aβ (p < 0.001). Scale bar in (i) is 10 µm and n = 9 for all groups in (B). increase following incubation in Aβ25-35 (Figure 3.5A ii). Bax is also expressed in low

levels in neurons exposed to afobazole alone (Figure 3.5A iii), and there is only a modest

increase when neurons are incubated in both 100 µM afobazole and the Aβ fragment 61

(Figure 3.5A iv). Analysis of images collected from multiple experiments shows that Bax

is detected in less than half of the control cells, but following exposure to Aβ25-35 there is

a 100 ± 5% increase in the number of cells testing positive for this pro-apoptotic gene

(Figure 5B). Application of afobazole reduces Bax expression below that observed in

control cells by 13 ± 5%, and the increase in expression occurring after Aβ25-35

incubation is reduced to 58 ± 5% (Figure 3.5B). The net result is a 39 ± 5% reduction in

the total number of cells expressing Bax in response to Aβ25-35 when afobazole was

applied.

Given that σ receptors have also been linked to decreased caspase-3 expression

(Tchedre and Yorio, 2008), immunocytochemistry was also employed to examine how

afobazole affects the levels of the active form of this death protease after neuronal

exposure to Aβ25-35. Representative photomicrographs of cells labeled with DAPI and

anti-activated caspase-3 antibody show an increase in the number of caspase-3 positive neurons 24 hr after incubation in Aβ25-35 relative to control (Figure 3.6A i & ii). Neuronal

expression of the active form of caspase-3 was not affected by incubation of the cells in

100 µM afobazole alone, but co-application of afobazole with Aβ25-35 significantly lowered

activated caspase-3 expression when compared to cells treated only with Aβ25-35 (Figure

3.6A iii & iv). Figure 3.6B shows a bar graph of mean (±SEM) percentages of active

caspase-3 positive neurons detected from multiple experiments using the same

conditions as in Figure 3.6A. There is a statistically significant upregulation of activated

caspase-3 expression following Aβ25-35 application, which results in a 104 ± 7% increase

in the percentage of neurons testing positive for the death protease. However, treatment

with afobazole reduces the increased cellular expression of activated caspase-3 62

Figure 3.6

Afobazole reduces expression of activated caspase-3 in response to application of Aβ25- 35 in neurons. A, Photomicrographs of merged images of cultured neurons exposed for 24 hr to media alone (Control) (i), or media containing 25 µM Aβ25-35 (Aβ) (ii), 100 µM afobazole (Afob) (iii) or 25 µM Aβ25-35 + 100 µM afobazole (Aβ + Afob) (iv). Neurons were double labeled with DAPI (blue) and anti-active caspase-3 antibody (green). Fields of view of each photomicrograph are representative regions within larger image used to calculate caspase-3 expression. B, Bar graph of mean percent (±SEM) of neurons testing positive for the active form of caspase-3 in experiments in which the cells were exposed to the same conditions as (A). Asterisk denotes significant difference between Control and Aβ within Media (p < 0.001). Pound symbol indicates significant difference between Media and Afob within Aβ (p < 0.001). Scale bar in (i) is 10 µm and n = 9 for all groups in (B).

produced by Aβ25-35 by 89 ± 7% (Figure 3.6B). Neuronal expression of active caspase-3

observed in cells treated with afobazole alone and afobazole with Aβ25-35 were not

statistically different (p = 0.20).

63

Activation of σ receptors has been shown to provide neuroprotection against

toxicity produced by the HIV-1 protein gp120 via upregulation of the anti-apoptotic gene

Bcl-2 (Zhang et al., 2012). Therefore, we examined the possible involvement of Bcl-2 in

the protection provided by afobazole against Aβ25-35-induced cytotoxicity. A significant

number of neurons were found to express Bcl-2 in our culture model under all conditions tested at both 24 and 48 hrs (Figure 3.7A i-vii). Quantification of the results indicates that

69 ± 6% of control neurons expressed Bcl-2, and that incubation in 25 µM Aβ25-35 for 24

hr increased this number to 90 ± 2% (Figure 3.7B). The addition of afobazole alone also

produced a 24 ± 2% increase in the percentage of Bcl-2 expressing cells, which was

statistically significant when compared to the control (Figure 3.7B). However, after a 24

hr incubation, no further increase in Bcl-2 was noted when afobazole was applied with

Aβ25-35 compared to incubation in Aβ25-35 alone. In contrast, increasing the incubation

time of the cells in Aβ25-35 to 48 hr resulted in reduction in the number of cells showing

positive labeling for Bcl-2 expression, with Bcl-2 being observed in 71 ± 1% of cells

incubated in media alone and 46 ± 2% of the cells incubated in media containing the Aβ

fragment (Figure 3.7B). Extending the application time of afobazole to 48 hr continued to

produce a similar increase in Bcl-2 expression as seen with the 24 hour incubation, with

85 ± 1% of the afobazole treated neurons showing Bcl-2 expression. However, the

addition of afobazole to Aβ25-35 prevented the Aβ-induced suppression of Bcl-2 seen at

48 hr, withBcl-2 being observed in 81 ± 1% of the neurons exposed to Aβ25-35 in the

presence of this drug (Figure 3.7B). In fact, in cultures exposed to afobazole + Aβ there

64

was a 14 ± 2% increase in the number of neurons testing positive for Bcl-2 expression compared to control (no Aβ or afobazole).

Figure 3.7

Afobazole prevents Bcl-2 down-regulation following 48 hr exposure of neurons to Aβ25-35. A, Photomicrographs of merged images of cultured neurons exposed for 24 or 48 hr to 100 µM afobazole (Afob, i and iv), 25 µM Aβ25-35 (Aβ, ii and v), or 100 µM afobazole + 25 µM Aβ25-35 (Afob + Aβ, iii and vi). For comparison, a photomicrograph of merged images of neurons exposed to media alone for 48 hr is shown (Control, vii). Neurons were double labeled with DAPI (blue) and anti-Bcl-2 antibody (green). Fields of view of each photomicrograph are representative regions within larger images used to calculate Bcl-2 expression. B, Bar graph of mean percent (±SEM) of Bcl-2-positive neurons exposed to the conditions described in (A). Asterisks indicate significant difference between Control and Aβ within the 24 hr and 48 hr groups, respectively. Pound symbols indicate significant difference between Control and Afob within the 24 hr and 48 hr groups, respectively, and between Aβ and Aβ + Afob within the 48 hr group. For 24 hr group n = 9 for all, for 48 hr group n = 12 for all, p ≤ 0.001 for all. Scale bar in (vii) is 10 µm.

65

Discussion

The major finding reported in this study is that afobazole, acting via σ-1 receptors, provides neuroprotection against Aβ25-35-induced toxicity in cortical neurons. Activation of

σ-1 receptors by afobazole appears to impinge on multiple molecular mechanisms

associated with the demise of neurons. First, afobazole, acting via σ-1 receptors,

2+ decreases neuronal [Ca ]i dyshomeostasis resulting from prolonged exposure (1 to 24

hr) of the cells to Aβ25-35. Second, afobazole mitigates Aβ25-35-evoked elevations in nitric

oxide production in cortical neurons that coincides with increases in ROS production,

thus nitrosative stress is likely lessened. Finally, afobazole blunts the pro-apoptotic

signaling induced by Aβ25-35 by inhibiting the upregulation of the pro-apoptotic gene

product Bax and the death protease, caspase-3, while preventing long-term

downregulation of the anti-apoptotic protein, Bcl-2.

2+ In our culture system, Aβ25-35 was found to produce increases in [Ca ]i after

cells were incubated in the Aβ fragment for ≥ 1 hr, with short applications ≤ 15 min failing

2+ 2+ to alter [Ca ]i. Maximal increases in [Ca ]i were observed after 12 hr incubation of the

2+ neurons in Aβ25-35 and [Ca ]i remained significantly elevated even after 24 hr incubation

2+ in the Aβ fragment. The observed 2-fold increase in [Ca ]i caused by Aβ25-35 is similar to

that previously reported for rat cortical neurons incubated for 24 hr in Aβ25-35 (Ferreiro et

al., 2004). Incubation of neurons in 100 µM afobazole reduced Aβ25-35-evoked increases

2+ 2+ in [Ca ]i by ~60%. This decrease in [Ca ]i is similar to that observed when afobazole

2+ was used to inhibit [Ca ]i increases produced in cortical neurons by acidosis (IC50 = 164

2+ µM) (Cuevas et al., 2011a). Reducing Aβ25-35 perturbation of [Ca ]i is likely to promote

66

2+ neuronal survival, since [Ca ]i destabilization has been linked to cell death in AD

2+ (Mattson and Chan, 2003b). Moreover, the Aβ25-35-induced changes in [Ca ]i precede pro-apoptotic signals in the model used here (e.g. decrease in Bcl-2 levels occurring 48 hrs after Aβ25-35 application).

2+ The inhibition of Aβ25-35-elicited increases in [Ca ]i by afobazole is due to

activation of σ-1, but not σ-2 receptors. Incubation in the σ-1 receptor antagonist, BD-

1047 (Matsumoto et al., 2004), reduced the effects of afobazole by ~40%. The concentration and degree of block reported here is consistent with previous studies showing BD-1047 inhibition of σ-1 receptor-mediated effects in cortical neurons and retinal ganglion cells (Katnik et al., 2006, Cuevas et al., 2011a, Mueller et al., 2013). The lack of involvement of σ-2 receptors is confirmed by results obtained using the σ-2 receptor antagonist, rimcazole (Gilmore et al., 2004). Rimcazole binds to σ-2 receptors with nanomolar affinity, but to σ-1 receptor with low micro molar affinity (Ferris et al.,

1986, Husbands et al., 1999, Rybczynska et al., 2008). Given that 300 nM rimcazole

2+ failed to inhibit the actions of afobazole on Aβ25-35-evoked increases in [Ca ]i, but that

the higher concentration of the drug reduced these effects, it is unlikely that afobazole

2+ activates σ-2 receptors to suppress Aβ25-35-evoked increases in [Ca ]i. Surprisingly, a

second pan-selective σ receptor agonist, DTG, failed to mimic the effects of afobazole on

2+ Aβ25-35-evoked [Ca ]i perturbation. DTG and afobazole both block acidosis- and

ischemia-induced Ca2+ overload in neurons to a similar extent (Cuevas et al., 2011a). It

2+ 2+ was shown that ischemia-induced [Ca ]i increases were due primarily to Ca influxes

through voltage-gated Ca2+ channels and NMDA receptors which were regulated by σ-1

67

receptor activation (Katnik et al., 2006). However, the difference between afobazole and

2+ 2+ DTG effects on Aβ modulation of [Ca ]i, suggest these long-term [Ca ]i increases

2+ involve distinct mechanisms from those producing ischemia-induced [Ca ]i overload.

2+ Additionally, the lack of a DTG effect on Aβ-evoked [Ca ]i overload suggests that not all

σ-1 agonists may promote neuronal survival in this model. This hypothesis is further

supported by our observation that DTG fails to decrease neuronal apoptosis following 24

hr incubation Aβ25-35. The difference in responses between afobazole and DTG may be

due to off-target effects of DTG counteracting the benefits of this pan-selective sigma

agonist. Such a possibility needs to be explored further.

2+ It has been suggested that one of the downstream consequences of [Ca ]i

dysregulation in neurons caused by Aβ25-35 is the production of ROS (Ekinci et al., 2000,

Abramov et al., 2004). Afobazole has been shown to reduce ROS accumulation in rat

brains after focal ischemia (Silkina et al., 2006). However, data presented here show that

afobazole does not block the ROS increases caused by the Aβ fragment. It has been

suggested that Aβ, and in particular Aβ25-35, can increase oxidative stress via

2+ mechanisms independent of [Ca ]i dysregulation (Varadarajan et al., 2001). Given that

2+ afobazole inhibits [Ca ]i dysregulation produced by Aβ25-35 in neurons but not the

2+ increase in ROS, it is unlikely that afobazole is affecting this [Ca ]i-independent ROS

production. It is important to note that while afobazole may not decrease ROS

production, it has been shown that σ-1 receptors decrease H2O2 evoked apoptosis

(Meunier and Hayashi, 2010). Thus, afobazole may in part decrease Aβ25-35-induced cell

death by acting downstream of the observed ROS increase.

68

Aβ25-35 has been shown to increase nitric oxide synthase (NOS) activity and

produce neuronal injury via nitrosative stress (Parks et al., 2001, Cho et al., 2009). Aβ25-

35-evoked increases in NO were significantly decreased following afobazole application.

Inhibition of inducible NOS (iNOS) has been proposed as a mechanism by which activation of σ-1 receptors is neuroprotective after stroke (Vagnerova et al., 2006).

Similarly, functional downregulation of neuronal NOS (nNOS) has been shown to

contribute to σ receptor neuroprotection following ischemia in striatal neurons (Yang et

al., 2010). Since Aβ25-35 appears to increase both iNOS and nNOS in neurons (Limon et

al., 2009), activation of σ receptors by afobazole is predicted to reduce NO production by

both of these enzymes, and will ultimately contribute to protection from Aβ25-35 induced

injury.

One of the pro-apoptotic proteins upregulated in neurons both by the pathological sequela induced by Aβ25-35 application and in patients with Alzheimer’s disease is Bax

(Paradis et al., 1996, Yao et al., 2005). Consistent with previous findings, the current

study shows that the number of Bax positive neurons is significantly increased following

24 hr incubation in Aβ25-35. However, when afobazole is applied in combination with Aβ25-

35, Bax upregulation is diminished significantly. It has also been shown that the σ-1

agonist, PRE-084, can reduce apoptosis in cortical neurons caused by Aβ25-35 via

reduction in Bax (Marrazzo et al., 2005). However, glutamatergic neurotransmission was

blocked in those experiments (Marrazzo et al., 2005). Glutamatergic transmission is a

factor in Aβ25-35-induced cell death, and elevated glutamate levels have been shown to

induce Bax upregulation in neurons (Schelman et al., 2004, Revett et al., 2013). Thus,

69

our data suggest that σ receptor activation by afobazole can reduce Bax expression and concomitant cell death in the presence of Aβ25-35 even when glutamatergic transmission

is not blocked.

In addition to decreasing Bax levels, afobazole effectively reduced activated

caspase-3 expression in response to Aβ25-35. The active form of caspase-3 has been

implicated in the loss of hippocampal neurons in Alzheimer’s disease (Selznick et al.,

1999). In addition to serving as a late event involved in neuronal death during

Alzheimer’s disease, caspase activation may be an early event that promotes the

pathology of AD. Several caspases, including caspase-3, have been shown to cause pathological tau filament assembly in neurons, which underlies AD etiology (Gamblin et al., 2003). Selective inhibition of caspase-3 has been shown to protect neurons from

Aβ25-35-induced apoptosis (Allen et al., 2001). Sigma receptor activation has been shown to decrease caspase-3 and to protect cortical neurons and RGC-5 cells against excitotoxicity (Tchedre and Yorio, 2008, Griesmaier et al., 2012). Such a mechanism is likely to contribute to afobazole neuroprotection from Aβ25-35 toxicity and may in part

explain the reduced neuronal death observed here.

The neuroprotective properties of afobazole following exposure to Aβ25-35 also

involves long-term upregulation of Bcl-2. Our data indicate that there is a biphasic

change in Bcl-2 levels in neurons following application of Aβ25-35 with an initial increase

at 24 hr followed by a suppression of Bcl-2 expression at 48 hr. A similar transient expression pattern was previously reported in rat hippocampal neurons exposed to Aβ25-

35 (Kim et al., 1998). It was proposed that this initial increase in Bcl-2 expression is a

non-sustainable cellular response to the apoptotic pathways induced by Aβ (Kim et al., 70

1998). Our observations indicate that when afobazole is applied along with Aβ25-35, Bcl-2

levels do not drop below control at the 48 hr time point. Previous studies have shown

that Bcl-2 is a target for σ receptor mediated neuroprotection. The HIV-1 protein gp120 was shown to downregulate Bcl-2 in cortical neurons, but activation of σ receptors with the σ ligand, PPBP, increased Bcl-2 expression (Zhang et al., 2012). PPBP was shown to have a similar effect in a glutamate excitotoxicity model (Yang et al., 2007), suggesting that Bcl-2 upregulation may occur after σ receptor stimulation under various pathophysiological conditions.

In conclusion, our experiments show that afobazole can decrease Aβ25-35-evoked

neuronal apoptosis by activating σ-1 receptors. The activation of σ-1 receptors prevents

2+ Aβ25-35-induced [Ca ]i overload and is associated with a decrease in NO levels. The

molecular mechanisms mediating neuroprotection by afobazole include downregulation

of both Bax and activated caspase-3, and long-term upregulation of Bcl-2. Given the importance of direct Aβ neurotoxicity in the etiology of Alzheimer’s disease, the findings reported here suggest that σ-1 receptors are a putative target for AD therapy and that the σ receptor agonist, afobazole, in particular, shows significant potential.

71

CHAPTER 4

Stimulation of Sigma Receptors with Afobazole Blocks Activation of Microglia and Reduces Toxicity Caused by Amyloid-β25-35

Introduction

Aβ has been shown to initiate a sequence of events in microglia, including a pro- inflammatory cascade and enhanced hemichannel formation, which ultimately produces neuronal death (Orellana et al., 2011). In contrast, data also suggest that microglia may be beneficial in AD by removing Aβ plaques (Wilcock et al., 2004, Herber et al., 2007). It has been proposed that microglial senescence, and not activation, is a contributing factor to late-onset Alzheimer's disease (Streit et al., 2009). Given the importance of microglial cells in AD pathology, there is great interest in identifying molecular targets for modulating the responses of these cells to Aβ, and for regulating their contribution to AD pathology in general.

One possible target that has been explored for therapeutics in other pathological states of the central nervous system but is not well-characterized in AD is sigma receptors. Sigma (σ) receptors are found in numerous cell types distributed throughout the mammalian body and modulate multiple signaling and regulatory pathways. Two subtypes of σ receptors have been pharmacologically identified, σ-1 and σ-2.

Reprinted with permission of the American Society for Pharmacology and Experimental Therapeutics. Copyright © 2013 by the American Society for Pharmacology and Experimental Therapeutics

72

To date only the σ-1 receptor has been cloned (Walker et al., 1990, Quirion et al.,

1992, Seth et al., 1998). Sigma-1 receptors are intracellular chaperones that form a

complex within the endoplasmic reticulum (ER) and the mitochondrion-associated ER

membrane (MAM) (Kourrich et al., 2012). Activation of σ-1 receptors promotes migration

of these receptors away from the MAM to the sub-plasmalemmal ER, where the receptors interact with and regulate numerous membrane channels (Zhang and Cuevas,

2002, 2005, Herrera et al., 2008, Zhang et al., 2009, Su et al., 2010, Cuevas et al.,

2011b). Less is known about σ-2 receptors, but these proteins seem to be involved in regulation of inflammation, cell survival and calcium homeostasis (Vilner and Bowen,

2000, Crawford and Bowen, 2002, Zhang and Cuevas, 2002, Iniguez et al., 2013). Our laboratory has shown that activation of σ receptors in microglia decreases ATP-induced membrane ruffling and cell migration and lipopolysaccharide-induced cytokine production

(Hall et al., 2009a). Activation of σ receptors also protects microglia from ischemia- induced cell death (Cuevas et al., 2011b). These observations led our laboratory to propose that the neuroprotection provided by σ receptor activation following stroke

involves both a decrease in the inflammatory response mediated by microglia and

enhanced survival of quiescent microglia (Cuevas et al., 2011b).

While the effects of σ receptor activation on microglial responses to ischemia

have been studied, there is little information on how σ receptors may affect microglial

responses to Aβ. In the absence of glutamatergic transmission, activation of σ-1

receptors has been shown to decrease neuronal apoptosis caused by Aβ25-35 (Marrazzo et al., 2005), but it is unclear how σ-1 activation will affect survival of microglia exposed

73

to Aβ. It has been reported that there is a reduction in the number of sigma ligand

binding sites and σ-1 receptors in the brains of AD patients (Jansen et al., 1993, Mishina

et al., 2008). If these receptors prevent microglial activation and/or senescence, a loss of

these receptors may contribute to AD progression. Thus, it is of significant interested to

determine how σ receptor activation influences microglial responses to Aβ.

Experiments were carried out to determine if afobazole, an agonist at both σ-1

and σ-2 receptors (Cuevas et al., 2011b), can reduce activation and toxicity of microglia

by Aβ25-35. Afobazole decreased membrane ruffling and migration evoked by Aβ25-35. The

effects of afobazole were dependent on activation of both σ-1 and σ-2 receptor and were blocked by inhibition of these receptors with BD-1047 and rimcazole. Furthermore, afobazole inhibited increases in the levels of the pro-apoptotic protein Bax and the death protease caspase-3 induced by Aβ25-35, resulting in an increase in microglia survival.

Finally, afobazole was shown to prevent disruption of ATP signaling in microglia

incubated in Aβ25-35, indicating that afobazole preserves microglial function following Aβ

exposure.

Materials and Methods

Primary Cultures of Microglia

Primary cultures of microglia were prepared from Sprague-Dawley mixed sex rat

pups (post-natal day 2-3) as previously described by our laboratory (Hall et al., 2009a,

Cuevas et al., 2011b). The mixed glial cultures were incubated for 7-12 days at 37 o C

prior to experiments being carried out. Microglia were mechanically separated from the

74

cultures by brief shaking. Isolated microglia were resuspended in DMEM PLUS

containing: 500 mL Dulbecco’s Modified Eagle Medium with 4.5 g/L glucose, L-glutamine

and sodium pyruvate (DMEM) (Corning Cellgro, Manassas, VA), 40 mL horse serum

(Corning Cellgro), 12.5 mL heat-inactivated fetal bovine serum (Thermo Scientific

Hyclone, Logan, UT ), and 5 mL 10x antibiotic/antimycotic (Corning Cellgro). These cells

were used immediately for migration and cytotoxicity assays or plated on glass

coverslips for one day for imaging experiments.

Membrane Morphology and Migration Assay

Morphological changes to microglia induced by ATP or Aβ25-35 were assessed by plating the cells on poly-L-lysine coated coverslips, serum starving the cells for 4 h in

DMEM, and exposing the cells to100 µM ATP or 25 µM Aβ25-35 for 10 min at 37º C.

When afobazole or DTG were used, microglia were incubated in the σ ligands (in

DMEM) for 10 min prior to ATP or Aβ25-35 exposure. Membrane ruffling was visualized by

labeling the cells with the filamentous actin probe, phalloidin (AlexaFluor 488 conjugated,

Life Technologies, Grand Island, NY). Quantification of membrane ruffling was carried

out as previously described (Hall et al., 2009a). Briefly, cell ruffling was scored as: “0” –

no ruffling and multiple filopodia; “1” – ruffling and filopodia; “2” – fully ruffled with no

filopodia.

Migration assays were carried out using a 48-well chemotaxis chamber (neuro

Probe, Inc., Gaithersburg, MD) as previously described (Cuevas et al., 2011b). The

bottom wells of the chamber were filled with 25 µM Aβ25-35 in DMEM and the top wells

contained 1x106 freshly isolated microglia. These chambers were separated by a

75

fibronectin coated polycarbonate membrane with 8 µm pores at a density of 1 x 103 pores/mm2. Each well had an exposed filter area of 8 mm2. Test compounds (i.e. σ ligands) were added to both the top and bottom wells at identical concentrations. The microglia were permitted to migrate for 2 hr while incubated at 37oC. The membranes containing microglia were processed as previously described and mounted on slides using Vectashield Hardset containing 4'-6-diamidino-2-phenylindole (DAPI) (Vector Labs,

Burlingame Ca) (Cuevas et al., 2011b). DAPI positive cells were counted in 4 random fields of view per well and averaged to determine migration for that well. For each experiment, a minimum of 2-3 wells were used, and at least three migration experiments were carried out for each condition. The n values given represent the total number of fields of view recorded.

Calcium Imaging Measurements

2+ 2+ Intracellular Ca concentrations ([Ca ]i) as a function of time were measured using fluorescent imaging techniques and fura-2 as previously reported (Hall et al.,

2009a, Cuevas et al., 2011b). Microglia plated on coverslips were incubated for 1 hour at room temperature in DMEM-PLUS containing 25 µM Aβ and 3 µM acetoxymethyl ester fura-2 and 0.3 % dimethyl sulfoxide. Prior to the experiments, the coverslips were washed in Aβ and fura-2-free physiological saline solution (PSS) containing (in mM): 140

NaCl, 5.4 KCl, 1.3 CaCl2, 1.0 MgCl2, 20 glucose, and 25 HEPES (pH to 7.4 with NaOH).

2+ For studies on ATP-evoked [Ca ]i changes, ATP was applied in PSS using a rapid application system as described previously (Cuevas and Berg, 1998).

76

ROS and NO Imaging

Intracellular ROS and NO concentrations were measured in isolated microglia

using the fluorometric indicators di-hydroethidiumbromide (DHE) and diacetate (4-amino-

5-methylamino-2',7'-difluorofluorescein diacetate) (DAF-FM), respectively. Cells plated on poly-L-lysine coated coverslips were incubated at 37°C in DMEM-PLUS containing 10

µM DHE with 0.25 % DMSO for 30 min or 5 µM DAF-FM with 0.1 % DMSO for 1 hr. Prior to beginning the experiments, the coverslips were rinsed with PSS. DHE and DAF-FM labeled microglia were measured by digital microscopy (CoolSnap HQ2; Photometrics,

Surrey, BC, Canada), excited at 518 nm and 495 nm and emission captured at 605 nm and 515 nm, respectively. Excitation exposure times were auto-calibrated to the control

(absence of drugs or Aβ) using this function on the acquisition software (Nikon Elements;

Nikon, Melville, NY, USA) and used for all slides in that dish. Image analysis was performed using the Nikon Elements software. Images were acquired every second for ten seconds and the average of the acquisitions was used for the fluorescence intensities (A.U.). Statistical analysis was performed using SigmaPlot 11 (Systat

Software, Inc., San Jose, CA).

Cell Death Assay

Microglia plated on coverslips were incubated at 37°C for 72 hr in DMEM-PLUS

with 25 µM Aβ25-35 in the absence (Control) and presence of 30 µM afobazole. The

coverslips were rinsed with phosphate buffered saline (PBS) and then incubated for 30

min in 4 µM EthD-1 solution at room temperature. The coverslips were washed with PBS

and deionized water, air dried and mounted on a microscope slide with Vectashield

77

Hardset mounting media (Vector Labs, Burlingame, CA). EthD-1 positive cells were visualized at 400x using a Zeiss Axioskop 2 and counted using Image-J software in four random fields per slide. For each experiment, a minimum of three slides were used, and the results of at least three experiments were averaged together. Coverslips were visualized in bright field to confirm homogeneous cell density.

Immunocytochemistry

Microglia plated on poly-L-lysine coverslips were incubated at 37°C for 24 hr in

DMEM-PLUS with 25 µM Aβ25-35 in the absence (Control) and presence of 30 µM afobazole. The coverslips were rinsed three times, followed by a step-wise ethanol fixation, permeabilized with 0.1 % Triton X, rehydrated and blocked with 2 % BSA.

Primary antibodies were diluted in PBS with 0.5 % BSA and incubated at 4°C for 24 hr.

The following dilutions were used: anti-Bax 1:20, anti-activated Caspase-3 1:25 and anti-

Bcl-2 1:100. The secondary antibodies used were Alexa Fluor 488 conjugated anti- mouse or anti-rabbit, as appropriate, and diluted at a ratio of 1:300 in PBS with 0.5%

BSA. Coverslips were mounted onto slides using VectaShield with DAPI. Excitation and emission wavelengths used were 359 nm and 461 nm (DAPI) and 485 nm and 530 nm

(Alexa Fluor 488), respectively. Cells were imaged at 400x and counted in four random fields per slide using Image-J software. The average for the fields was used as the value for each slide. For each experiment, a minimum of three slides were analyzed, and the results of at least three experiments were averaged together, with n values representing number of experiments.

78

Reagents and Antibodies

All test compounds were applied in DMEM-PLUS for 10 min prior to Aβ25-35

exposure, and throughout the Aβ25-35 incubation. The following drugs were used in this

investigation: DTG and BD-1047 (Tocris Biosciences, Ellisville, MO); rimcazole (Sigma-

Aldrich, St. Louis, MO); fura-2 acetoxymethyl ester, DHE, DAF-FM, Live/Dead

Viability/Cytotoxicity Kit, Alexa Fluor 488 Phalloidin (A-12379), Alexa Fluor 488 anti- mouse (A-11001) and anti-rabbit (A-11008) (Invitrogen, Grand Island, NY); Bax

(ab5714), activated caspase-3 (ab32351) and Bcl-2 (ab32370) (abcam, Cambridge, MA);

Afobazole was generously provided by IBC Generium (Moscow, Russian Federation).

Aβ25-35 was obtained from (American Peptides, Sunnyvale, CA).

Data Analysis

Fura-2 fluorescence intensities as a function of time were recorded from microglia

in the same manner as previously described (Katnik et al., 2006, Cuevas et al., 2011a,

2+ Cuevas et al., 2011b) and converted to [Ca ]i using an in situ calibration kit (Invitrogen).

Data were analyzed using SigmaPlot 11 (Systat Software, Inc., San Jose, CA). Data

points represent peak means ± standard error of the mean (S.E.M.). One- or two-way

analysis of variance were used for determining significant differences for multiple group

comparisons, as appropriate, followed by post hoc analysis using the Holm-Sidak test.

Results were considered statistically significant if p < 0.05.

Results

Previously, our laboratory has shown that the pan-selective σ agonist DTG prevents activation of microglia occurring in response to various stimuli, including 79

Figure 4.1

Afobazole blocks membrane ruffling induced by ATP in microglial cells. A, Photomicrographs of microglial cells exposed for 10 min (37° C) to media alone (Control), media containing 30 µM afobazole (Afob), or media containing 30 µM DTG (DTG) in the absence (i, iii, v) and presence (ii, iv, vi) of 100 µM ATP, respectively. Insets show zoomed in images of the indicated regions. Scale bar = 3 µm. B, Bar graph of mean degree of membrane ruffling (±SEM) observed in cells exposed to the same conditions as (A). Asterisks denote significant difference between Control and ATP groups within Media, Afob and DTG (p < 0.001, 0.05, & 0.01, respectively); pound symbols denote significant difference within ATP between Media and Afob or Media and DTG (p < 0.001 for both). For all groups, n > 67. lipopolysaccharide, ATP and MCP-1 (Hall et al., 2009a, Cuevas et al., 2011b). While afobazole preserves the functional capacity of microglia and responsiveness to ATP

(Figure 4.1), data from our laboratory suggest that afobazole can reduce activation of

80

these cells by ATP (Cuevas et al., 2011b). This previous work had shown that afobazole

inhibits migration of microglia, but did not examine if afobazole prevents the

morphological changes evoked by ATP in microglial. Incubation of the microglia in 100

µM ATP for 10 min (37° C) resulted in retraction of filopodia and increased membrane

ruffling (Figure 4.1A ii). Co-incubation of the cells in 30 µM afobazole or 30 µM DTG

decrease membrane ruffling elicited by ATP (Figure 4.1A iv & vi). Quantification of the

degree of membrane ruffling shows that ATP significantly increases membrane ruffling

by 271 ± 14% and that this change is statistically reduced by both afobazole and DTG,

49 ± 5 and 37 ± 5% respectively (Figure 4.1B).

Experiments were carried out to see afobazole could also block microglial

activation caused by application of amyloid-β25-35 (Aβ25-35). One of the events associated with microglial activation that has been shown to be regulated by σ receptors is membrane ruffling (Hall et al., 2009a). Thus, we examined how afobazole affects morphological changes induced by the amyloid peptide in these cells. In cultured, quiescent microglia, filopodia are readily observed, and these membrane processes are not directly affected by incubation of the cells in 30 µM afobazole or a second pan- selective σ agonist, DTG (30 µM) (Figure 4.2A). Incubation of the microglia in 25 µM

Aβ25-35 for 10 min (37° C) resulted in retraction of filopodia and increased membrane

ruffling (Figure 4.2A ii). When afobazole (30 µM) was applied along with the Aβ fragment,

there was a decrease in membrane ruffling compared to Aβ25-35 alone (Figure 4.2A iv).

Pronounced membrane ruffling was still observed when DTG was added with Aβ25-35

(Figure 4.2A vi). Quantification of the degree of membrane ruffling in microglia in

81

Figure 4.2

Afobazole blocks membrane ruffling induced by Aβ25-35 in microglial cells. A, Representative photomicrographs of microglial cells exposed for 10 min (37° C) to media, media containing 30 µM afobazole, or media containing 30 µM DTG in the absence (i, iii, v) and presence (ii, iv, vi) of 25 µM Aβ25-35, respectively. Fields of view of each photomicrograph are representative regions within larger image used to calculate membrane ruffling. Insets show expanded images of the indicated regions. Scale bar = 3 µm. B, Bar graph of mean degrees of membrane ruffling (±SEM) observed in cells incubated in media alone (Media), media with 30 µM Afobazole (Afob), and media with 30 µM DTG (DTG), in the absence (Control) or presence of 25 µM Aβ25-35 (Aβ). Asterisks denote significant difference between Control and Aβ groups within Media and DTG, respectively (p < 0.001 for both). Pound symbols denote significant difference from Media within Aβ (Afob, p < 0.001; DTG p < 0.05), and dagger indicates significant difference between Afob and DTG, also within Aβ (p < 0.001). For all groups, n ≥ 35.

experiments identical to those of Figure 4.2A shows that Aβ25-35 increases membrane

ruffling by more than 5-fold (Figure 4.2B). Incubation of the cells in either afobazole or

82

DTG significantly reduced the degree of membrane ruffling caused by Aβ25-35 (Figure

4.2B). However, the effect of afobazole on Aβ25-35-induced membrane ruffling was

significantly greater than that produced by DTG, with the membrane ruffling being

decreased by 70 ± 7% and 21 ± 9% by these drugs, respectively (Figure 4.2B).

In addition to membrane ruffling, Aβ25-35 induces chemotaxis of microglia, and σ receptor activation has been shown to disrupt migration of microglia in response to other stimuli (Hall et al., 2009a, McLarnon, 2012). Experiments were therefore carried out to determine if application of afobazole can inhibit microglial migration caused by Aβ25-35. A

concentration-response relationship for afobazole reduction in microglial migration when

Aβ25-35 is used as the chemoattractant is shown in Figure 4.3A. Afobazole, at concentrations as low as 10 µM, reduced the number of microglia migrating in response to Aβ25-35. Half maximal inhibition of Aβ25-35-induced migration was observed at 122 µM

afobazole and maximal recorded block was achieved at 1 mM afobazole (Figure 4.3A).

To verify that σ receptors are involved in afobazole suppression of microglial

activation by Aβ25-35, the σ receptor subtype-specific antagonists BD-1047 and rimcazole

were used to block the effects of afobazole on migration. Afobazole was applied at 30

µM in the absence and presence of the selective σ-1 and σ-2 receptor antagonist, BD-

1047 (10 µM) (Matsumoto et al., 1995) and rimcazole (300 nM) (Gilmore et al., 2004),

and migration in response to Aβ25-35 was assessed. Afobazole alone decreased

microglial migration caused by Aβ25-35 by 29 ± 1%.This inhibition was reduced to 10 ± 1%

and 8 ±1% when afobazole was combined with BD-1047 and rimcazole, respectively

83

(Figure 4.3B). Therefore, inhibition of either σ-1 or σ-2 significantly reduced afobazole- mediated suppression of microglial activation by Aβ25-35.

Figure 4.3

Afobazole blocks the migration of microglia elicited by Aβ25-35. A, Concentration- response relationship for afobazole inhibition of microglial migration evoked by 25 µM Aβ25-35. Solid line represents best fit to the data using a one-component Langmuir-Hill equation, with the IC50 value and Hill coefficient being 122 µM and 0.6, respectively. Points represent mean (±SEM) normalized to control values (no afobazole), for all points n = 36. B, Bar graph of the mean (± S.E.M.) percent inhibition of Aβ25-35-induced migration of microglial cells by afobazole (30 µM) in the absence (Control) and presence of 10 µM BD-1047 (BD-1047) or 300 nM rimcazole (Rimcazole). Asterisks denote significant difference from Control (p < 0.001 and n = 24 for all).

The microglia-mediated inflammatory response caused by Aβ is in part mediated via

upregulation in the production of ROS and NO (Weldon et al., 1998, Bianca et al., 1999,

Combs et al., 2001). Therefore, experiments were carried out to determine if afobazole

84

affects ROS or NO levels in microglia following incubation in Aβ25-35. Incubation in afobazole alone produced a decrease in basal levels of ROS in isolated microglia (Figure

4.4A). As predicted, incubation of microglia for 24 hr in Aβ25-35 increased ROS in the cells. However, afobazole failed to block this increase in ROS. Unlike ROS, basal NO

Figure 4.4 Afobazole does not decrease microglial ROS or NO production induced by Aβ25-35. A, Bar graph of mean (±SEM) DHE fluorescence intensity (ROS levels) measured in microglial cells incubated for 24 hr in the absence (Control) or presence of 25 µM Aβ25-35 (Aβ) without (Media) and with 30 µM Afobazole (Afob). Asterisks denote significant difference between Control and Aβ within Media and Afob groups, respectively (p < 0.05). Pound symbol indicates significant difference between Media and Afob groups within Control (p < 0.05). For all groups, n ≥ 149. B, Bar graph of mean (±SEM) DAF-FM fluorescence intensity (NO levels) measured in microglia incubated for 24 hr in absence (Control) or presence of 25 µM Aβ25-35 (Aβ) without (Media) and with 30 µM Afobazole (Afob). Asterisks denote significant difference between Control and Aβ groups within Media and Afob groups (p < 0.05 for both). For all groups, n ≥ 200.

85

levels in microglia were not significantly affected by afobazole (Figure 4.4B). Aβ25-35 did

produce a small but statistically significant increase in NO, and as with ROS, afobazole

failed to block this effect of the amyloid peptide (Figure 4.4B).

Given that microglial cell senescence has been suggested to contribute to AD pathology and that Aβ has been shown to produce microglial apoptosis (Korotzer et al.,

1993, Streit et al., 2009), experiments were carried out to determine how activation of σ receptors with afobazole affects microglial survival following application of Aβ25-35. Figure

4.5A shows representative photomicrographs of microglia labeled with EthD-1 following

72 hr (37º C) incubation in media alone or media containing Aβ25-35 in the absence and

presence of 30 µM afobazole. Few apoptotic microglia were detected when the cells

were incubated in media alone (Figure 4.5A i), but there was a significant increase in

EthD-1-positive cells following Aβ25-35 application (Figure 4.5A ii). Afobazole alone did

not visibly alter the number of EthD-1-positive cells (Figure 4.5A iii), but there was a

decrease in the number of apoptotic cells in the afobazole- Aβ25-35 group (Figure 4.5A iv)

relative to Aβ25-35 alone (Figure 4.5A ii). In identical experiments, Aβ25-35 was found to

produce a nearly 3-fold increase in the number of apoptotic cells relative to control (no

Aβ25-35) (Figure 4.5B). However, afobazole application significantly counteracted the pro-

apoptotic effects of Aβ25-35 such that no significant increase in EthD-1 labeling was

observed in the Aβ25-35 + afobazole group (Figure 4.5B).

Microglial cell death induced by Aβ25-35 has been linked to upregulation of the pro-

apoptotic gene, Bax (Jang et al., 2005). Immunocytochemical experiments were

conducted to determine if levels of Bax expression increase in our rat microglia model

86

and if afobazole can affect expression of this protein. Representative photomicrographs

of microglia double labeled with DAPI and anti-Bax antibody are shown in Figures 4.6A.

Whereas Bax labeling was rarely observed in control (untreated) microglia, Bax

immunolabeling was prominent in cultures treated for 24 hr (37º C) with Aβ25-35 (Figures

4.6A i & ii). Incubation of microglia in afobazole alone had no effect on Bax expression

Figure 4.5

Afobazole decreases microglial cell death produced by application of Aβ25-35. A, Photomicrographs showing EthD-1 labeling (red) of microglial cells following 72 hr incubation in media alone (i), or media containing 25 µM Aβ25-35 (Aβ) (ii), 30 µM afobazole (Afob) (iii), or 30 µM afobazole + 25 µM Aβ25-35 (iv). Fields of view shown are representative regions within larger images used to calculate cell death. B, Bar graph of the mean (±SEM) relative number of apoptotic cells identified by EthD-1 labeling in multiple experiments when the cultures were incubated under the same conditions as (A). Data for each experiment were normalized to the mean cell death observed with media alone (Media, Control). Asterisk denotes significant difference between Control and Aβ groups within Media (p < 0.05). Pound symbol indicates significant difference between Media and Afob groups within Aβ (p < 0.05) (n ≥ 12) Scale bar in (i) is 10 µm.

87

levels, but application of the σ agonist inhibited Aβ25-35 -induced elevated Bax expression

(Figure 4.6A iii & iv). Compilation of data from several identical experiments indicates

Figure 4.6

Afobazole reduces Aβ25-35-evoked expression of the pro-apoptotic gene product Bax, in microglia. A, Photomicrographs of merged images of cultured microglia exposed for 24 hr to media alone (i), or media containing 25 µM Aβ25-35 (Aβ) (ii), 30 µM afobazole (iii) or 25 µM Aβ25-35 + 30 µM afobazole (iv). Microglia were double labeled with DAPI (blue) and anti-Bax antibody (green). Fields of view shown are representative regions within larger images used to calculate Bax expression. B, Bar graph of the mean (±SEM) number of Bax-positive microglia in experiments in which the cells were exposed to media (Control) or 25 µM Aβ25-35 (Aβ) in the absence (Media) or presence of 30 µM afobazole (Afob). Data are expressed as the percent of Bax positive microglia, using DAPI to identify the number of microglia per slide. Asterisks denote significant difference between Control and Aβ within Media and Afob (p < 0.001 for both). Pound symbol indicates significant difference between Media and Afob within Aβ (p < 0.001). For all, n = 9. Scale bar in (i) is 10 µm.

88

that approximately 20% of control microglia expressed Bax, but that this number increases to over 80% following Aβ25-35 incubation (Figure 4.6B). While an increase in

Bax expression was noted when microglia were incubated in a combination of afobazole

and Aβ25-35, there was a 54 ± 4% reduction in the number of cells showing Bax reactivity

when compared to microglia incubated in Aβ25-35 alone (Figure 4.6B).

Caspase-3 has also been implicated in Aβ25-35-induced microglial apoptosis, and

σ-1 receptor activation has been shown to induce caspase-3 downregulation in these cells (Jang et al., 2005, Tchedre and Yorio, 2008). Thus, we examined the effects of

afobazole on the levels of the death protease, caspase-3, after microglial cell were

exposed to Aβ25-35 for 24 hr (37º C). Incubation of microglia in Aβ25-35 increased the

number of cells expressing the active form of caspase-3 relative to control (Figure 4.7A i

& ii). While afobazole alone had no significant effect on activated caspase-3 expression, afobazole inhibited the increases in active caspase-3 evoked by Aβ25-35 (Figure 4.7A iii & iv). In identical experiments, comparing the number of cells expressing activated caspase-3 in cultures treated with Aβ25-35 alone to those treated with Aβ25-35 + afobazole

indicates that afobazole reduces caspase-3 expression by 47 ± 5% (Figure 4.7B).

The anti-apoptotic gene product, Bcl-2, has been shown to be upregulated by

interventions that decrease microglial cell death caused by Aβ25-35 (Jang et al., 2005). In

neurons, the expression levels of this protein increase after 24 hr exposure to Aβ and

then decreases after 48 hr (Kim et al., 1998). Thus, to explore how afobazole protects

microglia from Aβ25-35-induced apoptosis, we used the 48 hr time point to assess how

afobazole affects Bcl-2 expression. Bcl-2 was readily detected at 48 hr in microglia under

all conditions tested (Figures 4.8A). Quantification of the data shows that afobazole 89

application produced a small, but significant, increase of 14 ± 1% in the number of

microglia expressing Bcl-2 (Figure 4.8B). Similarly, Aβ25-35 alone increased the number

of cells expressing Bcl-2 by 11 ± 1%. While afobazole co-application did not increase

Figure 4.7

Afobazole reduces expression of activated caspase-3 in response to application of Aβ25- 35 in microglia. A, Photomicrographs of merged images of cultured microglia exposed for 24 hr to media alone (i) or media containing 25 µM Aβ25-35 (ii), 30 µM afobazole (iii) or 25 µM Aβ25-35 + 30 µM afobazole (iv). Microglia were double labeled with DAPI (blue) and anti-activated caspase-3 antibody (green). Fields of view shown are representative regions within larger images used to calculate activated caspase-3 expression. Scale bar in (i) is 10 µm. B, Bar graph of mean (±SEM) percent of caspase-3 positive microglia in experiments in which the cells were exposed to media (Control) or 25 µM Aβ25-35 (Aβ) in the absence (Media) or presence of 30 µM afobazole (Afob). Asterisks denote significant difference between Control and Aβ groups within Media and Afob (p < 0.001); pound symbol indicates significant difference between Media and Afob within Aβ (p < 0.001). For all condition, n =9.

90

Bcl-2 expression significantly above afobazole alone, the combination of afobazole and

Aβ25-35 yielded a small but significant increase (4 ± 1%) in the number of microglia

expressing Bcl-2 after 48 hr when compared to Aβ alone (Figure 4.8B).

Figure 4.8

Afobazole has minor effects on Bcl-2 levels in microglia following application of Aβ25-35. A, Photomicrographs of merged images of cultured microglia exposed for 24 hr to media alone (i) or media containing 25 µM Aβ25-35 (ii), 30 µM afobazole (iii) or 25 µM Aβ25-35 + 30 µM afobazole (iv). Microglia were double labeled with DAPI (blue) and anti-Bcl-2 antibody (green). Areas shown are representative regions within larger images used to calculate Bcl-2 expression. Scale bar in (i) is 10 µm. B, Bar graph of mean (±SEM) percent of Bcl-2 positive microglia in experiments in which the cells were exposed to media (Control) or 25 µM Aβ25-35 (Aβ) in the absence (Media) or presence of 30 µM afobazole (Afob). Asterisk denotes significant difference between Control and Aβ within Media (p < 0.01). Pound symbols indicate significant difference between Media and Afob within Control and Aβ, respectively (p < 0.01 for both, n = 9 for all).

91

While our observations indicate that activation of σ receptors with afobazole

decreases microglial cell death, it is unclear if the surviving cells have been

compromised such that they fail to exhibit responses to normal stimuli. Thus, functional

2+ studies were carried out to evaluate microglia using ATP to stimulate changes in [Ca ]i

following Aβ25-35 application in the absence and presence of afobazole. Figure 4.9A

2+ shows a series of traces of [Ca ]i as a function of time recorded from four microglia

incubated in Aβ25-35 for 24 hr and exposed to ATP in the absence and presence of

2+ afobazole. Focal application of 100 µM ATP produced transient elevations in [Ca ]i in

microglia under control conditions and when the cells were incubated in 25 µM Aβ25-35

2+ (Figure 4.9A, top traces). However, the ATP-induced elevations in [Ca ]i were greatly

reduced in the microglia incubated in Aβ25-35. In contrast, microglia incubated in 30 µM

2+ afobazole displayed similar ATP-induced increases in [Ca ]i in absence and presence of

the Aβ fragment (Figure 4.9A, bottom traces). In identical experiments, ATP evoked

2+ comparable increases in [Ca ]i in microglia incubated in media alone (Control) and 30

µM afobazole (Figure 4.9B). However, microglia incubated in Aβ25-35 showed a

2+ statistically significant 34 ± 4% reduction in ATP-evoked [Ca ]i responses (Figure 4.9B).

While afobazole pre-incubation did not have a long-term effect on ATP-evoked responses, co-application of 30 µM afobazole along with the 25 µM Aβ25-35 rescued the

2+ ATP-evoked responses such that the increases in [Ca ]i observed in this group were

2+ statistically similar to both the Control group and the afobazole alone group. The [Ca ]i

responses in the afobazole + Aβ25-35 group were 44 ± 8% greater than the Aβ25-35 only

group (Figure 4.9B).

92

Figure 4.9

2+ Afobazole preserves [Ca ]i responses to ATP in microglial cells exposed to Aβ25-35. A, 2+ representative traces of [Ca ]i as a function of time recorded from four microglia during 10 second application of 100 µM ATP following 24 hr incubation in media alone (Control), or media containing 25 µM Aβ25-35 (Aβ), 30 µM Afobazole (Afob) or 25 µM Aβ25-35 + 30 µM Afobazole (Afob + Aβ). The afobazole and Aβ were washed out for 5 min 2+ prior to application of ATP. Line above [Ca ]i responses indicates duration of ATP 2+ 2+ application. B, Bar graph of mean (± SEM) peak increases in [Ca ]i (∆[Ca ]i) evoked by 2+ application of 100 µM ATP onto isolated microglia. [Ca ]i responses were recorded from microglia in experiments identical to those performed in (A). Asterisk symbolizes significant difference between Control and Aβ within Media (p < 0.001). Pound symbol denotes significant difference between Media and Afob within Aβ (p < 0.001). For all conditions, n ≥ 33.

93

Discussion

The most salient finding reported here is that activation of both σ-1 and σ-2 receptors by afobazole rescues microglia from the deleterious consequences of Aβ25-35

exposure. Afobazole application reduced the activation of microglia caused by incubation

of the cells in Aβ25-35 as evident by a decrease in membrane ruffling and cell migration

induced by the Aβ fragment. The apoptosis of microglia induced by Aβ25-35 was also

reduced by afobazole. This decrease in cell death is likely due to a reduction in the

expression of the pro-apoptotic gene, Bax, and the death protease, caspase-3.

Afobazole also preserved functional responses of the microglia to ATP after Aβ25-35

incubation, which has significant implications for Alzheimer’s disease progression.

Afobazole application reduced the activation of microglia caused by incubation of

the cells in ATP, as evident by a decrease in membrane ruffling. Previous studies from

our laboratory have shown that microglia quickly become activated and withdraw their

filopodia when exposed to various stimuli, such as LPS and ATP (Hall et al., 2009a).

This activated state, which allows the microglia to migrate to the site of injury, is

preceded by the release of neurotoxic substances (Kierdorf and Prinz, 2013). Previously

we have shown that microglia, in response to ATP, alter their morphology to a less

ramified state and this reaction was inhibited by DTG (Hall et al., 2009a). Afobazole

effectively blocked membrane ruffling by approximately 50% in response to ATP. For

comparison we used a second pan-selective σ receptor agonist, DTG, which reduced

ruffling comparable to afobazole and to the extent as previously reported.

Previous studies have shown that microglia rapidly respond to Aβ by withdrawing

their filopodia and assuming an amoeboid, ruffled morphology (Araujo and Cotman, 94

1992). This early event is indicative of microglial activation and precedes the inflammatory response that includes release of neurotoxic substances from the microglia

(Combs et al., 2001, Garcao et al., 2006, Orellana et al., 2011). Afobazole (30 µM)

effectively blocked membrane ruffling in response to Aβ25-35. While a second pan-

selective σ receptor agonist, DTG (30 µM), also decreased membrane ruffling caused by

Aβ25-35, this drug was significantly less potent than afobazole. It has been shown that

higher concentrations of DTG (100 µM) are also required to block ATP-induced

microglial membrane ruffling (Hall et al., 2009a).

Afobazole also effectively blocked microglial migration induced by Aβ25-35. It has

been shown previously that various Aβ fragments, including Aβ25-35, are microglial chemotactic agents (Davis et al., 1992). The Aβ-induced chemotaxis is in part due to release of ATP from microglia following Aβ25-35 application and subsequent activation of

P2Y2 receptors on these cells (Kim et al., 2012). We have previously shown that afobazole blocks microglial migration induced by activation P2Y receptors (Cuevas et al.,

2011b). Afobazole appears to inhibit microglial migration caused by UTP via σ receptor-

mediated disruption in P2Y signaling, since afobazole treatment also inhibited P2Y-

2+ induced elevation in [Ca ]i in microglia (Cuevas et al., 2011b). The IC50 for afobazole

inhibition of microglial migration caused by Aβ25-35 is 120 µM, which is similar to the low-

affinity site reported for afobazole inhibition of UTP-induced microglial migration (102

µM) (Cuevas et al., 2011b). Therefore, afobazole may in part prevent microglial

chemotaxis caused by Aβ25-35 via σ receptor-mediated inhibition of P2Y receptors in

these cells.

95

Data shown here indicate that both σ receptor subtypes are involved in afobazole

inhibition of Aβ25-35-evoked microglial activation. Afobazole-mediated inhibition of

microglial migration was reduced by ~50% by 10 µM BD 1047, which is indicative of a σ-

1 receptor mediated effect (Matsumoto et al., 1995). An identical concentration of BD

1047 had a similar effect on afobazole inhibition of ATP-induced microglial cell migration

(Cuevas et al., 2011b). Likewise, involvement of σ-2 receptors is confirmed by results obtained using the σ receptor antagonist, rimcazole, which binds to σ-2 receptors with nanomolar affinity (Ferris et al., 1986, Rybczynska et al., 2008). Nanomolar concentrations of rimcazole also blocked afobazole inhibition of microglial migration in response to ATP (Cuevas et al., 2011b).

Given that one of the consequences of microglial activation is increased oxidative and nitrosative stress, which leads to Aβ25-35 toxicity (Weldon et al., 1998, Bianca et al.,

1999, Combs et al., 2001), we examined how afobazole affects Aβ25-35-induced

increases in cellular ROS and NO levels. Consistent with previous reports, both ROS

and NO levels were increased significantly following Aβ25-35 application. However,

afobazole failed to attenuate the increases in ROS and NO evoked by the Aβ fragment.

Activation of σ receptors has been shown to decrease oxidative injury and nitrosative

stress in various models. It was recently shown, using σ receptor knockout mice, that σ-1

has a dual function of both reducing oxidative stress and activating antioxidant response

elements (Pal et al., 2012). In microglia, stimulation of σ receptors by DTG reduces NO

production in response to lipopolysaccharide (Hall et al., 2009a). Taken together,

however, our data suggest that the reduction in Aβ-evoked microglial cell toxicity

96

provided by afobazole application is not the result of σ receptor-mediated decrease in

oxidative or nitrosative stress.

While decreased activation of microglia will be beneficial in AD by reducing the

neuroinflammatory response, recent data suggests that enhanced microglial survival is

also important for improved outcomes given the ability of these cells to phagocytose Aβ

plaques (Streit et al., 2009, Solito and Sastre, 2012). Application of 30 µM afobazole

significantly reduced the number of microglia undergoing apoptosis following 72 hr

incubation in Aβ25-35. The concentrations of afobazole shown to be effective here are identical to those previously shown to decrease microglial cell death caused by ischemia in a σ receptor dependent manner and consistent with the binding affinity of afobazole for σ-1 receptors (Seredenin et al., 2009, Cuevas et al., 2011a, Cuevas et al., 2011b).

This observation suggests that afobazole is similarly acting through σ receptor activation to prevent microglial cell death. Both melatonin and N-acetylcysteine have been shown to decrease Aβ25-35-evoked apoptosis in microglia via a reduction in ROS (Jang et al.,

2005). Given that afobazole does not appear to affect Aβ25-35-elicited increases in ROS,

it is unlikely that afobazole provides cytoprotection via a similar mechanism to these

compounds.

Our findings demonstrate that afobazole mitigates Aβ25-35-induced apoptosis of

microglia by decreasing expression of the pro-apoptotic gene, Bax, and the death

protease caspase-3 while preserving the levels of the anti-apoptotic gene, Bcl-2. An increase in Bax levels has been observed in both neurons and microglia of AD brains

(Su et al., 1997). Moreover, interventions that decrease Aβ25-35-induced microglial

97

apoptosis have been shown to lower Bax expression (Jang et al., 2005, Shang et al.,

2012). Likewise, caspase activity has been detected in plaque-associated neurons and

microglia in AD brains (Yang et al., 1998). Caspase-3 levels have been shown to be upregulated in microglia following Aβ administration, while reduced levels of this death

protease have been associated with inhibition of Aβ toxicity (Shang et al., 2012).

Conversely, expression of the anti-apoptotic gene, Bcl-2, is inversely correlated with

disease severity in AD. Increases in Bcl-2 levels promote microglial survival following

exposure to Aβ25-35 (Satou et al., 1995, Jang et al., 2005). Therefore, afobazole likely

enhances microglial survival and resistance to Aβ25-35 toxicity via a complex response

involving both anti-apoptotic and pro-apoptotic genes. This multifaceted effect is

consistent with afobazole acting as a σ agonist, since σ receptor activation has been

shown to modulate Bax, caspase-3 and Bcl-2 levels (Tchedre and Yorio, 2008, Meunier

and Hayashi, 2010).

Importantly, not only did afobazole protect microglia from Aβ25-35-evoked

apoptosis, but application of the σ agonist preserved the functional capacity of these

2+ cells, as evident from the continued [Ca ]i responses to ATP. Ultimately, one of the

theoretical benefits to increased microglial survival would be continued clearance of Aβ, as this peptide has been shown to be phagocytosed by microglia (Wilcock et al., 2004,

Herber et al., 2007). This hypothesis is further supported by the recent observation that microglial clearance of Aβ directly correlates with expression of the gene CD33, such that a CD33 deficiency reduces brain levels of both insoluble Aβ1-42 and amyloid plaque

burden in a mouse AD model (Griciuc et al., 2013). Moreover, there is a decrease in both

98

CD33 expression and insoluble Aβ1-42 in the brains of individuals expressing the minor

allele of the CD33 (SNP rs3865444) gene, which was identified previously as imparting

protection against AD (Naj et al., 2011, Griciuc et al., 2013). The fact that responses to

ATP itself are preserved is significant, since the clearance of Aβ has been shown to be

dependent of the activation of P2Y2 receptors. Microglia respond to Aβ by releasing ATP

and this nucleotide functions in a paracrine manner, acting on P2Y2 receptors expressed

on the microglia to enhance Aβ uptake by these cells (Kim et al., 2012). We have

2+ previously shown that in our model these increases in [Ca ]i are due to activation of P2Y

receptors, since they can be elicited by UTP (Cuevas et al., 2011b). Therefore, by

preserving ATP responses in microglia, afobazole will permit these cells to respond to Aβ in vivo and potentially decrease amyloid burden.

In conclusion, our study demonstrates that afobazole can mitigate activation of microglial cells in response to Aβ25-35. Afobazole also reduced microglial toxicity caused

by Aβ25-35, but this cytoprotection does not involve regulation of either ROS or NO

production caused by the amyloid peptide. The observed decrease in apoptosis is due to

afobazole-mediated regulation of multiple genes, with a noted decrease in Bax and

caspase-3 and a concomitant increase in Bcl-2 expression. In addition to the decrease in cell death, afobazole preserved functional activity of the cells as evident by the

2+ conserved ATP-induced increases in [Ca ]i following prolonged incubation in Aβ25-35.

Taken together our data suggest that afobazole may prevent microglia from entering the

activated state associated with neurotoxicity following exposure to Aβ, and increasing

99

their survival, which will facilitate clearance of the amyloid peptide. These properties make afobazole an attractive drug for potential Alzheimer’s therapeutics.

100

CHAPTER 5

Activation of Sigma Receptors via Afobazole Modulates Microglial but not Neuronal Apoptotic Expression in Response to Long-Term Ischemia Exposure

Introduction

Our laboratory has previously shown that stroke is a multicellular disease and that any attempts to treat this disease must involve several different cell types (Hall et al.,

2009b, Leonardo et al., 2010, Cuevas et al., 2011a, Cuevas et al., 2011b). Neurons have previously been the sole target in the treatment of stroke, but we have shown that microglia cells play a dynamic roll the pathophysiology of stroke (Hall et al., 2009a,

Cuevas et al., 2011b). In response to stroke or other types of CNS injury, microglia become activated and migrate to the site of injury (Kierdorf and Prinz, 2013), while the initial action is neuroprotective in nature, overtime microglial cells become over-activated and neurotoxic (Hellwig et al., 2013). It is possible that the injury occurring, following the days and weeks after a stroke, is due to microglia over-activation (Hellwig et al., 2013).

However, completely inhibiting activation and migration of microglial cells would result in unintended, detrimental side effects as microglia play an important, neuroprotective role in the early stages following a stroke insult (Kitamura et al., 2004). Therefore, it is important to provide neuroprotection by regulating neuronal and microglial mechanisms.

Previously studies have shown that sigma (σ) receptors play a vital role in neuroprotection and cell regulation as a whole (Ajmo et al., 2006, Katnik et al., 2006, Su 101

et al., 2010). Largely distributed throughout the mammalian body, these receptors perform a number of functions such as regulating membrane ion channels (Hayashi et al., 2000, Tchedre et al., 2008), altering expression of antiapoptotic and proapoptotic transcription factors (Yang et al., 2007, Tchedre and Yorio, 2008, Meunier and Hayashi,

2010) and modulating activation and migration of microglial cells (Hall et al., 2009a).

Once thought to be a type of , sigma receptors were later placed into their own class of opioid-like receptors (Walker et al., 1990, Seth et al., 1998). Consisting of two subtypes, σ-1 & σ-2, these receptors have been shown to provide protection to both neurons and glial cells following in vitro ischemia and in vivo middle cerebral artery occlusion (MCAO) stroke models (Ajmo et al., 2006, Katnik et al., 2006). The σ-1 receptor is an inter-organelle chaperone localized to the mitochondrion-associated ER membrane (MAM) (Su et al., 2010). This σ-1 receptor-MAM complex plays an important role in cell signaling, mitochondrial function, maintaining intracellular calcium homeostasis and the expression of cellular proteins (Tsai et al., 2009). In contrast, σ-2 receptors are not well understood, in part because the gene encoding this receptor has not been cloned. However, σ-2 receptors are known to play important roles in regulating intracellular calcium (Vilner and Bowen, 2000) and are involved in the activation and migration of microglial cells (Cuevas et al., 2011b, Iniguez et al., 2013).

Our laboratory has previously shown the pan-selective sigma receptor agonist afobazole (5-ethoxy-2-[2-(morpholino)-ethylthio]benzimidazole), an anxiolytic currently used clinically in Russia, to be protective in neurons (Cuevas et al., 2011a) and microglia

(Cuevas et al., 2011b) in an in vitro chemical ischemia and acidosis model. Furthermore, afobazole prevented neuronal and glial cell death in an in vivo MCAO stroke model 102

(Katnik et al., 2013). Other studies have suggested that afobazole may provide neuroprotection in a variety of pathophysiological diseased states, which involve oxidative stress and glutamate toxicity (Zenina et al., 2005). Recently our laboratory showed that afobazole maintained neuronal and microglial cell viability when exposed to amyloid-β25-35 by the downregulation of the pro-apoptotic protein Bax and the death protease caspase-3, while preserving Bcl-2 expression (Behensky et al., 2013a, b).

Experiments were carried out to determine how afobazole affects neuronal and

microglial responses to chemical ischemia. Afobazole also reduced ischemia-induced

cell death in both microglia and neurons. In addition, afobazole application prevented

upregulation of the proapoptotic protein Bax and death protease caspase-3 in microglia but not in neurons. Conversely, Bcl-2 expression was preserved in microglia, but not neurons, by afobazole following exposure to ischemia. This study demonstrates that one way in which afobazole maintains cellular viability in microglia cells is through the regulation of apoptotic proteins. We also show that afobazole provides neuroprotection against chemical ischemia in neurons, but by mechanisms other than direct regulation of apoptosis.

Materials and Methods

Primary Cultures of Microglia

Primary cultures of microglia were prepared from Sprague-Dawley mixed sex rat

pups (post natal day 2-3) as previously described by our laboratory (Hall et al., 2009a,

Cuevas et al., 2011b). The mixed glial cultures were incubated for 7-12 days at 37 o C

prior to experiments being carried out. Microglia were mechanically separated from the 103

cultures by brief shaking. Isolated microglia were resuspended in DMEM PLUS

containing: 500 mL Dulbecco’s Modified Eagle Medium with 4.5 g/L glucose, L-glutamine

and sodium pyruvate (DMEM) (Corning Cellgro, Manassas, VA), 40 mL horse serum

(Corning Cellgro), 12.5 mL heat-inactivated fetal bovine serum (Thermo Scientific

Hyclone, Logan, UT ), and 5 mL 10x antibiotic/antimycotic (Corning Cellgro). These cells

were used immediately for cytotoxicity assays or plated on glass coverslips for one day

for imaging experiments.

Preparation of Cortical Neurons

All experiments were carried out on cultured cortical neurons from mixed sex

embryonic day 18 (E18) Sprague-Dawley rats (Harlan, Indianapolis, IN). Methods used

here were identical to those previously reported for the isolation and culturing of these

cells (Katnik et al., 2006). Cortical neurons were plated on poly-L-lysine coated

coverslips and cultured in B27 and L-glutamine supplemented Neurobasal medium

(Neurobasal Complete) (Life Technologies, Grand Island, NY). Cells were used for experiments after 10-21 days in culture, which permits synaptic contact formation and yields robust responses to other pathophysiological conditions such as in vitro ischemia and acidosis (Katnik et al., 2006, Herrera et al., 2008). Animals were cared for in accordance with the regulations and guidelines set forth by the University of South

Florida’s College of Medicine Institution on Animal Care and Use Committee.

Live/Dead Cell assay

Neurons or microglia plated on poly-L-lysine coverslips were incubated at 37°C for

72 hr in Neurobasal Complete or DMEM Plus, respectively, with 4 mM sodium azide in 104

the absence (Control) and presence of 100 µM afobazole for neurons and 30 µM

afobazole or for microglia. The coverslips were rinsed with PBS followed by 30 min

incubation at room temperature in PBS with 4 µL of a 2mM ethidium homodimer-1 (EthD-

1) dissolved in 1:4 DMSO:H2O solution. The coverslips were washed with PBS and

deionized water, dried, and mounted on a microscope slide with Vectashield Hardset

mounting media (Vector Labs, Burlingame, CA). EthD-1 loaded cells were illuminated

with light at 530 nm and visualized at 645 nm using a Zeiss Axioskop 2 equipped with a

20X objective. EthD-1-positive cells were identified and counted using Image-J in four

random fields per slide, and the average for the fields used as the value for the slide. For

each experiment, a minimum of three slides were used, and the results of at least three

experiments were averaged together for each condition tested.

Immunocytochemistry

Neurons or microglia plated on poly-L-lysine coverslips were incubated at 37°C for

24 hr in Neurobasal complete or DMEM Plus with 4 mM sodium azide in the absence

and presence of 100 µM or 30 µM afobazole, respectively. The coverslips were rinsed

with phosphate-buffered saline (PBS) followed by an ethanol step-wise fixation. The cells were permeabilized with 0.1% Triton X in PBS for 15 min, then rehydrated with PBS and washed with 0.5% bovine serum albumin (BSA) in PBS. Blocking was achieved with 45 min incubation in 2% BSA, followed by multiple washes with 0.5% BSA. Primary antibodies were diluted in PBS with 0.5% BSA and applied onto cells at 4°C for 24 hr.

Primary antibody dilutions were as follows: Bax 1/20, Caspase-3 1/25 and Bcl-2 1/100.

The cells were then washed multiply times with 0.5% BSA in PBS and then incubated in

105

secondary antibodies for 60 min at room temperature. Secondary antibodies were either

Alexa Fluor 488 conjugated anti-mouse or anti-rabbit diluted at a ratio of 1/300 in PBS with 0.5% BSA. Following incubation in secondary antibodies, cells were washed with

0.5% BSA in PBS and then with PBS alone. Coverslips were rinsed with deionized water and inverted onto a slide VectaShield containing DAPI. DAPI and the Alexa Fluor 488 conjugated secondary antibodies were illuminated at 359 and 485 nm and visualized at

461 and 530 nm, respectively, using a Zeiss Axioskop 2 outfitted with a 40X objective.

Images of DAPI and Alexa Fluor 488 positive cells were counted and merged to demonstrate co-localization using Image-J software. Four random fields per slide were

used to determine an average value for the slide. For each experiment, a minimum of

three slides were used, and the results of at least three experiments were averaged

together.

Calcium Imaging

Intracellular Ca2+ concentrations were measured in isolated cortical neurons using

ratiometric fluorometry as previously described (Katnik et al., 2006). Neurons were

loaded with fura-2 by adding 3 µg/ml of the acetoxymethylester form of fura-2 (fura-2

AM) in 0.3% DMSO to the media and incubating the cells at room temperature for 1 hr.

Prior to beginning the experiments, the coverslips were rinsed with physiological saline

solution (PSS) consisting of (in mM): 140 NaCl, 5.4 KCl, 25 HEPES, 20 glucose, 1.3

2+ CaCl2, and 1.0 MgCl2 (pH to 7.4 with NaOH). For each cell, [Ca ]i was measured once a

second for a 100 second period and the values averaged to determine mean peak and

2+ net [Ca ]i.

106

Compounds, Reagents and Antibodies

The following compounds and reagents were used in this investigation: DTG

(Tocris Biosciences, Ellisville, MO); Live/Dead Viability/Cytotoxicity Kit (Life

Technologies); Sodium Azide (ACROS Organics, Fair Lawn, NJ). The following

antibodies were also used: Alexa Fluor 488 anti-mouse (A-11001) and anti-rabbit (A-

11008) (Life Technologies); anti-Bax (ab5714), anti-activated caspase-3 (ab32351) and anti-Bcl-2 (ab32370) (abcam, Cambridge, MA). Afobazole was generously provided by

IBC Generium (Moscow, Russian Federation). Vehicle for drugs and reagents used were either water (H2O), ethanol (EtOH) or dimethyl sulfoxide (DMSO), and appropriate

vehicle controls were carried out for each study.

Data Analysis

Fluorescence intensities were recorded from cortical neurons and microglia in the

same manner as previously described (Katnik et al., 2006, Cuevas et al., 2011a, Cuevas

et al., 2011b). Data were analyzed using Image J or SigmaPlot 11 software (Systat

Software, Inc., San Jose, CA). Data points represent peak means ± standard error of the mean (S.E.M.). One-, two- and three-way analysis of variance were used for determining

significant differences for multiple group comparisons, as appropriate, followed by post

hoc analysis using the Holm-Sidak method. Results were considered statistically

significant if p < 0.05.

Results

Previous studies suggested that activation of σ receptors decreased microglial cell death

in response to ischemia. To further examine the effects of ischemia on survival of 107

microglia, the Live/Dead cytotoxicity assay was used. Ethidium Homodimer-1 (EthD-1) labeled cells were counted in microglia cultures in the absence (Control) and presence of ischemia (Isch). There was significant microglia death when exposed to 72 hr ischemia

Figure 5.1

Afobazole reduces microglia death produced by application of ischemia. A, Photomicrographs showing EthD-1 labeling (red) of microglial cells following 72 hr incubation in media alone (i), or media containing 4 mM Azide (in vitro ischemia, Isch) (ii), 30 µM afobazole (Afob) (iii), or 30 µM afobazole + 4 mM Azide (iv). Fields of view shown are representative regions within larger images used to calculate cell death. B, Bar graph of relative cell death observed following 72 hr incubation of microglia in control (Control) or ischemic condition (Isch), with (Afob) or without (Media) 30 µM afobazole (n > 12). Data were normalized to cell death observed with media alone (Control). Asterisks denote significant difference between Control and Isch within Media and Afob, respectively (p < 0.001 & = 0.001, respectively); pound symbol indicates difference between Afob and Media within Isch (p < 0.001).

108

(Figure 5.1). Afobazole significantly mitigated microglial cell death when co-incubated with ischemia. 72hr incubation in media alone or media with 30µM afobazole had no effect on microglia death, respectively. Counting the number EthD-1-positive cells showed that afobazole mitigates ischemia induced cell death by 75 ± 8%.

The mechanism by which activation of σ receptors may decrease cell death remain unknown. Thus, experiments were carried out to determine if activation of σ receptors alters the expression of molecules involved in cell apoptosis. One pro- apoptotic gene product that has been shown to be affected by ischemia and may regulated by σ receptors is the protein Bax (Schelman et al., 2004, Tchedre and Yorio,

2008). Immunocytochemical experiments were conducted to measure Bax levels following 24 hr ischemia incubation to ascertain and whether afobazole application affects the levels of protein. Ischemia significantly increases the pro-apoptotic gene, Bax, in microglia (Figure 5.2). Co-labeling of microglia with DAPI and an anti-Bax antibody shows that Bax is rarely detected in control cells but is readily visible in microglia following ischemia. Afobazole alone had no effect on Bax levels, but this drug reduced

Bax expression after ischemia incubation. Nearly all microglia show Bax expression after ischemia, but this is reduced by 41 ± 1% when afobazole is co-applied. This reduction in

Bax expression was found to be statistically significant (Figure 5.2).

Bax-dependent caspase-3 activation is a key component in cellular apoptosis

(Cregan et al., 1999). Therefore, we examined how afobazole effects the levels of the death protease, caspase-3, in microglia after ischemia treatment. Following ischemic exposure, the activated form of caspase-3 was observed in a greater number of cells

109

Figure 5.2

Afobazole prevents upregulation of the pro-apoptotic gene, Bax, observed in response to ischemia in microglia. Microglia were double labeled with DAPI (blue) and anti-Bax antibody followed by Alexa Fluor 488 conjugated secondary antibody (green). A, Photomicrographs of merged images of cultured microglia exposed for 24 hr to media alone (i) or media containing 4 mM Azide (Isch) (ii), 30 µM afobazole (iii) or 4 mM Azide + 30 µM afobazole (iv). Microglia were double labeled with DAPI (blue) and anti-Bax antibody (green). Fields of view shown are representative regions within larger images used to calculate Bax expression. Scale bar in (i) is 10 µm. B, Bar graph of mean number of Bax-positive microglia in experiments in which the cells were exposed to normal conditions (Control) or ischemia (Isch) in the absence (Media) or presence of 30 µM afobazole (Afob). Data are expressed as percent (± SEM) of total number of microglia per slide (n = 9). Asterisk denotes significant difference between Control and Isch within Media and Afob, respectively (p < 0.001 for both). Pound symbol indicates significant difference between Media and Afob within Isch (p < 0.001).

relative to control (Figure 5.3). Afobazole alone significantly decreased caspase-3 levels

relative to the control, and diminished the increases in caspase-3 evoked by ischemia.

110

Examining the number of cells showing caspase-3 signal demonstrates that afobazole

reduced the number of caspase-3 positive cells by 53 ± 6% in the absence of ischemia.

Figure 5.3

Afobazole reduces expression of caspase-3 in response to ischemia in microglia. Microglia were double labeled with DAPI (blue) and anti-caspase-3 antibody followed by Alexa Fluor 488 conjugated secondary antibody (green). A, Photomicrographs of merged images of cultured microglia exposed for 24 hr to media alone (i) or media containing 4 mM Azide (Isch) (ii), 30 µM afobazole (iii) or 4 mM Azide + 30 µM afobazole (iv). Fields of view shown are representative regions within larger images used to calculate activated caspase-3 expression. Scale bar in (i) is 10 µm. B, Bar graph of mean number of caspase-3-positive microglia in experiments in which the cells were exposed to normal conditions (Control) or ischemia (Isch) using media alone (Media) of media with 30 µM afobazole (Afob). Data are expressed as percent (± SEM) of total number of microglia per slide (n = 9). Asterisks denote significant difference between Control and Isch within Media and Afob (p < 0.001 for both). Pound symbols indicate significant difference between Media and Afob within Control and Isch, respectively (p < 0.001 for both).

111

Following ischemia, afobazole reduced caspase-3 levels by 59 ± 5%, such that the levels of caspase-3 were similar to those observed under control conditions (no ischemia or afobazole) both of which were statistically significant.

The effects of Bax can be counteracted by upregulation of the anti-apoptotic protein Bcl-2 (Hockenbery et al., 1993, Korsmeyer et al., 1993, Oltvai et al., 1993), which may be a second mechanism by which σ receptors reduce cell death following ischemia.

The relationship between afobazole and expression of the anti-apoptotic gene, Bcl-2, during ischemia in microglia was examined via immunocytochemistry. Following ischemia, the levels of Bcl-2 expressed by microglia decreased by ~20% compared to that of control (Figure 5.4). Afobazole alone produced a ~10% increase in the number of cells expressing Bcl-2. Moreover, afobazole rescued Bcl-2 expression following ischemia. Both afobazole and afobazole with ischemia treated microglia showed nearly

100% expression in all cells.

Previous studies showed that σ receptor activation can decrease neuronal death following a 24 hr ischemic event in vitro. However, it has not been determined if neuroprotection occurs following longer ischemic events such as those used for microglia here. Thus, experiments were carried out to see if afobazole can decrease neuronal death under these conditions. Neurons labeled with EthD-1 were counted in the absence (Control) and presence of ischemia (Isch). There was significant neuronal death when exposed to 72 hr Isch (Figure 5.5). Afobazole significantly mitigated neuronal cell death when co-incubated with ischemia. Incubation in media alone (Control) set the limits of normal neuronal death, whereas media with 100µM afobazole had reduced

112

neuron death beyond control levels. Counting the number EthD-1 positive cells showed that afobazole mitigates ischemia-induced cell death by 49 ± 9%.

Figure 5.4

Afobazole enhances Bcl-2 expression under control conditions and following ischemia in microglia. Microglia were double labeled with DAPI (blue) and anti-Bcl-2 antibody followed by Alexa Fluor 488 conjugated secondary antibody (green). A, Photomicrographs of merged images of cultured microglia exposed for 24 hr to media alone (i) or media containing 4 mM Azide (ii), 30 µM afobazole (iii) or 4 mM Azide + 30 µM afobazole (iv). Areas shown are representative regions within larger images used to calculate Bcl-2 expression. Scale bar in (i) is 10 µm. B, Bar graph of mean number of Bcl-2-positive microglia in experiments in which the cells were exposed to normal conditions (Control) or ischemia (Isch), in the absence (Media) and presence of 30 µM afobazole (Afob). Data are expressed as percent (± SEM) of total number of microglia per slide (n = 9). Asterisk denotes significant difference between Control and Isch within Media (p < 0.001); pound symbols indicate significant difference between Media and Afob within Control and Isch, respectively (p < 0.01 for Control & < 0.001 for Isch).

113

Experiments were next carried out to determine if the enhanced neuronal survival observed following σ receptor stimulation in neurons involves a pathway similar to that observed in microglial cells. As in microglia, ischemia significantly increased the pro- apoptotic gene, Bax, in neurons (Figure 5.6). Co-labeling neurons with DAPI and an anti-

Bax antibody showed that Bax is detected in control cells but increased in neurons

Figure 5.5

Afobazole reduces neuronal death induced under ischemic conditions. Bar graph of relative cell death observed following 72 hr incubation of neurons in media (Media) or media containing ischemia (Isch), 100 µM afobazole (Afob) or ischemia + 100 µM afobazole (Isch + Afob) (n > 12). Data were normalized to cell death observed with media alone (Control). Asterisks denote significant difference between Control and Isch within Media and Afob, respectively (p < 0.001 for both); pound symbol indicates significant difference between Media and Afob within Control and Ischemia, respectively (p < 0.05 for Control & < 0.001 for Isch).

114

following ischemia. Afobazole alone showed a slight reduction in Bax levels but this was not significant. When afobazole is co-applied with ischemia, Bax expression was reduced by ~ 10%, however this was not significant. Thus, unlike the case in microglia, σ receptor activation fails to significantly block ischemia-induced upregulation of the pro- apoptotic gene, Bax, in neurons.

Figure 5.6

Afobazole fails to prevent upregulation of the pro-apoptotic gene, Bax, observed in response to extended ischemia in neurons A, Photomicrographs of merged images of cultured neurons exposed for 24 hr to media alone (Control) (i), or media containing 4 mM Azide (ii), 100 µM afobazole (Afob) (iii) or 4 mM Azide + 100 µM afobazole (Isch + Afob) (iv). Neurons were double labeled with DAPI (blue) and anti-Bax antibody (green). Fields of view shown are representative regions within larger images used to calculate Bax expression. Scale bar in (i) is 10 µm. B, Bar graph of mean number of Bax-positive neurons in experiments in which the cells were exposed to the same conditions as A. Data are expressed as percent (± SEM) of total number of neurons. Asterisks denote difference between Control and Isch within Media and Afob, respectively (p < 0.001) (n = 9). 115

We further examined how afobazole effects the levels of the death protease, caspase-3, in neurons after an ischemic insult. Following ischemia, caspase-3 expression was observed in a greater number of cells relative to control (Figure 5.7).

Figure 5.7

Afobazole fails to reduce expression of caspase-3 in response to prolonged ischemia in neurons. A, Photomicrographs of merged images of cultured neurons exposed for 24 hr to media alone (Control) (i), or media containing 4 mM Azide (Isch) (ii), 100 µM afobazole (Afob) (iii) or 4 mM Azide + 100 µM afobazole (Isch + Afob) (iv). Neurons were double labeled with DAPI (blue) and anti-active caspase-3 antibody (green). Fields of view of each photomicrograph are representative regions within larger image used to calculate caspase-3 expression. Scale bar in (i) is 10 µm. B, Bar graph of mean percent of caspase-3-positive neurons in experiments in which the cells were exposed to conditions identical to A. Data are expressed as percent (± SEM) of total number of neurons. Asterisks denote significant difference between Control and Isch within Media and Afob groups (p < 0.001 for both); pound symbol indicates significant difference between Media and Afob within Isch (p < 0.05) (n = 9).

116

Afobazole alone decreased caspase-3 levels relative to control, but failed to diminished the increase in caspase-3 levels evoked by ischemia. Examining the number of cells showing caspase-3 signal demonstrates that afobazole reduces the number of caspase-

3 positive cells by 38 ± 16% in the absence of ischemia. Following ischemia, afobazole

Figure 5.8

Afobazole alone upregulates the anti-apoptotic gene, Bcl-2, but does not enhance Bcl-2 levels following 24 hr ischemic incubation in neurons. Neurons were double labeled with DAPI (blue) and anti-Bcl-2 antibody followed by Alexa Fluor 488 conjugated secondary antibody (green). A, Photomicrographs of merged images of cultured neurons exposed for 24 hr to media alone (i) or media containing 4 mM Azide (ii, Isch), 100 µM afobazole (iii) or 4 mM Azide + 100 µM afobazole (iv). Areas shown are representative regions within larger images used to calculate Bcl-2 expression. Scale bar in (i) is 10 µm. B, Bar graph of mean percent of Bcl-2-positive neurons in experiments in which the cells were exposed to conditions identical to A. Data are expressed as percent (± SEM) of total number of neurons. Asterisk denote significant difference between Control and Isch within Media and Afob, respectively (p < 0.05 & < 0.01) (n = 9). 117

reduced caspase-3 levels by ~14%, unlike microglia levels of caspase-3, which were significantly different than the control, neuronal levels where similar to those observed under ischemic conditions.

We wanted to see if the relationship between afobazole and the expression of the anti-apoptotic gene, Bcl-2, was similar to what we saw in microglia after ischemia incubation. Following ischemia, the levels of Bcl-2 expressed by the neurons decreased by ~20% compared to that of control levels (Figure 5.8). Afobazole alone produced a

Figure 5.9

Afobazole prolongs neuronal survival but fails to preserve function after prolonged ischemia. A, bar graph of mean change in depolarization-induced increase in peak 2+ + [Ca ]i, evoked by application of high K (30 mM KCl) (±S.E.M.) in isolated cortical neurons incubated for 72 hr in media alone (Media) or media containing 4mM azide + 100µM afobazole (Isch + Afob). B, in experiments are identical to (A) shows the total net calcium influx. For each condition, n ≥ 79. Asterisk denotes significant difference between Media and Isch + Afob (p < 0.001).

118

~10% increase in the number of cells expressing Bcl-2. However, afobazole failed to rescue Bcl-2 expression following ischemia. Both ischemia and afobazole with ischemia treated neurons showed a statistically significant decrease in the expression of Bcl-2 by

21 ± 3 and 36 ± 8%, respectively.

The failure of σ receptors to affect Bax, caspase 3 and Bcl-2 in neurons following ischemia suggests that a distinct pathway may be involved in σ receptor protection of neurons and microglia. However, a second possibility is that these neurons are significantly injured and will eventually undergo programmed cell death. Thus, experiments were carried out to determine if the surviving neurons were functionally compromised, and thus likely to proceed towards apoptosis. Fluorometric calcium imaging experiment were carried out to determine if surviving cortical neurons showed functional responses consistent with membrane depolarizations. Neurons incubated under ischemic conditions in the absence and presence of 100 µM afobazole for 72 hrs followed by focal application of 30mM KCl failed to have any response compared to control neurons. These data suggest that the surviving neurons are not depolarizing and are thus functionally compromised (Figure 5.9).

Discussion

The novel findings reported in this study are that the σ receptor agonist, afobazole, rescues microglia from ischemia-induced cell death through mechanisms involving the regulated expression of apoptosis-regulating proteins. Subsequently, afobazole provides neuroprotection against ischemia-induced toxicity in cortical neurons but not through changes in the expression of these proteins. This decrease in microglial

119

cell death is likely due to a reduction in the expression of the pro-apoptotic gene, Bax

and the death protease, caspase-3. Further reduction in microglial cell death likely

results from the preserved Bcl-2 expression caused by afobazole. In contrast, afobazole

failed to inhibit the pro-apoptotic signaling induced by ischemia as the pro-apoptotic gene

product, Bax and the death protease, caspase-3, were not depressed following

afobazole application nor was the ischemia-induced downregulation of the anti-apoptotic

protein, Bcl-2 prevented. These findings suggest that activation of σ receptors differentially affects survival mechanisms in neurons and microglia. Furthermore, treatment with the sigma receptor agonist, afobazole did not preserve neuronal function after prolonged ischemia.

It has been suggested that over-activation of microglia result in neuronal death

(Hellwig et al., 2013). However, the need to maintain viable microglia that respond to sites of injury are equally necessary. Neurons exposed to ischemic conditions release cytotoxic and neurotropic factors that both activate and attract microglia to the area of injury (Hellwig et al., 2013, Kierdorf and Prinz, 2013). Unfortunately, in disease states like stroke, over-activation of microglia is a common occurrence and often times result in

neuronal and microglial cell death. Thus it is of great importance to identify factors that

contribute to the demise of microglia and find treatments to combat these mechanisms.

Exposure of microglia cells to ischemia resulted in approximately 150-fold increase in cell death from control alone. Afobazole reduced the number of dyeing microglia by 75 ±

8% when co-incubated with ischemia.

Our laboratory has previously shown that afobazole can mitigate intracellular calcium dyshomeostasis in neurons and microglia following ischemia, acidosis and ATP 120

exposure (Katnik et al., 2006, Herrera et al., 2008, Cuevas et al., 2011a, Cuevas et al.,

2011b). A rise in intracellular concentration, if unresolved, can promote the initiation of apoptosis (Mattson and Chan, 2003a). In addition to regulating intracellular calcium homeostasis, our findings show that afobazole mitigates ischemia-evoked apoptosis of microglia by decreasing expression of the pro-apoptotic gene, Bax, and death protease caspase-3 while preserving the levels of the anti-apoptotic gene, Bcl-2. Experiments were conducted to ascertain the mechanism by which afobazole mitigates microglia cell death following ischemia. Following a stroke, cells in the core undergo necrosis and in the days and weeks following the insult if the penumbra is not rescued from undergoing apoptosis, it too will die and become part of the core (Butler et al., 2002). Previous studies have shown that activation of σ-1 receptors protect RGC-5 cells from apoptosis by regulating the expression of the proapoptotic protein, Bax and death protease, caspase-3 (Tchedre and Yorio, 2008). We carried out immunocytochemical experiments to determine if afobazole affects expression of the pro-apoptotic protein Bax or the death caspase-3 following 24 hr ischemic exposure. Both Bax and caspase-3 expression increased > 270 and 150% from the control, respectively. We found that afobazole alone decreased caspase-3 expression and when co-incubated with ischemia both Bax and caspase-3 expression were mitigated approximately 42 and 59%, respectively. Further experiments were conducted to determine if levels of Bcl-2 were affected by ischemia and to what if affect afobazole had on its expression. Microglia cells were incubated for

24 hr in the absence (Control) and presents of ischemia, with and with afobazole.

Following ischemia incubation, microglial Bcl-2 expression was decreased by ~ 21% from that of the control. Incubation with afobazole alone increased Bcl-2 expression by a 121

modest but significant 10%. However, when co-incubated with ischemia, afobazole

preserved Bcl-2 expression by approximately 24%, making it comparable to afobazole

alone.

Following suit, we wanted to know if afobazole conveyed neuroprotection in a

similar mechanism as shown previously with microglia. Immunocytochemical

experiments were carried out to determine if one mechanism by which afobazole is

neuroprotective is through sigma receptor regulation of apoptotic expression. Both Bax

and caspase-3 expression were observed under all conditions. Following ischemic

exposure, both Bax and caspase-3 levels increased by ~ 47 and 135%, respectively.

However, unlike the results we observed in microglia cells, afobazole did not mitigate

increases in Bax or caspase-3 expression in neurons. The ratio of Bcl-2:Bax has been shown to positively correlate to cell survival in neurons exposed to Aβ (Patel and Brewer,

2008). Likewise, the ratio of Bcl-2:Bax is important in regulating cell survival following

glutamate excitotoxicity (Schelman et al., 2004) and previous studies have shown that

Bcl-2 is a target for σ receptor mediated neuroprotection (Yang et al., 2007, Meunier and

Hayashi, 2010). Additionally, experiments were performed to determine if afobazole could preserve expression of Bcl-2 levels following ischemia. As we observed with

microglia, Bcl-2 expression decreased in neurons under ischemic conditions. We found

that afobazole alone increased Bcl-2 expression; however, when co-incubated with

ischemia, afobazole failed to preserve Bcl-2 expression levels and were comparable to ischemia alone. Exposing cells to ischemic conditions has been shown to decrease cell viability. Our laboratory has shown that afobazole mitigates death following 2 hr ischemia

exposure (Cuevas et al., 2011a, Cuevas et al., 2011b). This model resembles injury 122

caused by reperfusion of the tissues with oxygen, more of a transient ischemic model.

We wanted to see to what extent afobazole could be neuroprotective following 72 hr

exposure to ischemic conditions, more a permanent occlusion model. There was an

approximant 3-fold increase in the number of expired neurons from that of the control.

This increase in the demise of neurons was mitigated by approximately 55% when

neurons were co-incubated with afobazole and ischemia. Incidentally, afobazole alone reduced neuronal cell death from control levels by ~ 49%. This data indicates that even under prolonged ischemic conditions afobazole was to some extent neuroprotective.

However, when functional integrity of these surviving neurons was examined by attempting to depolarize them with a focal application of 30mM KCl, we found them unresponsive.

In conclusion, our study demonstrates that afobazole reduce microglial toxicity and cell death following ischemic exposure. This cytoprotection most likely involves regulation of multiple genes, with a noted decrease in Bax and caspase-3 and a concomitant increase in Bcl-2 expression. Furthermore, our experiments show that afobazole can decrease ischemia-evoked neuronal cell death. However, the molecular mechanisms mediating neuroprotection by afobazole following an ischemic insult appear to be different than with microglia and do not involve the downregulation of both Bax and activated caspase-3, nor preserving upregulation of Bcl-2 expression. Taken together our

data suggest that afobazole may prevent microglia from entering the activated state

associated with neurotoxicity following exposure to ischemia, and increasing the survival

of both microglia and neurons following an ischemic insult. These properties make

afobazole an attractive drug for treatment of ischemia. 123

CHAPTER 6

Afobazole Abolishes Amyloid-β25-35 Potentiation of Ischemia-Acidosis Evoked Increases in Neuronal Intracellular Calcium Levels

Introduction

In addition to large vessel ischemic strokes there are a subset known as lacunar infarcts, which account for approximately 25% of all ischemic insults (Samuelsson et al.,

1996). These strokes result from the blockage of small penetrating arteries to the subcortical regions of the brain (Kang et al., 2012). While the injury from these strokes were once thought to be negligible, lacunar infarcts can and do result in death and long-

term disability. This type infarct is a leading cause of cognitive impairment and vascular

dementia (Charidimou and Werring, 2012). While the risk factors resulting in lacunar

verse large artery infarcts differ, the resulting cellular cascade promoting cerebral cell

death may have overlapping mechanisms. One of these mechanisms may be the

increased production and toxicity of the amyloid-β peptide, which has been shown to be

upregulated following an ischemic event (Popa-Wagner et al., 1998, Guglielmotto et al.,

2+ 2009, Pluta et al., 2013). The presence of Aβ has been shown to increase [Ca ]i levels,

increase ROS production and promote apoptosis (Behensky et al., 2013a, b). In addition,

the accumulation of Aβ can contribute to vascular dementia, which has been shown to

significantly increase the chances of having a recurrent stroke (Pendlebury and Rothwell,

2009). Furthermore, this Aβ-induced arterial weakening may potentiate the chances of 124

having a hemorrhagic stroke significantly increasing the patient’s chance of developing serious long-term disability or even death (Sudduth et al., 2013, Sudduth et al., 2014).

The mechanisms that contribute to the injury of both lacunar and large vessel ischemia are still unclear. However, the activation of NMDA receptors is known to contribute, in part, to both of these types of strokes (Shih et al., 2013). This brings into question the prospect that even though lacunar and large-vessel strokes occur via different vascular pathologies the mechanisms promoting their injury may overlap. Data from our laboratory has shown that activation of sigma receptors mitigates ischemia, acidosis and Aβ-induced calcium overload (Katnik et al., 2006, Herrera et al., 2008,

Behensky et al., 2013a, b). This calcium overload if left unchecked will promote apoptosis through pathways including NMDA receptor activation. Our laboratory has also shown that acid-sensing ion channel 1a (ASIC1a) contributes to the downstream activity of NMDA receptors (Mari et al., 2014). Aβ levels increase following ischemia, which may lead to potentiated responses that increase the risk for various vascular dementias and ultimately result increased cell death. Currently, the mechanisms contributing to this interaction remain unknown.

Experiments were carried out to determine the effects of Aβ on ischemia + acidosis evoked elevations in intracellular Ca2+ overload in cortical neurons. Aβ incubation was found to significantly potentiate the ischemia + acidosis induced

2+ increases in [Ca ]i but required 24 hr incubation before affects were seen, indicating that non-acute mechanisms are involved. Furthermore, afobazole co-incubation was found to

2+ 2+ abolish the Aβ-induce intracellular Ca increase. The kinetics of the [Ca ]i overload produced by ischemia + acidosis following pre-incubation in Aβ were indicative of acid- 125

sensing ion channels 1a (ASIC1a) involvement. In conclusion, our data show a

2+ mechanism by which Aβ may enhance neuronal injury during ischemia, increased [Ca ]i

overload, and identify ASIC1a as the channel mediating this phenomenon. Finally, our

data suggest that activation of σ receptors may represent a therapeutic strategy for mitigating injury produced by Aβ and stroke.

Materials and Methods

Preparation of Rat Cortical Neurons

All experiments were carried out on cultured cortical neurons from mixed sex

embryonic day 18 (E18) Sprague-Dawley rats (Harlan, Indianapolis, IN). Methods used

here were identical to those previously reported for the isolation and culturing of these

cells (Katnik et al., 2006). Cortical neurons were plated on poly-L-lysine coated

coverslips and cultured in B27 and L-glutamine supplemented Neurobasal medium

(Neurobasal Complete) (Life Technologies, Grand Island, NY). Cells were used for experiments after 10-21 days in culture, which permits synaptic contact formation and yields robust responses to other pathophysiological conditions such as in vitro ischemia and acidosis (Katnik et al., 2006, Herrera et al., 2008). Animals were cared for in accordance with the regulations and guidelines set forth by the University of South

Florida’s College of Medicine Institution on Animal Care and Use Committee.

Calcium Imaging

Intracellular Ca2+ concentrations were measured in isolated cortical neurons using

ratiometric fluorometry as previously described (Katnik et al., 2006, Cuevas et al., 2011a,

126

Cuevas et al., 2011b). For most experiments, cultured neurons were incubated in 25 µM

2+ Aβ25-35 for 24 hr prior to [Ca ]i being measured. Neurons were loaded with fura-2 by adding 3-5 µg/ml of the acetoxymethylester form of fura-2 (fura-2 AM) in 0.3% DMSO to the media and incubating the cells at room temperature for 1 hr. Prior to beginning the experiments, the coverslips were rinsed with physiological saline solution (PSS) consisting of (in mM): 140 NaCl, 5.4 KCl, 25 HEPES, 20 glucose, 1.3 CaCl2, and 1.0

2+ MgCl2 (pH to 7.4 with NaOH). For neurons, [Ca ]i was measured once every second for approximately a 4 min period and the ischemia-acidosis induced values for baseline,

2+ peak, area and slope (± SEM) [Ca ]i was calculated.

Electrical Recordings

Proton-activated membrane currents were recorded using the protocol previously described by our laboratory (Herrera et al., 2008). Briefly, neurons plated on glass coverslips were transferred to a recording chamber and membrane currents were amplified, filtered at 1 kHz, digitized at 5 kHz, and acquired using Clampex 9 (Axon), a

MultiClamp 700A amplifier and a Digidata 1322A. Electrical access was achieved using the amphotericin B perforated patch method to preserve intracellular integrity of neurons

(Rae et al., 1991). An amphotericin B stock solution (60 mg/ml in DMSO) was made fresh daily and diluted to 240 μg/ml (0.4% DMSO) in control pipette solution immediately prior to seal formation. The control pipette solution consisted of (in mM): 75 K2SO4, 55

KCl, 5 MgSO4, and 10 HEPES (titrated to pH 7.2 with N-methyl-d-glutamine). Patch electrodes were pulled from thin-walled borosilicate glass (World Precision Instruments

Inc., Sarasota, FL) using a Sutter Instruments P-87 pipette puller (Novato, CA) and had

127

resistances of 1.0–1.5 MΩ. Access resistance (Rs) was monitored throughout experiments for stable values ≤ 30 MΩ and were compensated at 40% (lag, 10 μs). In all electrical recordings, cells were voltage-clamped at −70 mV.

Compounds, Reagents and Peptides

The following compounds and reagents were used in this investigation: Amyloid-β fragments 25-35 (62-0-72), 35-25 (62-0-33) 1-40 (62-0-78) and 1-42 (62-0-30)

(American Peptides, Sunnyvale, CA); Tetrodotoxin and YM244769 (Tocris Biosciences,

Ellisville, MO); fura-2 acetoxymethyl ester (Life Technologies); Psalmotoxin-1

(ab120483) (abcam, Cambridge, MA. Vehicle for drugs and reagents will be either water

(H2O) or dimethyl sulfoxide (DMSO), and appropriate vehicle controls will be carried out for each study. For the Aβ oligomerization experiment, Aβ25-35 was made to stock concentration (1mM) and allowed to oligomerize for 24 hr at 37º C prior to cell incubation. Afobazole was generously provided by IBC Generium (Moscow, Russian

Federation).

Data Analysis

Fura-2 fluorescence intensities as a function of time were recorded in the same manner as previously described (Katnik et al., 2006, Cuevas et al., 2011a, Cuevas et al.,

2+ 2011b) and converted to [Ca ]i using control values obtained with an in situ calibration kit (Invitrogen). Electrophysiology data was converted to text files and imported into

Clampfit 9 for analysis. Data was analyzed using SigmaPlot 11 (Systat Software, Inc.,

San Jose, CA). Data points represent peak means ± standard error of the mean

(S.E.M.). Statistical analysis involved the use of paired or unpaired t-tests, as 128

appropriate, and one- or two-way analysis of variance for determining significant

differences for multiple group comparisons, followed by post hoc analysis using the

Holm-Sidak test. Results were considered statistically significant if p < 0.05.

Results

Experiments were carried out to determine if incubation of 25 µM amyloid-β25-35

2+ affects the elevations in [Ca ]i in response to ischemia, acidosis and ischemia + acidosis

2+ application in rat cortical neurons. Figure 6.1A-C shows representative traces of [Ca ]i

as a function of time recorded from two cells during focal application of ischemia,

2+ acidosis or ischemia + acidosis that produced robust elevations in [Ca ]i in the neurons, consistent with previous reports (Katnik et al., 2006, Herrera et al., 2008, Mari et al.,

2014). Following 12 hour incubation in media with 25 µM amyloid-β25-35, neurons were

exposed to a focal application of ischemia, acidosis or ischemia + acidosis, which

2+ produced increases in [Ca ]i levels similar to the control. Quantification of these calcium

increases showed that 12 hr of Aβ incubation did not increase the peak calcium levels.

However, there was a small but significant increase in the total net calcium influx evoked

from all three conditions (Figure 6.1D).

Given that Aβ modulation of these responses may require a longer incubation time,

neurons were exposed to applications of ischemia, acidosis or ischemia + acidosis

following 24 hr incubation in media alone, or media containing 25 µM of Aβ25-35. Figure

2+ 6.2A-C shows representative traces of [Ca ]i as a function of time recorded from two

cells during focal application of ischemia, acidosis or ischemia + acidosis that produced

2+ robust elevations in [Ca ]i in the neurons. Calcium increases in the Aβ-treated group

129

Figure 6.1

Incubation for 12 hr with amyloid-β25-35 does not potentiate intracellular calcium responses to focal ischemia, acidosis or ischemia + acidosis application. A-C, show 2+ representative traces of [Ca ]i as a function of time recorded from two neurons during application of ischemia, acidosis and ischemia + acidosis following 12 hr incubation in media alone (Control), or media containing 25 µM Aβ25-35 (Aβ), respectively. Aβ was 2+ washed out for 1hr prior to focal application. Line above [Ca ]i responses indicates duration of ischemia, acidosis or ischemia + acidosis application. D, Bar graph of mean 2+ (± SEM) peak and total net increases in [Ca ]i evoked by focal applications following 12 2+ hr Aβ in isolated cortical neurons. [Ca ]i responses were recorded from neurons in experiments identical to those performed in (A-C). Asterisk symbolizes significant difference between Control and Aβ (p < 0.05). For all conditions, n ≥ 74.

were significantly higher than the respective controls. When quantified, Aβ evoked a

significant increase in both the peak and the total net calcium influx following ischemia by

130

Figure 6.2

24 hr incubation with amyloid-β25-35 significantly potentiates the intracellular calcium responses to ischemia, acidosis and ischemia + acidosis application. A-C, show 2+ representative traces of [Ca ]i as a function of time recorded from two neurons during application of ischemia, acidosis and ischemia + acidosis following 24 hr incubation in media alone (Control), or media containing 25 µM Aβ25-35 (Aβ), respectively. Aβ was 2+ washed out for 1hr prior to focal application. Line above [Ca ]i responses indicates duration of ischemia, acidosis or ischemia + acidosis application. D, Bar graph of mean 2+ (± SEM) peak and total net increases in [Ca ]i evoked by focal applications following 24 2+ hr Aβ in isolated cortical neurons. [Ca ]i responses were recorded from neurons in experiments identical to those performed in (A-C). Asterisk symbolizes significant difference between Control and Aβ (p < 0.001). For all conditions, n ≥ 81.

131

72 ± 6 and 82 ± 6%, acidosis 42 ± 7 and 33 ± 10% and ischemia + acidosis 43 ± 4 and

85 ± 2% applications, respectively (Figure 6.2D).

Amyloid-β peptide has been shown to induce toxicity in several disease states including AD and cerebral amyloid angiopathy through a number of mechanisms including calcium toxicity, reactive species and hemichannel formation to name a few

(Mantha et al., 2006, Rezende et al., 2007, Orellana et al., 2011, Lazzari et al., 2015). To ensure that the fragment Aβ25-35 shows similar toxicity to other more researched

Figure 6.3

2+ Amyloid-β fragments 25-35 and 1-42 potentiate ischemia + acidosis induce [Ca ]i increases. A, shows a bar graph of mean peak (± SEM) of neurons incubated for 24 hr in 25µM Amyloid-β, either fragment 25-35, 1-40 or 1-42. Asterisk indicates a significant difference from the Control, whereas the pound symbol shows significance from fragment 1-40. B, is a bar graph of the mean total net Ca2+ influx (± SEM) in experiments identical to A. Asterisk indicates a significant difference from the Control, whereas the pound symbol shows significance from fragment 1-42 and 25-35 or 1-40 (p < 0.001 for all) (n ≥ 158). 132

fragments, experiments were conducted to assess various Aβ fragments effects on

2+ potentiating increases in [Ca ]i evoked by ischemia + acidosis application. Figure 6.3 shows a bar graph of the peak and total net calcium influx-induced by ischemia + acidosis application following 24 hr incubation in media with Aβ fragments 25-35, 1-40 or

1-42. While fragment 1-40 showed no significance from the control, both fragment 25-35

2+ and 1-42 showed a potentiation in the peak [Ca ]i induced by ischemia + acidosis by 65

2+ ± 1 and 51 ± 9%, respectively. This potentiation in peak [Ca ]i level by the Aβ fragments

25-35 and 1-42 were not significantly different from each other. Moreover, pre-incubation with these fragments resulted in an increase in the total net calcium influx by 19 ± 2 and

37 ± 3% following focal application of ischemia + acidosis.

Figure 6.4

Oligomerized amyloid-β25-35 fails to potentiate ischemia + acidosis evoked increases in 2+ 2+ [Ca ]i following a 1 hr incubation. Bar graph of the mean peak and total net Ca influx (±SEM) in neurons incubated for 1 hr in media alone or media (Control) with 25 µM oligomerized Aβ25-35 (Aβ) (n ≥ 43). 133

Even though Aβ has been shown to bind directly to various ion channels including

NMDA and nAChRs (Nagele et al., 2002, Texido et al., 2011) it has been suggested that

2+ the effects seen on Aβ-potentiated increases in [Ca ]i be due to the oligomerized and not the free Aβ. Therefore, experiments were conducted to determine if 1 hr incubation

2+ with the oligomerized Aβ25-35 produced significant increases in [Ca ]i following ischemia

+ acidosis application. Figure 6.4 shows a bar graph of the peak and total net calcium influx in neurons incubated in media alone or media with the oligomerized Aβ. Following application of ischemia + acidosis there was no significant difference between the media

2+ or Aβ incubated increases in [Ca ]i levels.

Given that afobazole has been shown to reduced ischemia and acidosis evoked

2+ elevations in [Ca ]i (Cuevas et al., 2011a) and that σ-1 receptor activation has been shown to reduce Aβ-induced toxicity (Marrazzo et al., 2005), experiments were

2+ conducted to determine if afobazole could mitigate the Aβ potentiated increase in [Ca ]i following ischemia + acidosis application. Figure 6.5A shows representative traces of

2+ [Ca ]i as a function of time recorded from 3 cells during ischemia + acidosis. Increases

2+ in [Ca ]i were observed in all groups with a potentiated increase in the Aβ incubated cells with responses similar to those observed in experiments from Figure 6.2C. In cells

2+ were afobazole was co-incubated with Aβ, [Ca ]i elevations were not statistically different from the control (Figure 6.5B&C), indicating that afobazole blocked the Aβ-

2+ induced potentiation of [Ca ]i overload.

2+ The kinetics of the [Ca ]i overload produced by ischemia + acidosis following pre- incubation with Aβ suggest that the acid-sensing ion channels 1a (ASIC1a) is involved in the potentiation. To determine if Aβ is potentiating ASIC or only downstream effector 134

targets of these channels, patch-clamp electrophysiological recordings were carried out.

Figure 6.6A shows proton-evoked membrane currents recorded from two neurons

Figure 6.5

2+ Afobazole inhibits increases in [Ca ]i evoked by ischemia + acidosis application 2+ following 24hr Aβ25-35 incubation. A, Representative traces of [Ca ]i as a function of time recorded from three neurons during focal application of ischemia + acidosis in the absence (Control) and presence of 25µM amyloid-β25-35 (Aβ) or 25µM amyloid-β25-35 and 100 µM afobazole (Afob + Aβ). Afobazole was co-incubated with Aβ for 24 hr and washed out 1 hr prior to Ischemia + acidosis application. In experiments identical to A, bar graphs of mean peak and total net Ca2+ influx (±SEM) measured in neurons. Asterisks denote significant difference between Aβ and Control or Aβ + Afob (p < 0.001) (B & C) (n ≥ 137).

135

following 24 hr incubation in media alone or media with 25 µM Aβ25-35 held at a

membrane potential of -70 mV. Application of protons evoked a rapid inward current

which was potentiated by 68 ± 17% in Aβ incubated neurons. Varying pH showed that

Aβ potentiated responses at both pH 6.5 and pH 6.0, but not pH 7.0 (Figure 6.6B).

These data suggest that Aβ is potentiating the responses rather than shifting the half maximal activation of ASIC to a higher pH.

2+ The elevations of [Ca ]i evoked by protons in cortical neurons are due to

activation of ASIC, but these channels themselves account for only a small portion of the

2+ increase in [Ca ]i observed during acidosis (Herrera et al., 2008). One possible

2+ contributor to Aβ increases in [Ca ]i that are downstream of ASIC are the ion

Figure 6.6

Amyloid-β25-35 incubation potentiates proton-evoked currents in cortical neurons. A, Representative current traces recorded from two neurons held at -70 mV during acidosis in neurons incubated in media alone (Control) or media with 25µM Amyloid-β25-35 (Aβ). B, Bar graph of mean peak (±SEM) of the proton-evoked inward current in experiments identical to A (n ≥ 8). Asterisk denotes significant difference between Aβ and respective controls (p < 0.05).

136

exchangers. Thus, calcium imaging experiments were conducted to determine if reversal of the sodium/calcium exchanger (NCX) contributed to the calcium overload evoked by

2+ Aβ incubation. Figure 6.7 is a bar graph of the mean peak and area of [Ca ]i elevations evoked by focal application of ischemia + acidosis following 24 hr incubation in media alone or media with 25 µM Aβ. Similar to previous experiments, incubation with Aβ

2+ produced a potentiated increase in the [Ca ]i levels. Following the initial recording, neurons were incubated with the NCX reversal mode inhibitor YM244769 (YM,1µM) for

Figure 6.7

Aβ potentiated calcium influx involves reversal of the sodium/calcium exchanger. A, Bar 2+ 2+ graphs of mean (±SEM) increases in peak [Ca ]i or net [Ca ]i (Area, area under the trace) elicited by focal ischemia + acidosis application in neurons incubated for 24 hr in media alone (Media) or media with 25 µM amyloid-β25-35 (Aβ), before (Control) and after 20 min pre-incubation with YM224769 (YM, 1µM). Asterisks denote significant difference between Control and YM within Media and Aβ groups (p < 0.001). Pound symbol indicates significant difference between Controls within Media and Aβ groups (p < 2+ 0.001). B, Bar graph of relative YM inhibition of [Ca ]i. Asterisks denote significant difference (p <0.01). For all groups, n ≥ 171.

137

20 minutes. YM incubation produced a significantly reduced increase in the ischemia +

2+ acidosis evoked [Ca ]i peak and net elevations by 38 ± 2 and 36 ± 2%, respectively.

This YM-induced reduction in calcium influx through the reversal NCX was also observed

in Aβ incubated neurons, which lead to a 40 ± 2 and 44 ± 1% reduction in the peak and

2+ net Aβ-potentiated increase in [Ca ]i, respectively. This resulted in relative inhibition of

YM blocking a small but significant 3.2 ± 0.7 and 10 ± 1.8% influx in the peak and total net calcium potentiated by Aβ incubation, respectively. While blocking the reversal NCX in neurons that were incubated in Aβ reduced the total calcium influx, we cannot yet be sure that Aβ is having a direct effect on NCX or if it is an upstream mechanism(s), which is increasing the sodium loading and thus increasing the ion concentration gradient.

Continuing to assess the role of ion exchangers in the Aβ potentiated increase in

2+ [Ca ]i elevations following focal application of ischemia + acidosis, we decided to look at

the sodium/calcium potassium-dependent exchanger (NCKX). Experiments were

conducted following a 24 hr incubation in media or media with 25 µM Aβ25-35, using a physiological saline solution (PSS) with varying concentrations of K+ in attempts to

2+ assess the role of NCKX. Figure 6.8A shows representative traces of [Ca ]i as a

function of time recorded from 2 cells during ischemia + acidosis. Responses under

control conditions (K+=5.4mM), were identical to previous experiments for both media

and Aβ incubated neurons with Aβ having statistically higher peak and total net calcium

influx then media. However, when we changed the PSS to low or high K+ (2.7 & 10.8

2+ mM, respectively) the ischemia + acidosis induced elevations in [Ca ]i levels were not

as pronounced. Furthermore, there was no longer a significant difference between the

138

media and Aβ incubated neurons. Indicating to us that the NCKX may not be contributing

to the potentiation we observe in Aβ treated neurons (Figure 6.8B&C).

Figure 6.8

2+ Amyloid-β25-35 potentiation of [Ca ]i increases evoked by ischemia + acidosis is not dependent of the sodium/calcium potassium-dependent exchanger (NCKX). A, representative traces of focal application of ischemia + acidosis recorded from two neurons following 24 hr incubation in media (Media) or media with 25 µM Aβ25-35 (Aβ). Neuron were pre-incubated for 10 min in PSS with varying concentrations of K+ (in mM, 2+ normal = 5.4, low = 2.7, high = 10.8). B, bar graph of mean change in peak [Ca ]i from experiments identical to A. Asterisk identifies significance Control and Aβ within 5.4 (p < 0.001). Pound represents significance between Control or Aβ within groups 5.4, 2.7 and 10.8 (Control p < 0.01, Aβ p < 0.001). Dagger denotes significance between Aβ within 2+ 2.7 and 10.8. C, bar graph of mean net [Ca ]i from experiments identical to A. Asterisk identifies significance between Control and Aβ within 5.4 (p < 0.001). Pound represents significance between Control or Aβ within groups 5.4, 2.7 and 10.8 (both p < 0.001). Dagger denotes significance between Control within 2.7 and 10.8 (n ≥ 80).

139

Figure 6.9

2+ Amyloid-β25-35 potentiation of the net [Ca ]i increases evoked by ischemia + acidosis may involve intracellular protons. A, representative traces of focal application of ischemia + acidosis recorded from two neurons following 24 hr incubation in media (Media) or media with 25 µM Aβ25-35 (Aβ). Neuron were pre-incubated for 10 min in PSS without - - 2+ (Control) and with 26 mM HCO3 (HCO3 ). B, bar graph of mean change in peak [Ca ]i - from experiments identical to A. Asterisk identifies significance Control and HCO3 within Media and Aβ groups (p < 0.001). Pound represents significance between Control or - HCO3 within groups Media and Aβ (Peak p < 0.05, Net p < 0.001). C, bar graph of - 2+ relative HCO3 inhibition of [Ca ]i from experiments identical to A. Asterisk identifies significance between Media and Aβ within Area (p < 0.001) (n ≥ 66).

Since following an ischemic event cells undergo acidosis we were interested in looking at the roles of proton exchangers, specifically the sodium/hydrogen exchanger

140

(NHE) and the mitochondrial calcium/hydrogen exchanger (LETM1). To achieve this we

- used PSS with 26 mM HCO3 to sequester the intracellular protons. Prior to recording

neurons were incubated for 24 hr in media alone or media with 25 µM Aβ25-35. Figure

2+ 6.8A shows representative traces of [Ca ]i as a function of time recorded from 2 cells

- during ischemia + acidosis before and after 10 min incubation in HCO3 PSS. As we have previously seen in identical experiments Aβ incubation potentiates both the peak and total net calcium influx following ischemia + acidosis application. Pre-incubation with

- HCO3 reduced the media and Aβ incubated peak by 36 ± 4 and 31 ± 3%, as well as the

total net calcium influx by 49 ± 1 and 34 ± 1%, respectively (Figure 6.8B). When

- comparing the relative inhibitions that HCO3 had between the media and Aβ treated

cells we found no significance in the peak levels, however there was a 30 ± 2% reduction

in the total net calcium influx of media incubated neurons (Figure 6.8C). While still

unclear, intracellular protons may be contributing to the calcium overload, possibly

through the aforementioned exchangers.

Discussion

The major findings reported in the present study are that amyloid-β potentiates

2+ ischemia + acidosis evoked increases in [Ca ]i in a time-dependent manner and through an acid-sensing ion channel 1a-dependent mechanism. The responses were also dependent on the use of amyloid-β fragments 25-35 or 1-42, whereas the fragment 1-40 did not potentiate the increase in intracellular calcium. Moreover, co-incubation with the pan-selective σ receptor agonist, afobazole, completely abolished this potentiation.

141

Neuronal cultures were used to determine if incubation with 25 µM amyloid-β25-35

2+ produced an increase in the [Ca ]i levels evoked by ischemia + acidosis application.

Incubation for 12 hr failed to potentiate Ca2+ elevations. It has been shown that longer

periods are needed (i.e. 24 hr) to up-regulate gene expression (Behensky et al., 2013a,

b). Therefore, neurons were incubated for 24 hr with Aβ and found to have potentiated

2+ ischemia, acidosis and ischemia + acidosis induced increases in [Ca ]i levels. We found

2+ that acidosis application alone produced an increase in mean peak [Ca ]i by ~42%.

Furthermore, application of ischemia alone or ischemia + acidosis also produced

potentiated elevations in both the peak and total net calcium influx by ~72 and 82% for

ischemia and ~43 and 85% for ischemia + acidosis, respectively. This dual potentiation

2+ in [Ca ]i may indicated that the mechanism in these response involve multiple pathways

including ASIC1a, VGCCs, NMDA and AMPA/kainate receptors. Previous studies from

2+ our laboratory have shown that ischemia-induced elevations in [Ca ]i involved the

activation of NMDA receptors (Katnik et al., 2006), whereas acidosis application

activates ASIC1a receptors (Herrera et al., 2008). Moreover, the ischemia + acidosis

2+ activation of NMDA and ACIS1a receptors results in a synergistic increase in [Ca ]i

levels. This is a complicated interaction with NMDA receptors functionally up-regulating

ASIC1a, while at the same time ASIC1a potentiates NMDA receptor activity (Mari et al.,

2014).

2+ Since activation of ASIC1a has been shown to potentiate ischemia-induced [Ca ]i

levels through NMDA receptors, we were interested in determining where Aβ potentiation

occurred. Previous studies have shown that only a small part of the ischemia + acidosis

evoked Ca2+ influx is directly due to ACIS1a with VGCCs, NMDA and AMPA/kainate 142

receptors contributing to the bulk influx (Gonzalez-Alvear and Werling, 1995, Alonso et

al., 2000). However, fluorometric calcium imaging showed that Aβ-induced a potentiated

proton activated Ca2+ influx. Therefore, whole-cell patch-clamp experiments were

performed to determine whether this potentiation was due to directly to ASIC1a activity

or via ASIC1a enhanced downstream channels. Neurons incubated with Aβ for 24 hr

were voltage-clamped at -70 mV, which prevents NMDA receptor and VGCC activation,

demonstrated a ~68% increase in the proton evoked inward current. Taken together, our

results indicate that Aβ incubation is directly up-regulating ASIC1a activity as well as the

ASIC1a potentiated down-stream channels.

2+ These channels contribute directly and indirectly to the [Ca ]i overload. Following

ischemia there is a loss of ATP production and consequently Na+ loading due to failure in

the Na+/K+ ATPase. This shift in the Na+ concentration gradient results in the reversal of

the Na+/Ca2+ exchanger, which can contribute to the Ca2+ overload. Imaging experiments

found that following Aβ incubation there was an increase NCX function, which was

blocked by the NCX (reversal) inhibitor YM244769 (1µM). However, we are not able to

say whether this increase in reversal NCX activity is the result of Aβ on NCX or due to

increased Na+ loading, which increases the driving force of the exchanger. An additional

consequence of the Na+/K+ ATPase failure is the accumulation of extracellular K+, which

can result in the depolarization of neighboring neurons. However, it can also increase the

activity of the Na+/Ca2+ K+-dependent exchanger (NCKX), which can further contribute to

2+ 2+ the [Ca ]i overload. To assess the role of NCKX on Aβ potentiated increase in [Ca ]i

evaluations following focal application of ischemia + acidosis experiments were

conducted using a physiological saline solution (PSS) with varying concentrations of K+. 143

Responses under control conditions (K+ = 5.4mM), were identical to previous

experiments for both media and Aβ incubated neurons with Aβ having statistically higher

peak and total net calcium influx then media. However, when we changed the PSS to

low or high K+ (2.7 & 10.8 mM, respectively) the ischemia + acidosis induced elevations

2+ in [Ca ]i levels were not as pronounced. Furthermore, there was no longer a significant

difference between the media and Aβ incubated neurons. Indicating to us that the NCKX

may not be contributing to the potentiation we observe in Aβ treated neurons.

Reducing the Ca2+ overload produced by an ischemic insult is crucial to mitigating

stroke injury. Previous studies from our laboratory have shown that σ-1 receptor activation in neurons can elevate the Ca2+ overload induced by ischemia, acidosis and

Aβ (Cuevas et al., 2011a, Behensky et al., 2013a). However, not all sigma receptor

agonists were shown to be as effective under these conditions (Behensky et al., 2013a).

The pan-selective agonist afobazole was one such drug that did mitigate Ca2+ overload

induced under all three conditions. Therefore, 100 µM afobazole was co-incubated with

Aβ for 24 hr to assess what function activation of sigma receptors would have on the Aβ

up-regulated function. Co-incubation with afobazole completely abolished the potentiated

2+ increases in [Ca ]i levels-induced via focal application of ischemia + acidosis. Reducing

the Aβ up-regulated Ca2+ increases will likely translate to an increase in neuronal

survival.

In conclusion, our data show one mechanism by which Aβ may potentiate

2+ neuronal injury during ischemia-acidosis is through increased [Ca ]i overload, and

identify ASIC1a as the channel mediating this phenomenon. Finally, our data suggest

144

that activation of σ receptors by afobazole may represent a therapeutic strategy for mitigating injury produced by Aβ and stroke.

145

CHAPTER 7

Discussion

Conclusions

Stroke is the fifth leading cause of death and continues to be a major cause of serious long-term disability in the United States (Mozaffarian et al., 2015). An ischemic

stroke accounts for about 87% of all strokes and occurs when blood flow to the brain is

occluded by a thrombus or embolus. The only treatment currently approved by the FDA

for ischemic stroke is recombinant tissue plasminogen activator (rtPA), a thrombolytic

agent used to restore blood-flow interrupted by a blood clot (Weintraub, 2006). However,

rtPA therapy is time sensitive, and the drug must be administered < 4.5 hours post-

infarct. In addition, aging patients presenting various comorbidities leads to an increase

in rtPA’s risk to benefit ratio, resulting in less than 4% of patients being able to use it

(Armstead et al., 2010). The goal of future therapies would be to treat these patients at

24 to 48 hrs follow the insult.

Our laboratory has shown that DTG can mitigate neuronal and glial cell injury

following in vitro stroke models through activation of σ receptors (Katnik et al., 2006, Hall

et al., 2009a). In attempts to improve upon the efficacy of DTG we conducted

experiments, which involved shifting and removal of the methyl moiety of DTG. Our

findings with the DTG analogues show that rearrangement of the methyl moiety did not

146

produce a compound which behaves differently from the parent compound in terms of

2+ modulating [Ca ]i increases induced via ischemia and acidosis. Removal of the group entirely (DPhG) produces a significant 18.3 ± 0.05% increase over o-DTG in inhibiting

2+ elevations in [Ca ]i during acidosis. Similarly, DPhG show a 12.2 ± 0.04% greater inhibition than o-DTG in the ischemia assay, but this difference was not statistically significant. Previous studies have reported that the σ receptor binding affinities for o-, m-, and p-DTG are approximately 30, 50, and 530 nM, respectively; whereas the affinity of

DPhG for σ receptors is ~400 nM (Scherz et al., 1990, Reddy et al., 1994). Results from our functional studies, therefore, do not reflect the decrease in affinity observed for the p-

DTG or DPhG. Calcium elevations evoked by ischemia and acidosis are in part due to activation of NMDA receptors (Katnik et al., 2006, Herrera et al., 2008). However, neither p-DTG nor DPhG shows increased affinity for NMDA receptors compared to o-DTG

(Scherz et al., 1990, Reddy et al., 1994), and thus actions on that receptor cannot account for the discrepancy between σ receptor binding and functional effects. Since the earlier studies did not examine specifically σ receptor subtype binding affinity, the difference may be due to changes in affinity for one receptor subtype versus the other

(i.e. σ-2 receptor affinity decreases significantly but σ-1 receptor affinity remains unchanged).

While migration and removal of the methyl moiety failed to alter functional properties of the molecule significantly, pronounced enhancement in potency were found with halide substitutions. Data presented in this study show that substituting a chloro for a methyl moiety in the meta and para positions of DTG produced compounds that had

147

~15% higher potency relative to the parent compound (o-DTG) at 100 µM concentration

2+ in the ischemia-induced increases in [Ca ]i assay. The enhancement in potency was

2+ even more pronounced in the acidosis-evoked [Ca ]i dysregulation assay. In this assay,

m- and p-ClDPhG (100 µM) produced blocks of >100% greater than o-DTG. In contrast,

substitution of a chloro moiety in the ortho position did not alter the functional effects of

the parent compound in either the ischemia or acidosis assay. Similar observations were

made using compounds with bromo substitutions (BrDPhG) of DTG in the meta and para

positions with an ~22% increase of inhibition during ischemia and an ~160% increase

during acidosis applications over o-DTG, respectively. The most potent compound tested

was p-BrDPhG, which had an IC50 indicating 8-fold and 34-fold greater potency over that

2+ 2+ of o-DTG in the acidosis [Ca ]i and ischemia [Ca ]i assays, respectively.

Functional assays indicated that p-BrDPhG was significantly more potent than o-

DTG, but a radioligand binding assay showed decreased affinity for both σ receptor

subtypes. Our experiments showed that o-DTG binds σ-1 and σ-2 receptors with an affinity in the range of 60-90 nM, which is similar to what was previously reported in the literature (Torrence-Campbell and Bowen, 1996). In contrast, p-BrDPhG was found to bind σ-1 and σ-2 receptors with 296 ± 46 nM and 800 ± 57 nM affinities, respectively, with the σ-2 receptor affinity being significantly lower. These values are similar to a previous report which indicates that p-BrDPhG binds guinea pig brain membrane extracts with an affinity of 540 ± 25 nM (Scherz et al., 1990). That study also demonstrated that p-BrDPhG has lower binding affinity for the NMDA receptor than the parent compound (o-DTG), and therefore, the increased functional potency cannot be explained by increased action at the NMDA receptor. 148

2+ Confirmation that p-BrDPhG mitigates [Ca ]i dysregulation via activation of σ-1

receptors was achieved via the use of the σ-1 receptor antagonist BD-1063. Using the

2+ acidosis [Ca ]i functional assay, BD-1063 was shown to inhibit the effects of p-BrDPhG

by >50% at a concentration of 10 nM, which is consistent with p-BrDPhG acting via σ-1

receptors (Matsumoto et al., 1995, Herrera et al., 2008). To date, only activation of the σ-

2+ 1 receptor has been shown to affect ischemia- and acidosis-evoked [Ca ]i dysregulation

(Katnik et al., 2006, Herrera et al., 2008), and thus effects by the σ-2 receptor were not

tested in these assays.

The current study shows that p-BrDPhG also inhibits microglial activation and this

DTG analogue is 4-fold more potent than the parent compound and a second pan-

selective σ agonist, afobazole (Hall et al., 2009a, Cuevas et al., 2011b). Unlike ischemia-

2+ and acidosis-evoked [Ca ]i dysregulation, which appears to be exclusively affected by σ-

1 receptor activation, both σ-1 and σ-2 receptors can modulate microglial migration

(Cuevas et al., 2011b). This observation may explain why p-BrDPhG, which has even lower affinity for σ-2 than σ-1, is only 4-fold more potent than o-DTG in the microglial

2+ migration assay but 8- to 34-fold more potent than the parent compound in the [Ca ]i

dysregulation experiments. The observation that p-BrDPhG is more potent than o-DTG

in inhibiting microglial activation may have important therapeutic implications. Previous

studies have demonstrated that activation of microglia following an ischemic insult

results in the release of pro-inflammatory cytokines by these cells, which promotes

neuronal death (Lee et al., 2001).

149

One mechanism which may explain the discrepancy between decreased binding

affinity but increased functional potency of p-BrDPhG may be increased membrane permeability of the compound. It has been shown that chloro and bromo substitution in the para position on phenylalanine increases hydrophobicity and blood-brain barrier

permeability of a drug (Gentry et al., 1999). Likewise, halogenation of small molecules

has been demonstrated to increase membrane permeation (Gerebtzoff et al., 2004). It

has been shown that increasing membrane permeability of a σ ligand by deprotonation

increases potency of the compound, which was interpreted to suggest that the binding

site for ligands such as BD-1063 are intracellular (Vilner and Bowen, 2000). Immuno-

labeling studies have confirmed this hypothesis, showing that σ-1 receptors localize to

the endoplasmic reticulum (Dussossoy et al., 1999, Seminerio et al., 2012). Moreover,

the proposed binding site for drugs such as o-DTG are, in part, located in the

transmembrane domain of the receptor (Pal et al., 2007). Thus, by increasing membrane

permeability, chloro and bromo substitution more than compensate for the decreased

binding affinity.

Our data, however, also suggest that factors other than membrane permeability

are likely involved in the effects of the halogenation and methylation. Electronegativity

and conformation of the molecule are also important. For example, fluoro substitution of

a distal phenyl group of a piperidine greatly increased the lipophilicity of the compound

(de Candia et al., 2009). Despite the likely increased lipid permeability, our findings show

2+ that fluoro substitution does not result in increased inhibition of [Ca ]i dysregulation

evoked via ischemia or acidosis relative to o-DTG. One possibility is that while the fluoro

substitution may increase the drugs lipophilicity, the added increase in electronegativity 150

caused by addition of a fluoro moiety may have disrupted binding of the compound to the

σ receptors. While fluorine has an electronegativity of 4.0, that of chlorine and bromine

are ~ 3.0. Moreover, our data suggest that location of chloro and bromo substitutions on

the phenyl ring for enhanced potency were limited to either the meta or para positions. A

previous report on structure-activity relationship of DTG analogues suggested that the

ortho position was the most tolerant to structural modifications, including addition of

electronegative moiety (Scherz et al., 1990). However, that report did not distinguish

between σ-1 and σ-2 receptor binding, and thus the effects of those modifications on the

respective receptors remains to be determined.

It has been shown that following a stroke there is an increase in the production

of the amyloid-β peptide (Stephenson et al., 1992, Jablonski et al., 2011, Pluta et al.,

2013). There are currently no studies that have looked at potential therapeutic targets to mitigate post-stroke amyloid-β induce toxicity. Aβ25-35 was found to produce increases in

2+ [Ca ]i after cells were incubated in the Aβ fragment for ≥ 1 hr, with short applications ≤

2+ 2+ 15 min failing to alter [Ca ]i. Maximal increases in [Ca ]i were observed after 12 hr

2+ incubation of the neurons in Aβ25-35 and [Ca ]i remained significantly elevated even after

2+ 24 hr incubation in the Aβ fragment. The observed 2-fold increase in [Ca ]i caused by

Aβ25-35 is similar to that previously reported for rat cortical neurons incubated for 24 hr in

Aβ25-35 (Ferreiro et al., 2004). Despite the DTG analogue p-BrDPhG showing a greater

2+ potency for mitigating ischemia- and acidosis-induced increases in [Ca ]i elevations, we

decided to use another pan-selective sigma receptor agonist. The decision to use

afobazole was predominately two-fold. First, the guanidine compounds are known to be

151

“dirty” (i.e. bind to many receptors), whereas afobazole has minimal non-target effects.

Second, afobazole has been better characterized and is currently being used clinically.

Therefore, neurons were incubated in 100 µM afobazole and found to reduce Aβ25-35-

2+ 2+ evoked increases in [Ca ]i by ~60%. This decrease in [Ca ]i is similar to that observed

2+ when afobazole was used to inhibit [Ca ]i increases produced in cortical neurons by

2+ acidosis (IC50 = 164 µM) (Cuevas et al., 2011a). Reducing Aβ25-35 perturbation of [Ca ]i

2+ is likely to promote neuronal survival, since [Ca ]i destabilization has been linked to cell

death in AD (Mattson and Chan, 2003b). Moreover, the Aβ25-35-induced changes in

2+ [Ca ]i precede pro-apoptotic signals in the model used here (e.g. decrease in Bcl-2 levels occurring 48 hrs after Aβ25-35 application).

2+ The inhibition of Aβ25-35-elicited increases in [Ca ]i by afobazole is due to

activation of σ-1, but not σ-2 receptors. Incubation in the σ-1 receptor antagonist, BD-

1047 (Matsumoto et al., 2004), reduced the effects of afobazole by ~40%. The concentration and degree of block reported here is consistent with previous studies showing BD-1047 inhibition of σ-1 receptor-mediated effects in cortical neurons and retinal ganglion cells (Katnik et al., 2006, Cuevas et al., 2011a, Mueller et al., 2013). The lack of involvement of σ-2 receptors is confirmed by results obtained using the σ-2 receptor antagonist, rimcazole (Gilmore et al., 2004). Rimcazole binds to σ-2 receptors with nanomolar affinity, but to σ-1 receptor with low micro molar affinity (Ferris et al.,

1986, Husbands et al., 1999, Rybczynska et al., 2008). Given that 300 nM rimcazole

2+ failed to inhibit the actions of afobazole on Aβ25-35-evoked increases in [Ca ]i, but that

the higher concentration of the drug reduced these effects, it is unlikely that afobazole

152

2+ activates σ-2 receptors to suppress Aβ25-35-evoked increases in [Ca ]i. Surprisingly, a second pan-selective σ receptor agonist, DTG, failed to mimic the effects of afobazole on

2+ Aβ25-35-evoked [Ca ]i perturbation. DTG and afobazole both block acidosis- and ischemia-induced Ca2+ overload in neurons to a similar extent (Cuevas et al., 2011a). It

2+ 2+ was shown that ischemia-induced [Ca ]i increases were due primarily to Ca influxes through voltage-gated Ca2+ channels and NMDA receptors which were regulated by σ receptor activation (Katnik et al., 2006). However, the difference between afobazole and

2+ 2+ DTG effects on Aβ modulation of [Ca ]i, suggest these long-term [Ca ]i increases

2+ involve distinct mechanisms from those producing ischemia-induced [Ca ]i overload.

2+ Additionally, the lack of a DTG effect on Aβ-evoked [Ca ]i overload suggests that not all

σ-1 agonists may promote neuronal survival in this model. This hypothesis is further supported by our observation that DTG fails to decrease neuronal apoptosis following 24 hr incubation Aβ25-35. The difference in responses between afobazole and DTG may be due to off-target effects of DTG counteracting the benefits of this pan-selective sigma agonist.

2+ It has been suggested that one of the downstream consequences of [Ca ]i dysregulation in neurons caused by Aβ25-35 is the production of ROS (Ekinci et al., 2000,

Abramov et al., 2004). Afobazole has been shown to reduce ROS accumulation in rat brains after focal ischemia (Silkina et al., 2006). However, data presented here show that afobazole does not block the ROS increases caused by the Aβ fragment. It has been suggested that Aβ, and in particular Aβ25-35, can increase oxidative stress via

2+ mechanisms independent of [Ca ]i dysregulation (Varadarajan et al., 2001). Given that

153

2+ afobazole inhibits [Ca ]i dysregulation produced by Aβ25-35 in neurons but not the

2+ increase in ROS, it is unlikely that afobazole is affecting this [Ca ]i-independent ROS

production. It is important to note that while afobazole may not decrease ROS

production, it has been shown that σ-1 receptors decrease H2O2 evoked apoptosis

(Meunier and Hayashi, 2010). Thus, afobazole may in part decrease Aβ25-35-induced cell

death by acting downstream of the observed ROS increase.

Aβ25-35 has been shown to increase nitric oxide synthase (NOS) activity and

produce neuronal injury via nitrosative stress (Parks et al., 2001, Cho et al., 2009). Aβ25-

35-evoked increases in NO were significantly decreased following afobazole application.

Inhibition of inducible NOS (iNOS) has been proposed as a mechanism by which activation of σ-1 receptors is neuroprotective after stroke (Vagnerova et al., 2006).

Similarly, functional downregulation of neuronal NOS (nNOS) has been shown to

contribute to σ receptor neuroprotection following ischemia in striatal neurons (Yang et

al., 2010). Since Aβ25-35 appears to increase both iNOS and nNOS in neurons (Limon et

al., 2009), activation of σ receptors by afobazole is predicted to reduce NO production by

both of these enzymes, and will ultimately contribute to protection from Aβ25-35 induced

injury.

One of the pro-apoptotic proteins upregulated in neurons both by the

pathological sequela induced by Aβ25-35 application and in patients with Alzheimer’s

disease is Bax (Paradis et al., 1996, Yao et al., 2005). Consistent with previous findings,

the current study shows that the number of Bax positive neurons is significantly

increased following 24 hr incubation in Aβ25-35. However, when afobazole is applied in

154

combination with Aβ25-35, Bax upregulation is diminished significantly. It has also been

shown that the σ-1 agonist, PRE-084, can reduce apoptosis in cortical neurons caused

by Aβ25-35 via reduction in Bax (Marrazzo et al., 2005). However, glutamatergic

neurotransmission was blocked in those experiments (Marrazzo et al., 2005).

Glutamatergic transmission is a factor in Aβ25-35-induced cell death, and elevated

glutamate levels have been shown to induce Bax upregulation in neurons (Schelman et

al., 2004, Revett et al., 2013). Thus, our data suggest that σ receptor activation by

afobazole can reduce Bax expression and concomitant cell death in the presence of

Aβ25-35 even when glutamatergic transmission is not blocked.

In addition to decreasing Bax levels, afobazole effectively reduced activated

caspase-3 expression in response to Aβ25-35. The active form of caspase-3 has been

implicated in the loss of hippocampal neurons in Alzheimer’s disease (Selznick et al.,

1999). In addition to serving as a late event involved in neuronal death during

Alzheimer’s disease, caspase activation may be an early event that promotes the

pathology of AD. Several caspases, including caspase-3, have been shown to cause

pathological tau filament assembly in neurons, which underlies AD etiology (Gamblin et al., 2003). Selective inhibition of caspase-3 has been shown to protect neurons from

Aβ25-35-induced apoptosis (Allen et al., 2001). Sigma receptor activation has been shown to decrease caspase-3 and to protect cortical neurons and RGC-5 cells against excitotoxicity (Tchedre and Yorio, 2008, Griesmaier et al., 2012). Such a mechanism is likely to contribute to afobazole neuroprotection from Aβ25-35 toxicity and may in part

explain the reduced neuronal death observed here.

155

The neuroprotective properties of afobazole following exposure to Aβ25-35 also

involves long-term upregulation of Bcl-2. Our data indicate that there is a biphasic

change in Bcl-2 levels in neurons following application of Aβ25-35 with an initial increase

at 24 hr followed by a suppression of Bcl-2 expression at 48 hr. A similar transient expression pattern was previously reported in rat hippocampal neurons exposed to Aβ25-

35 (Kim et al., 1998). It was proposed that this initial increase in Bcl-2 expression is a

non-sustainable cellular response to the apoptotic pathways induced by Aβ (Kim et al.,

1998). Our observations indicate that when afobazole is applied along with Aβ25-35, Bcl-2

levels do not drop below control at the 48 hr time point. Previous studies have shown

that Bcl-2 is a target for σ receptor mediated neuroprotection. The HIV-1 protein gp120 was shown to downregulate Bcl-2 in cortical neurons, but activation of σ receptors with

the σ ligand, PPBP, increased Bcl-2 expression (Zhang et al., 2012). PPBP was shown

to have a similar effect in a glutamate excitotoxicity model (Yang et al., 2007),

suggesting that Bcl-2 upregulation may occur after σ receptor stimulation under various

pathophysiological conditions.

Previous studies from our laboratory have shown that microglia quickly become

activated and withdraw their filopodia when exposed to various stimuli, such as LPS and

ATP (Hall et al., 2009a). This activated state, which allows the microglia to migrate to the

site of injury, is preceded by the release of neurotoxic substances (Kierdorf and Prinz,

2013). Previously we have shown that microglia, in response to ATP, alter their

morphology to a less ramified state and this reaction was inhibited by DTG (Hall et al.,

2009a). Afobazole effectively blocked membrane ruffling by approximately 50% in

156

response to ATP. For comparison we used a second pan-selective σ receptor agonist,

DTG, which reduced ruffling comparable to afobazole and to the extent as previously

reported. In addition, afobazole was recently shown to mitigate microglial migration in a

concentration dependent manor in response to ATP and UTP. Other studies have shown

that microglia rapidly respond to Aβ by withdrawing their filopodia and assuming an

amoeboid, ruffled morphology (Araujo and Cotman, 1992). This early event is indicative

of microglial activation and precedes the inflammatory response that includes release of

neurotoxic substances from the microglia (Combs et al., 2001, Garcao et al., 2006,

Orellana et al., 2011). Afobazole (30 µM) effectively blocked membrane ruffling in

response to Aβ25-35. While a second pan-selective σ receptor agonist, DTG (30 µM), also

decreased membrane ruffling caused by Aβ25-35, this drug was significantly less potent than afobazole. It has been shown that higher concentrations of DTG (100 µM) are also

required to block ATP-induced microglial membrane ruffling (Hall et al., 2009a).

Afobazole also effectively blocked microglial migration induced by Aβ25-35. It has

been shown previously that various Aβ fragments, including Aβ25-35, are microglial chemotactic agents (Davis et al., 1992). The Aβ-induced chemotaxis is in part due to release of ATP from microglia following Aβ25-35 application and subsequent activation of

P2Y2 receptors on these cells (Kim et al., 2012). We have previously shown that afobazole blocks microglial migration induced by activation P2Y receptors (Cuevas et al.,

2011b). Afobazole appears to inhibit microglial migration caused by UTP via σ receptor-

mediated disruption in P2Y signaling, since afobazole treatment also inhibited P2Y-

2+ induced elevation in [Ca ]i in microglia (Cuevas et al., 2011b). The IC50 for afobazole

157

inhibition of microglial migration caused by Aβ25-35 is 120 µM, which is similar to the low-

affinity site reported for afobazole inhibition of UTP-induced microglial migration (102

µM) (Cuevas et al., 2011b). Therefore, afobazole may in part prevent microglial

chemotaxis caused by Aβ25-35 via σ receptor-mediated inhibition of P2Y receptors in

these cells.

Data shown here indicate that both σ receptor subtypes are involved in afobazole

inhibition of Aβ25-35-evoked microglial activation. Afobazole-mediated inhibition of

microglial migration was reduced by ~50% by 10 µM BD 1047, which is indicative of a σ-

1 receptor mediated effect (Matsumoto et al., 1995). An identical concentration of BD

1047 had a similar effect on afobazole inhibition of ATP-induced microglial cell migration

(Cuevas et al., 2011b). Likewise, involvement of σ-2 receptors is confirmed by results obtained using the σ receptor antagonist, rimcazole, which binds to σ-2 receptors with nanomolar affinity (Ferris et al., 1986, Rybczynska et al., 2008). Nanomolar concentrations of rimcazole also blocked afobazole inhibition of microglial migration in response to ATP (Cuevas et al., 2011b).

Given that one of the consequences of microglial activation is increased oxidative and nitrosative stress, which leads to Aβ25-35 toxicity (Weldon et al., 1998, Bianca et al.,

1999, Combs et al., 2001), we examined how afobazole affects Aβ25-35-induced

increases in cellular ROS and NO levels. Consistent with previous reports, both ROS

and NO levels were increased significantly following Aβ25-35 application. However,

afobazole failed to attenuate the increases in ROS and NO evoked by the Aβ fragment.

Activation of σ receptors has been shown to decrease oxidative injury and nitrosative

158

stress in various models. It was recently shown, using σ receptor knockout mice, that σ-1

has a dual function of both reducing oxidative stress and activating antioxidant response

elements (Pal et al., 2012). In microglia, stimulation of σ receptors by DTG reduces NO

production in response to lipopolysaccharide (Hall et al., 2009a). Taken together,

however, our data suggest that the reduction in Aβ-evoked microglial cell toxicity

provided by afobazole application is not the result of σ receptor-mediated decrease in

oxidative or nitrosative stress.

While decreased activation of microglia will be beneficial following a stroke by

reducing the neuroinflammatory response, recent data suggests that enhanced

microglial survival is also important for improved outcomes given the ability of these cells

to phagocytose Aβ plaques (Streit et al., 2009, Solito and Sastre, 2012). Application of

30 µM afobazole significantly reduced the number of microglia undergoing apoptosis

following 72 hr incubation in Aβ25-35. The concentrations of afobazole shown to be effective here are identical to those previously shown to decrease microglial cell death

caused by ischemia in a σ receptor dependent manner and consistent with the binding

affinity of afobazole for σ-1 receptors (Seredenin et al., 2009, Cuevas et al., 2011a,

Cuevas et al., 2011b). This observation suggests that afobazole is similarly acting

through σ receptor activation to prevent microglial cell death. Both melatonin and N-

acetylcysteine have been shown to decrease Aβ25-35-evoked apoptosis in microglia via a

reduction in ROS (Jang et al., 2005). Given that afobazole does not appear to affect

Aβ25-35-elicited increases in ROS, it is unlikely that afobazole provides cytoprotection via

a similar mechanism to these compounds.

159

Our findings demonstrate that afobazole mitigates Aβ25-35-induced apoptosis of

microglia by decreasing expression of the pro-apoptotic gene, Bax, and the death

protease caspase-3 while preserving the levels of the anti-apoptotic gene, Bcl-2. An increase in Bax levels has been observed in both neurons and microglia of AD brains

(Su et al., 1997). Moreover, interventions that decrease Aβ25-35-induced microglial apoptosis have been shown to lower Bax expression (Jang et al., 2005, Shang et al.,

2012). Likewise, caspase activity has been detected in plaque-associated neurons and

microglia in AD brains (Yang et al., 1998). Caspase-3 levels have been shown to be upregulated in microglia following Aβ administration, while reduced levels of this death protease have been associated with inhibition of Aβ toxicity (Shang et al., 2012).

Conversely, expression of the anti-apoptotic gene, Bcl-2, is inversely correlated with disease severity in AD. Increases in Bcl-2 levels promote microglial survival following exposure to Aβ25-35 (Satou et al., 1995, Jang et al., 2005). Therefore, afobazole likely

enhances microglial survival and resistance to Aβ25-35 toxicity via a complex response

involving both anti-apoptotic and pro-apoptotic genes. This multifaceted effect is

consistent with afobazole acting as a σ agonist, since σ receptor activation has been

shown to modulate Bax, caspase-3 and Bcl-2 levels (Tchedre and Yorio, 2008, Meunier

and Hayashi, 2010).

Importantly, not only did afobazole protect microglia from Aβ25-35-evoked

apoptosis, but application of the σ agonist preserved the functional capacity of these

2+ cells, as evident from the continued [Ca ]i responses to ATP. Ultimately, one of the

theoretical benefits to increased microglial survival would be continued clearance of Aβ,

160

as this peptide has been shown to be phagocytosed by microglia (Wilcock et al., 2004,

Herber et al., 2007). This hypothesis is further supported by the recent observation that

microglial clearance of Aβ directly correlates with expression of the gene CD33, such

that a CD33 deficiency reduces brain levels of both insoluble Aβ1-42 and amyloid plaque

burden in a mouse AD model (Griciuc et al., 2013). Moreover, there is a decrease in both

CD33 expression and insoluble Aβ1-42 in the brains of individuals expressing the minor

allele of the CD33 (SNP rs3865444) gene, which was identified previously as imparting

protection against AD (Naj et al., 2011, Griciuc et al., 2013). The fact that responses to

ATP itself are preserved is significant, since the clearance of Aβ has been shown to be

dependent of the activation of P2Y2 receptors. Microglia respond to Aβ by releasing ATP

and this nucleotide functions in a paracrine manner, acting on P2Y2 receptors expressed

on the microglia to enhance Aβ uptake by these cells (Kim et al., 2012). We have

2+ previously shown that in our model these increases in [Ca ]i are due to activation of P2Y

receptors, since they can be elicited by UTP (Cuevas et al., 2011b). Therefore, by

preserving ATP responses in microglia, afobazole will permit these cells to respond to Aβ in vivo and potentially decrease amyloid burden.

It has been suggested that over-activation of microglia result in neuronal death

(Hellwig et al., 2013). However, the need to maintain viable microglia that respond to sites of injury are equally necessary. Neurons exposed to ischemic conditions release cytotoxic and neurotropic factors that both activate and attract microglia to the area of injury (Hellwig et al., 2013, Kierdorf and Prinz, 2013). Unfortunately, in disease states like stroke, over-activation of microglia is a common occurrence and often times result in

161

neuronal and microglial cell death. Thus it is of great importance to identify factors that

contribute to the demise of microglia and find treatments to combat these mechanisms.

Exposure of microglia cells to ischemia resulted in approximately 150-fold increase in

cell death from control alone. Afobazole reduced the number of dyeing microglia by 75 ±

8% when co-incubated with ischemia.

Our laboratory has previously shown that afobazole can mitigate intracellular

calcium dyshomeostasis in neurons and microglia following ischemia, acidosis and ATP

exposure (Katnik et al., 2006, Herrera et al., 2008, Cuevas et al., 2011a, Cuevas et al.,

2011b). A rise in intracellular concentration, if unresolved, can promote the initiation of

apoptosis (Mattson and Chan, 2003a). Immunocytochemical experiments were

conducted to determine if afobazole affects microglial expression of the pro-apoptotic

protein Bax or the death protease, caspase-3, following 24 hr ischemic exposure. Both

Bax and caspase-3 expression increased > 270 and 150% from the control, respectively.

We found that afobazole alone decreased caspase-3 expression and when co-incubated

with ischemia both Bax and caspase-3 expression were mitigated approximately 42 and

59%, respectively. Further experiments were conducted to determine if levels of Bcl-2

were affected by ischemia and to what if affect afobazole had on its expression.

Following ischemic incubation, microglial Bcl-2 expression was decreased by ~ 21% from that of the control. However, when co-incubated with ischemia, afobazole preserved

Bcl-2 expression by approximately 24%, making it comparable to afobazole alone.

Exposing cells to ischemic conditions has been shown to decrease cell viability.

Our laboratory has shown that afobazole mitigates death following 2 hr ischemia exposure (Cuevas et al., 2011a, Cuevas et al., 2011b). This model resembles injury 162

caused by reperfusion of the tissues with oxygen, more of a transient ischemic model.

Experiments were conducted to determine to what extent afobazole could be

neuroprotective following prolonged ischemic exposure (72 hr). There was an

approximant 3-fold increase in the number of expired neurons from that of the control.

This increase in the demise of neurons was mitigated by approximately 55% when co-

incubated with afobazole. This data indicates that even under prolonged ischemic

conditions afobazole was to some extent neuroprotective. Immunocytochemical

experiments were carried out to determine if one mechanism by which afobazole is

neuroprotective is through sigma receptor regulation of apoptotic expression. Both Bax

and caspase-3 expression were observed under all conditions. Following ischemic

exposure, both Bax and caspase-3 levels increased by ~ 47 and 35%, respectively.

However, unlike the results we observed in microglia cells, afobazole did not mitigate increases in Bax or caspase-3 expression in neurons. The ratio of Bcl-2:Bax has been shown to positively correlate to cell survival in neurons exposed to Aβ (Patel and Brewer,

2008). Likewise, the ratio of Bcl-2:Bax is important in regulating cell survival following

glutamate excitotoxicity (Schelman et al., 2004) and previous studies have shown that

Bcl-2 is a target for σ receptor mediated neuroprotection (Yang et al., 2007, Meunier and

Hayashi, 2010). Additionally, experiments were performed to determine if afobazole could preserve expression of Bcl-2 levels following ischemia. As we observed with

microglia, Bcl-2 expression decreased in neurons under ischemic conditions. We found

that afobazole alone increased Bcl-2 expression; however, when co-incubated with

ischemia, afobazole failed to preserve Bcl-2 expression levels and were comparable to ischemia alone. 163

However, when functional integrity of these surviving neurons was examined by

attempting to depolarize them with a focal application of 30mM KCl, we found them

unresponsive. It appeared that afobazole might not be as neuroprotective as we first

thought against prolonged ischemic exposure. However we realized that the conditions

in which these neurons were exposed resembled a near core environment and not the

penumbra. Neuronal necrosis contributes to the expansion of the core into the

penumbra. The fact that afobazole prevents the dumping of neuronal cellular contents

into surrounding tissue and allows for these damaged cells to undergo controlled cell

death.

Lastly, In addition to large vessel ischemic strokes there are small vessel ischemic

stroke known as lacunar infarcts, which account for approximately 25% of all ischemic

insults (Samuelsson et al., 1996). These strokes result from the blockage of small

penetrating arteries to the subcortical regions of the brain (Kang et al., 2012). This type infarct is a leading cause of cognitive impairment and vascular dementia (Charidimou and Werring, 2012). While the risk factors resulting in lacunar verse large artery infarcts differ, the resulting cellular cascade promoting cerebral cell death may have overlapping mechanisms. One of these mechanisms may be the increased production and toxicity of the amyloid-β peptide, which has been shown to be upregulated following an ischemic event (Popa-Wagner et al., 1998, Guglielmotto et al., 2009, Pluta et al., 2013). Neuronal cultures were used to determine if incubation with 25 µM amyloid-β25-35 produced an

2+ increase in the [Ca ]i levels evoked by ischemia + acidosis application. Incubation for 12

hr failed to potentiate Ca2+ elevations. Given that Aβ modulation of these responses may

require longer incubation period for gene up-regulation. Therefore, neurons were 164

incubated for 24 hr with Aβ and found to have potentiated ischemia, acidosis and

2+ ischemia + acidosis induced increases in [Ca ]i levels. We found that acidosis

2+ application alone produced an increase in mean peak [Ca ]i by ~42%. Furthermore,

application of ischemia alone or ischemia + acidosis also produced potentiated

elevations in both the peak and total net calcium influx by ~72 and 82% for ischemia and

2+ ~43 and 85% for ischemia + acidosis, respectively. This dual potentiation in [Ca ]i may indicated that the mechanism in these response involve multiple pathways including

ASIC1a, VGCCs, NMDA and AMPA/kainate receptors. Previous studies from our

2+ laboratory have shown that ischemia-induced elevations in [Ca ]i involved the activation

of NMDA receptors (Katnik et al., 2006), whereas acidosis application activates ASIC1a receptors (Herrera et al., 2008). Moreover, the ischemia + acidosis activation of NMDA

2+ and ACIS1a receptors results in a synergist increase in [Ca ]i levels. This is a

complicated interaction with NMDA receptors functionally up-regulating ASIC1a, while at

the same time ASIC1a potentiates NMDA receptor activity (Mari et al., 2014).

Since activation of ASIC1a has been shown to potentiate ischemia-induced

2+ [Ca ]i levels through NMDA receptors, we were interested in determining were Aβ

potentiation occurred. Previous studies have shown that only a small part of the ischemia

+ acidosis evoked Ca2+ influx is directly due to ACIS1a with VGCCs, NMDA and

AMPA/kainate receptors contributing to the bulk influx (Gonzalez-Alvear and Werling,

1995, Alonso et al., 2000). However, fluorometric calcium imaging showed that Aβ-

induced a potentiated proton activated Ca2+ influx. Therefore, whole-cell patch-clamp

experiments were performed to determine whether this potentiation was due to directly to

ASIC1a activity or via ASIC1a enhanced downstream channels. Neurons Aβ-incubated 165

for 24 hr were voltage-clamped at -70 mV, which prevents NMDA receptor and VGCC

activation, demonstrated a ~68% increase in the proton evoked inward current. Taken

together, our results indicate that Aβ incubation is directly up-regulating ASIC1a activity

as well as the ASIC1a potentiated down-stream channels.

Reducing the Ca2+ overload produced by an ischemic insult is crucial to mitigating

stroke injury. Previous studies from our laboratory have shown that σ-1 receptor activation in neurons can elevate the Ca2+ overload induced by ischemia, acidosis and

Aβ (Cuevas et al., 2011a, Behensky et al., 2013a). However, not all sigma receptor

agonists were shown to be as effective under these conditions (Behensky et al., 2013a).

The pan-selective agonist afobazole was one such drug that did mitigate Ca2+ overload

induced under all three conditions. Therefore, 100 µM afobazole was co-incubated with

Aβ for 24 hr to assess what function activation of sigma receptors would have on the Aβ

up-regulated function. Co-incubation with afobazole completely abolished the potentiated

2+ increases in [Ca ]i levels-induced via focal application of ischemia + acidosis. Reducing

the Aβ up-regulated Ca2+ increases will likely translate to an increase in neuronal

survival.

The overall significance of this study shows that sigma receptor activation via

afobazole can decrease both ischemic- and Aβ-induced injury following a stroke. The

2+ activation of σ-1 receptors prevented Aβ25-35-induced [Ca ]i overload and mediated neuroprotection by downregulation of both Bax and activated caspase-3, and long-term upregulation of Bcl-2. This translated to a decrease in Aβ25-35-evoked neuronal

apoptosis. In addition, afobazole through σ-1 and σ-2 receptors mitigated the activation

and migration of microglial cells in response to Aβ25-35. The observed decrease in 166

apoptosis is due to afobazole-mediated regulation of multiple genes, with a noted

decrease in Bax and caspase-3 and a concomitant increase in Bcl-2 expression. In

addition to the decrease in cell death, afobazole preserved functional activity of the cells

2+ as evident by the conserved ATP-induced increases in [Ca ]i following prolonged

incubation in Aβ25-35. Taken together our data suggest that afobazole may prevent microglia from entering the activated state associated with neurotoxicity following exposure to Aβ, and increasing their survival, which will facilitate clearance of the

amyloid peptide. Our data show one mechanism by which Aβ may potentiate neuronal

2+ injury during ischemia-acidosis is through increased [Ca ]i overload, and identify

ASIC1a as the channel mediating this phenomenon. Finally, our data suggest that

activation of σ receptors by afobazole may represent a therapeutic strategy for mitigating

concurrent injury produced by Aβ and stroke.

167

LIST OF REFERENCES

Abate C, Elenewski J, Niso M, Berardi F, Colabufo NA, Azzariti A, Perrone R, Glennon RA (2010) Interaction of the sigma(2) receptor ligand PB28 with the human nucleosome: computational and experimental probes of interaction with the H2A/H2B dimer. ChemMedChem 5:268-273. Abate C, Niso M, Infantino V, Menga A, Berardi F (2015) Elements in support of the 'non-identity' of the PGRMC1 protein with the sigma2 receptor. Eur J Pharmacol 758:16-23. Abramov AY, Canevari L, Duchen MR (2004) Calcium signals induced by amyloid beta peptide and their consequences in neurons and astrocytes in culture. Biochim Biophys Acta 1742:81-87. Ajmo CT, Jr., Vernon DO, Collier L, Pennypacker KR, Cuevas J (2006) Sigma receptor activation reduces infarct size at 24 hours after permanent middle cerebral artery occlusion in rats. Curr Neurovasc Res 3:89-98. Alberdi E, Sanchez-Gomez MV, Cavaliere F, Perez-Samartin A, Zugaza JL, Trullas R, Domercq M, Matute C (2010) Amyloid beta oligomers induce Ca2+ dysregulation and neuronal death through activation of ionotropic glutamate receptors. Cell Calcium 47:264-272. Allen JW, Eldadah BA, Huang X, Knoblach SM, Faden AI (2001) Multiple caspases are involved in beta-amyloid-induced neuronal apoptosis. J Neurosci Res 65:45-53. Alonso G, Phan V, Guillemain I, Saunier M, Legrand A, Anoal M, Maurice T (2000) Immunocytochemical localization of the sigma(1) receptor in the adult rat central nervous system. Neuroscience 97:155-170. Antipova TA, Sapozhnikova DS, Stepanichev MY, Onufriev MV, Gulyaeva NV, Seredenin SB (2010) Effects of selective anxiolytic afobazole on active caspase-3. Bull Exp Biol Med 149:201-203. Araujo DM, Cotman CW (1992) Beta-amyloid stimulates glial cells in vitro to produce growth factors that accumulate in senile plaques in Alzheimer's disease. Brain Res 569:141-145. Armstead WM, Ganguly K, Kiessling JW, Riley J, Chen XH, Smith DH, Stein SC, Higazi AA, Cines DB, Bdeir K, Zaitsev S, Muzykantov VR (2010) Signaling, delivery and age as emerging issues in the benefit/risk ratio outcome of tPA For treatment of CNS ischemic disorders. J Neurochem 113:303-312. Baykova VS, Kadnikov IA, Voronin MV, Ganshina TS, Gnezdilova AV, Gorbunov AA, Mirzoyan PC, Seredenin SB (2011) Effects of afobazole on the content of neurotransmitter amino acids in the striatum in global transient ischemia. Bull Exp Biol Med 151:593-596. Behensky AA, Yasny IE, Shuster AM, Seredenin SB, Petrov AV, Cuevas J (2013a) Afobazole Activation of Sigma-1 Receptors Modulates Neuronal Responses to Amyloid-beta25-35. J Pharmacol Exp Ther. Behensky AA, Yasny IE, Shuster AM, Seredenin SB, Petrov AV, Cuevas J (2013b) Stimulation of Sigma Receptors with Afobazole Blocks Activation of Microglia and Reduces Toxicity Caused by Amyloid-beta25-35. J Pharmacol Exp Ther.

168

Besancon E, Guo S, Lok J, Tymianski M, Lo EH (2008) Beyond NMDA and AMPA glutamate receptors: emerging mechanisms for ionic imbalance and cell death in stroke. Trends Pharmacol Sci 29:268-275. Bianca VD, Dusi S, Bianchini E, Dal Pra I, Rossi F (1999) beta-amyloid activates the O-2 forming NADPH oxidase in microglia, monocytes, and neutrophils. A possible inflammatory mechanism of neuronal damage in Alzheimer's disease. J Biol Chem 274:15493-15499. Bowen WD (2000) Sigma receptors: recent advances and new clinical potentials. Pharm Acta Helv 74:211-218. Butler TL, Kassed CA, Sanberg PR, Willing AE, Pennypacker KR (2002) Neurodegeneration in the rat hippocampus and striatum after middle cerebral artery occlusion. Brain Res 929:252-260. Camello-Almaraz C, Gomez-Pinilla PJ, Pozo MJ, Camello PJ (2006) Mitochondrial reactive oxygen species and Ca2+ signaling. Am J Physiol Cell Physiol 291:C1082-1088. Cassano G, Gasparre G, Niso M, Contino M, Scalera V, Colabufo NA (2009) F281, synthetic agonist of the sigma-2 receptor, induces Ca2+ efflux from the endoplasmic reticulum and mitochondria in SK-N-SH cells. Cell Calcium 45:340-345. Cha MY, Han SH, Son SM, Hong HS, Choi YJ, Byun J, Mook-Jung I (2012) Mitochondria- specific accumulation of amyloid beta induces mitochondrial dysfunction leading to apoptotic cell death. PLoS One 7:e34929. Charidimou A, Werring DJ (2012) Cerebral microbleeds and cognition in cerebrovascular disease: an update. J Neurol Sci 322:50-55. Cho DH, Nakamura T, Fang J, Cieplak P, Godzik A, Gu Z, Lipton SA (2009) S-nitrosylation of Drp1 mediates beta-amyloid-related mitochondrial fission and neuronal injury. Science 324:102-105. Cobos EJ, Entrena JM, Nieto FR, Cendan CM, Del Pozo E (2008) Pharmacology and therapeutic potential of sigma(1) receptor ligands. Curr Neuropharmacol 6:344-366. Colabufo NA, Berardi F, Abate C, Contino M, Niso M, Perrone R (2006) Is the sigma2 receptor a histone binding protein? J Med Chem 49:4153-4158. Combs CK, Karlo JC, Kao SC, Landreth GE (2001) beta-Amyloid stimulation of microglia and monocytes results in TNFalpha-dependent expression of inducible nitric oxide synthase and neuronal apoptosis. J Neurosci 21:1179-1188. Cortes-Salva M, Nguyen BL, Cuevas J, Pennypacker KR, Antilla JC (2010) Copper-catalyzed guanidinylation of aryl iodides: the formation of N,N'-disubstituted guanidines. Org Lett 12:1316-1319. Crawford KW, Bowen WD (2002) Sigma-2 receptor agonists activate a novel apoptotic pathway and potentiate antineoplastic drugs in breast tumor cell lines. Cancer Res 62:313-322. Cregan SP, MacLaurin JG, Craig CG, Robertson GS, Nicholson DW, Park DS, Slack RS (1999) Bax-dependent caspase-3 activation is a key determinant in p53-induced apoptosis in neurons. J Neurosci 19:7860-7869. Cuevas J, Behensky A, Deng W, Katnik C (2011a) Afobazole modulates neuronal response to ischemia and acidosis via activation of sigma-1 receptors. J Pharmacol Exp Ther 339:152-160. Cuevas J, Berg DK (1998) Mammalian nicotinic receptors with alpha7 subunits that slowly desensitize and rapidly recover from alpha-bungarotoxin blockade. J Neurosci 18:10335- 10344. Cuevas J, Rodriguez A, Behensky A, Katnik C (2011b) Afobazole modulates microglial function via activation of both sigma-1 and sigma-2 receptors. J Pharmacol Exp Ther 339:161- 172.

169

Davis JB, McMurray HF, Schubert D (1992) The amyloid beta-protein of Alzheimer's disease is chemotactic for mononuclear phagocytes. Biochem Biophys Res Commun 189:1096- 1100. de Candia M, Liantonio F, Carotti A, De Cristofaro R, Altomare C (2009) Fluorinated benzyloxyphenyl piperidine-4-carboxamides with dual function against thrombosis: inhibitors of factor Xa and platelet aggregation. J Med Chem 52:1018-1028. Dheen ST, Kaur C, Ling EA (2007) Microglial activation and its implications in the brain diseases. Curr Med Chem 14:1189-1197. Dumont M, Lemaire S (1991) Interaction of 1,3-di(2-[5-3H]tolyl) guanidine with sigma 2 binding sites in rat heart membrane preparations. Eur J Pharmacol 209:245-248. Dussossoy D, Carayon P, Belugou S, Feraut D, Bord A, Goubet C, Roque C, Vidal H, Combes T, Loison G, Casellas P (1999) Colocalization of sterol isomerase and sigma(1) receptor at endoplasmic reticulum and nuclear envelope level. Eur J Biochem 263:377-386. Eichhoff G, Brawek B, Garaschuk O (2011) Microglial calcium signal acts as a rapid sensor of single neuron damage in vivo. Biochim Biophys Acta 1813:1014-1024. Ekinci FJ, Linsley MD, Shea TB (2000) Beta-amyloid-induced calcium influx induces apoptosis in culture by oxidative stress rather than tau phosphorylation. Brain Res Mol Brain Res 76:389-395. Feher A, Juhasz A, Laszlo A, Kalman J, Jr., Pakaski M, Kalman J, Janka Z (2012) Association between a variant of the sigma-1 receptor gene and Alzheimer's disease. Neurosci Lett 517:136-139. Ferreira IL, Ferreiro E, Schmidt J, Cardoso JM, Pereira CM, Carvalho AL, Oliveira CR, Rego AC (2015) Abeta and NMDAR activation cause mitochondrial dysfunction involving ER calcium release. Neurobiol Aging 36:680-692. Ferreiro E, Oliveira CR, Pereira C (2004) Involvement of endoplasmic reticulum Ca2+ release through ryanodine and inositol 1,4,5-triphosphate receptors in the neurotoxic effects induced by the amyloid-beta peptide. J Neurosci Res 76:872-880. Ferris RM, Tang FL, Chang KJ, Russell A (1986) Evidence that the potential agent rimcazole (BW 234U) is a specific, competitive antagonist of sigma sites in brain. Life Sci 38:2329-2337. Finnegan KT, Calder L, Clikeman J, Wei S, Karler R (1993) Effects of L-type calcium channel antagonists on the serotonin-depleting actions of MDMA in rats. Brain Res 603:134-138. Fontanilla D, Johannessen M, Hajipour AR, Cozzi NV, Jackson MB, Ruoho AE (2009) The hallucinogen N,N-dimethyltryptamine (DMT) is an endogenous sigma-1 receptor regulator. Science 323:934-937. Galaeva IP, Garibova TL, Voronina TA, Seredenin SB (2005) Neuroprotective effects of afobazol in experimental cerebral hemorrhage. Bull Exp Biol Med 140:535-537. Gamblin TC, Chen F, Zambrano A, Abraha A, Lagalwar S, Guillozet AL, Lu M, Fu Y, Garcia- Sierra F, LaPointe N, Miller R, Berry RW, Binder LI, Cryns VL (2003) Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer's disease. Proc Natl Acad Sci U S A 100:10032-10037. Garcao P, Oliveira CR, Agostinho P (2006) Comparative study of microglia activation induced by amyloid-beta and prion peptides: role in neurodegeneration. J Neurosci Res 84:182-193. Gaugler J, James B, Johnson T, Scholz K, Weuve J (2013) Alzheimer’s Disease Facts and Figures. Alzheimer’s Association: Alzheimer’s & Dementia 9:1-71. Gentry CL, Egleton RD, Gillespie T, Abbruscato TJ, Bechowski HB, Hruby VJ, Davis TP (1999) The effect of halogenation on blood-brain barrier permeability of a novel peptide drug. Peptides 20:1229-1238.

170

Gerebtzoff G, Li-Blatter X, Fischer H, Frentzel A, Seelig A (2004) Halogenation of drugs enhances membrane binding and permeation. Chembiochem 5:676-684. Gilmore DL, Liu Y, Matsumoto RR (2004) Review of the pharmacological and clinical profile of rimcazole. CNS Drug Rev 10:1-22. Glennon RA (2005) Pharmacophore identification for sigma-1 (sigma1) receptor binding: application of the "deconstruction-reconstruction-elaboration" approach. Mini Rev Med Chem 5:927-940. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Magid D, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER, Moy CS, Mussolino ME, Nichol G, Paynter NP, Schreiner PJ, Sorlie PD, Stein J, Turan TN, Virani SS, Wong ND, Woo D, Turner MB (2013) Heart disease and stroke statistics--2013 update: a report from the American Heart Association. Circulation 127:e6-e245. Gonzalez-Alvear GM, Werling LL (1995) sigma1 Receptors in rat striatum regulate NMDA- stimulated [3H]dopamine release via a presynaptic mechanism. Eur J Pharmacol 294:713-719. Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, Hooli B, Choi SH, Hyman BT, Tanzi RE (2013) Alzheimer's Disease Risk Gene CD33 Inhibits Microglial Uptake of Amyloid Beta. Neuron. Griesmaier E, Posod A, Gross M, Neubauer V, Wegleiter K, Hermann M, Urbanek M, Keller M, Kiechl-Kohlendorfer U (2012) Neuroprotective effects of the sigma-1 receptor ligand PRE-084 against excitotoxic perinatal brain injury in newborn mice. Exp Neurol 237:388- 395. Guglielmotto M, Aragno M, Autelli R, Giliberto L, Novo E, Colombatto S, Danni O, Parola M, Smith MA, Perry G, Tamagno E, Tabaton M (2009) The up-regulation of BACE1 mediated by hypoxia and ischemic injury: role of oxidative stress and HIF1alpha. J Neurochem 108:1045-1056. Guitart X, Codony X, Monroy X (2004) Sigma receptors: biology and therapeutic potential. Psychopharmacology (Berl) 174:301-319. Hall AA, Herrera Y, Ajmo CT, Jr., Cuevas J, Pennypacker KR (2009a) Sigma receptors suppress multiple aspects of microglial activation. Glia 57:744-754. Hall AA, Leonardo CC, Collier LA, Rowe DD, Willing AE, Pennypacker KR (2009b) Delayed treatments for stroke influence neuronal death in rat organotypic slice cultures subjected to oxygen glucose deprivation. Neuroscience 164:470-477. Hanisch UK, Kettenmann H (2007) Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 10:1387-1394. Hayashi T, Maurice T, Su TP (2000) Ca(2+) signaling via sigma(1)-receptors: novel regulatory mechanism affecting intracellular Ca(2+) concentration. J Pharmacol Exp Ther 293:788- 798. Hellewell SB, Bowen WD (1990) A sigma-like binding site in rat pheochromocytoma (PC12) cells: decreased affinity for (+)-benzomorphans and lower molecular weight suggest a different sigma receptor form from that of guinea pig brain. Brain Res 527:244-253. Hellewell SB, Bruce A, Feinstein G, Orringer J, Williams W, Bowen WD (1994) Rat liver and kidney contain high densities of sigma 1 and sigma 2 receptors: characterization by ligand binding and photoaffinity labeling. Eur J Pharmacol 268:9-18. Hellwig S, Heinrich A, Biber K (2013) The brain's best friend: microglial neurotoxicity revisited. Front Cell Neurosci 7:71.

171

Herber DL, Mercer M, Roth LM, Symmonds K, Maloney J, Wilson N, Freeman MJ, Morgan D, Gordon MN (2007) Microglial activation is required for Abeta clearance after intracranial injection of lipopolysaccharide in APP transgenic mice. J Neuroimmune Pharmacol 2:222-231. Herrera Y, Katnik C, Rodriguez JD, Hall AA, Willing A, Pennypacker KR, Cuevas J (2008) sigma- 1 receptor modulation of acid-sensing ion channel a (ASIC1a) and ASIC1a-induced Ca2+ influx in rat cortical neurons. J Pharmacol Exp Ther 327:491-502. Hockenbery DM, Oltvai ZN, Yin XM, Milliman CL, Korsmeyer SJ (1993) Bcl-2 functions in an antioxidant pathway to prevent apoptosis. Cell 75:241-251. Husbands SM, Izenwasser S, Kopajtic T, Bowen WD, Vilner BJ, Katz JL, Newman AH (1999) Structure-activity relationships at the monoamine transporters and sigma receptors for a novel series of 9-[3-(cis-3, 5-dimethyl-1-piperazinyl)propyl]carbazole (rimcazole) analogues. J Med Chem 42:4446-4455. Iniguez MA, Punzon C, Nieto R, Burgueno J, Vela JM, Fresno M (2013) Inhibitory effects of sigma-2 receptor agonists on T lymphocyte activation. Front Pharmacol 4:23. Jablonski M, Maciejewski R, Januszewski S, Ulamek M, Pluta R (2011) One year follow up in ischemic brain injury and the role of Alzheimer factors. Physiol Res 60 Suppl 1:S113-119. Jang MH, Jung SB, Lee MH, Kim CJ, Oh YT, Kang I, Kim J, Kim EH (2005) Melatonin attenuates amyloid beta25-35-induced apoptosis in mouse microglial BV2 cells. Neurosci Lett 380:26-31. Jansen KL, Faull RL, Storey P, Leslie RA (1993) Loss of sigma binding sites in the CA1 area of the anterior hippocampus in Alzheimer's disease correlates with CA1 pyramidal cell loss. Brain Res 623:299-302. Joseph R, Han E (1992) Amyloid beta-protein fragment 25-35 causes activation of cytoplasmic calcium in neurons. Biochem Biophys Res Commun 184:1441-1447. Ju Y, Asahi T, Sawamura N (2014) Arctic mutant Abeta40 aggregates on alpha7 nicotinic acetylcholine receptors and inhibits their functions. J Neurochem 131:667-674. Kaminsky YG, Marlatt MW, Smith MA, Kosenko EA (2010) Subcellular and metabolic examination of amyloid-beta peptides in Alzheimer disease pathogenesis: evidence for Abeta(25-35). Exp Neurol 221:26-37. Kang DW, Han MK, Kim HJ, Yun SC, Jeon SB, Bae HJ, Kwon SU, Kim JS (2012) New ischemic lesions coexisting with acute intracerebral hemorrhage. Neurology 79:848-855. Katnik C, Garcia A, Behensky AA, Yasny IE, Shuster AM, Seredenin SB, Petrov AV, Seifu S, McAleer J, Willing A, Cuevas J (2013) Treatment with afobazole at delayed time points following ischemic stroke improves long-term functional and histological outcomes. Neurobiol Dis 62C:354-364. Katnik C, Garcia A, Behensky AA, Yasny IE, Shuster AM, Seredenin SB, Petrov AV, Seifu S, McAleer J, Willing A, Cuevas J (2014) Treatment with afobazole at delayed time points following ischemic stroke improves long-term functional and histological outcomes. Neurobiol Dis 62:354-364. Katnik C, Guerrero WR, Pennypacker KR, Herrera Y, Cuevas J (2006) Sigma-1 receptor activation prevents intracellular calcium dysregulation in cortical neurons during in vitro ischemia. J Pharmacol Exp Ther 319:1355-1365. Kiedrowski L (2007) NCX and NCKX operation in ischemic neurons. Ann N Y Acad Sci 1099:383-395. Kierdorf K, Prinz M (2013) Factors regulating microglia activation. Front Cell Neurosci 7:44. Kim ES, Kim RS, Ren RF, Hawver DB, Flanders KC (1998) Transforming growth factor-beta inhibits apoptosis induced by beta-amyloid peptide fragment 25-35 in cultured neuronal cells. Brain Res Mol Brain Res 62:122-130. 172

Kim HJ, Ajit D, Peterson TS, Wang Y, Camden JM, Gibson Wood W, Sun GY, Erb L, Petris M, Weisman GA (2012) Nucleotides released from Abeta(1)(-)(4)(2) -treated microglial cells increase cell migration and Abeta(1)(-)(4)(2) uptake through P2Y(2) receptor activation. J Neurochem 121:228-238. Kitamura Y, Takata K, Inden M, Tsuchiya D, Yanagisawa D, Nakata J, Taniguchi T (2004) Intracerebroventricular injection of microglia protects against focal brain ischemia. J Pharmacol Sci 94:203-206. Koike MA, Lin AJ, Pham J, Nguyen E, Yeh JJ, Rahimian R, Tromberg BJ, Choi B, Green KN, LaFerla FM (2012) APP knockout mice experience acute mortality as the result of ischemia. PLoS One 7:e42665. Koistinaho M, Koistinaho J (2005) Interactions between Alzheimer's disease and cerebral ischemia--focus on inflammation. Brain Res Brain Res Rev 48:240-250. Korotzer AR, Pike CJ, Cotman CW (1993) beta-Amyloid peptides induce degeneration of cultured rat microglia. Brain Res 624:121-125. Korsmeyer SJ, Shutter JR, Veis DJ, Merry DE, Oltvai ZN (1993) Bcl-2/Bax: a rheostat that regulates an anti-oxidant pathway and cell death. Semin Cancer Biol 4:327-332. Kourrich S, Su TP, Fujimoto M, Bonci A (2012) The sigma-1 receptor: roles in neuronal plasticity and disease. Trends Neurosci 35:762-771. Kurokawa K, Mizuno K, Kiyokage E, Shibasaki M, Toida K, Ohkuma S (2011a) Dopamine D1 receptor signaling system regulates ryanodine receptor expression after intermittent exposure to in primary cultures of midbrain and cerebral cortical neurons. J Neurochem 118:773-783. Kurokawa K, Shibasaki M, Mizuno K, Ohkuma S (2011b) Gabapentin blocks methamphetamine- induced sensitization and conditioned place preference via inhibition of alpha/delta-1 subunits of the voltage-gated calcium channels. Neuroscience 176:328-335. Lahmy V, Meunier J, Malmstrom S, Naert G, Givalois L, Kim SH, Villard V, Vamvakides A, Maurice T (2013) Blockade of Tau Hyperphosphorylation and Abeta Generation by the Aminotetrahydrofuran Derivative ANAVEX2-73, a Mixed Muscarinic and sigma Receptor Agonist, in a Nontransgenic Mouse Model of Alzheimer's Disease. Neuropsychopharmacology. Langa F, Codony X, Tovar V, Lavado A, Gimenez E, Cozar P, Cantero M, Dordal A, Hernandez E, Perez R, Monroy X, Zamanillo D, Guitart X, Montoliu L (2003) Generation and phenotypic analysis of sigma receptor type I (sigma 1) knockout mice. Eur J Neurosci 18:2188-2196. Lawrence JL, Tong M, Alfulaij N, Sherrin T, Contarino M, White MM, Bellinger FP, Todorovic C, Nichols RA (2014) Regulation of presynaptic Ca2+, synaptic plasticity and contextual fear conditioning by a N-terminal beta-amyloid fragment. J Neurosci 34:14210-14218. Lazzari C, Kipanyula MJ, Agostini M, Pozzan T, Fasolato C (2015) Abeta42 oligomers selectively disrupt neuronal calcium release. Neurobiol Aging 36:877-885. Lee G, Dallas S, Hong M, Bendayan R (2001) Drug transporters in the central nervous system: brain barriers and brain parenchyma considerations. Pharmacol Rev 53:569-596. Lee L, Kosuri P, Arancio O (2014) Picomolar amyloid-beta peptides enhance spontaneous astrocyte calcium transients. J Alzheimers Dis 38:49-62. Leonardo CC, Hall AA, Collier LA, Green SM, Willing AE, Pennypacker KR (2010) Administration of a Sigma Receptor Agonist Delays MCAO-Induced Neurodegeneration and White Matter Injury. Transl Stroke Res 1:135-145. Limon ID, Diaz A, Mendieta L, Chamorro G, Espinosa B, Zenteno E, Guevara J (2009) Amyloid- beta(25-35) impairs memory and increases NO in the temporal cortex of rats. Neurosci Res 63:129-137. 173

Lipton SA (2004) Failures and successes of NMDA receptor antagonists: molecular basis for the use of open-channel blockers like in the treatment of acute and chronic neurologic insults. NeuroRx 1:101-110. Mantha AK, Moorthy K, Cowsik SM, Baquer NZ (2006) Membrane associated functions of neurokinin B (NKB) on Abeta (25-35) induced toxicity in aging rat brain synaptosomes. Biogerontology 7:19-33. Mari Y, Katnik C, Cuevas J (2010) ASIC1a channels are activated by endogenous protons during ischemia and contribute to synergistic potentiation of intracellular Ca(2+) overload during ischemia and acidosis. Cell Calcium 48:70-82. Mari Y, Katnik C, Cuevas J (2014) sigma-1 Receptor Inhibition of ASIC1a Channels is Dependent on a Pertussis Toxin-Sensitive G-Protein and an AKAP150/Calcineurin Complex. Neurochem Res. Marin N, Romero B, Bosch-Morell F, Llansola M, Felipo V, Roma J, Romero FJ (2000) Beta- amyloid-induced activation of caspase-3 in primary cultures of rat neurons. Mech Ageing Dev 119:63-67. Mark LP, Prost RW, Ulmer JL, Smith MM, Daniels DL, Strottmann JM, Brown WD, Hacein-Bey L (2001) Pictorial review of glutamate excitotoxicity: fundamental concepts for neuroimaging. AJNR Am J Neuroradiol 22:1813-1824. Marrazzo A, Caraci F, Salinaro ET, Su TP, Copani A, Ronsisvalle G (2005) Neuroprotective effects of sigma-1 receptor agonists against beta-amyloid-induced toxicity. Neuroreport 16:1223-1226. Martin WR, Eades CG, Thompson JA, Huppler RE, Gilbert PE (1976) The effects of - and nalorphine- like drugs in the nondependent and morphine-dependent chronic spinal dog. J Pharmacol Exp Ther 197:517-532. Matsumoto RR, Bowen WD, Tom MA, Vo VN, Truong DD, De Costa BR (1995) Characterization of two novel sigma receptor ligands: antidystonic effects in rats suggest sigma receptor antagonism. Eur J Pharmacol 280:301-310. Matsumoto RR, Gilmore DL, Pouw B, Bowen WD, Williams W, Kausar A, Coop A (2004) Novel analogs of the sigma receptor ligand BD1008 attenuate -induced toxicity in mice. Eur J Pharmacol 492:21-26. Mattson MP (2000) Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol 1:120-129. Mattson MP, Chan SL (2003a) Calcium orchestrates apoptosis. Nat Cell Biol 5:1041-1043. Mattson MP, Chan SL (2003b) Neuronal and glial calcium signaling in Alzheimer's disease. Cell Calcium 34:385-397. Maurice T, Su TP, Privat A (1998) Sigma1 (sigma 1) receptor agonists and neurosteroids attenuate B25-35-amyloid peptide-induced amnesia in mice through a common mechanism. Neuroscience 83:413-428. McLarnon JG (2012) Microglial chemotactic signaling factors in Alzheimer's disease. Am J Neurodegener Dis 1:199-204. Mei J, Pasternak GW (2001) Molecular cloning and pharmacological characterization of the rat sigma1 receptor. Biochem Pharmacol 62:349-355. Melani A, Turchi D, Vannucchi MG, Cipriani S, Gianfriddo M, Pedata F (2005) ATP extracellular concentrations are increased in the rat striatum during in vivo ischemia. Neurochem Int 47:442-448. Meunier J, Hayashi T (2009) Sigma-1 receptors regulate Bcl-2 expression by ROS-dependent transcriptional regulation of NF-{kappa}B. J Pharmacol Exp Ther. Meunier J, Hayashi T (2010) Sigma-1 receptors regulate Bcl-2 expression by reactive oxygen species-dependent transcriptional regulation of nuclear factor kappaB. J Pharmacol Exp Ther 332:388-397. 174

Mishina M, Ohyama M, Ishii K, Kitamura S, Kimura Y, Oda K, Kawamura K, Sasaki T, Kobayashi S, Katayama Y, Ishiwata K (2008) Low density of sigma1 receptors in early Alzheimer's disease. Ann Nucl Med 22:151-156. Molnar Z, Soos K, Lengyel I, Penke B, Szegedi V, Budai D (2004) Enhancement of NMDA responses by beta-amyloid peptides in the hippocampus in vivo. Neuroreport 15:1649- 1652. Monassier L, Bousquet P (2002) Sigma receptors: from discovery to highlights of their implications in the cardiovascular system. Fundam Clin Pharmacol 16:1-8. Morgan D (2009) The role of microglia in antibody-mediated clearance of amyloid-beta from the brain. CNS Neurol Disord Drug Targets 8:7-15. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB (2015) Heart disease and stroke statistics--2015 update: a report from the American Heart Association. Circulation 131:e29-322. Mueller BH, 2nd, Park Y, Daudt DR, 3rd, Ma HY, Akopova I, Stankowska DL, Clark AF, Yorio T (2013) Sigma-1 receptor stimulation attenuates calcium influx through activated L-type Voltage Gated Calcium Channels in purified retinal ganglion cells. Exp Eye Res 107:21- 31. Muir KW (2006) Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Curr Opin Pharmacol 6:53-60. Munoz Maniega S, Cvoro V, Chappell FM, Armitage PA, Marshall I, Bastin ME, Wardlaw JM (2008) Changes in NAA and lactate following ischemic stroke: a serial MR spectroscopic imaging study. Neurology 71:1993-1999. Nagele RG, D'Andrea MR, Anderson WJ, Wang HY (2002) Intracellular accumulation of beta- amyloid(1-42) in neurons is facilitated by the alpha 7 nicotinic in Alzheimer's disease. Neuroscience 110:199-211. Naj AC, Jun G, Beecham GW, Wang LS, Vardarajan BN, Buros J, Gallins PJ, Buxbaum JD, Jarvik GP, Crane PK, Larson EB, Bird TD, Boeve BF, Graff-Radford NR, De Jager PL, Evans D, Schneider JA, Carrasquillo MM, Ertekin-Taner N, Younkin SG, Cruchaga C, Kauwe JS, Nowotny P, Kramer P, Hardy J, Huentelman MJ, Myers AJ, Barmada MM, Demirci FY, Baldwin CT, Green RC, Rogaeva E, St George-Hyslop P, Arnold SE, Barber R, Beach T, Bigio EH, Bowen JD, Boxer A, Burke JR, Cairns NJ, Carlson CS, Carney RM, Carroll SL, Chui HC, Clark DG, Corneveaux J, Cotman CW, Cummings JL, DeCarli C, DeKosky ST, Diaz-Arrastia R, Dick M, Dickson DW, Ellis WG, Faber KM, Fallon KB, Farlow MR, Ferris S, Frosch MP, Galasko DR, Ganguli M, Gearing M, Geschwind DH, Ghetti B, Gilbert JR, Gilman S, Giordani B, Glass JD, Growdon JH, Hamilton RL, Harrell LE, Head E, Honig LS, Hulette CM, Hyman BT, Jicha GA, Jin LW, Johnson N, Karlawish J, Karydas A, Kaye JA, Kim R, Koo EH, Kowall NW, Lah JJ, Levey AI, Lieberman AP, Lopez OL, Mack WJ, Marson DC, Martiniuk F, Mash DC, Masliah E, McCormick WC, McCurry SM, McDavid AN, McKee AC, Mesulam M, Miller BL, Miller CA, Miller JW, Parisi JE, Perl DP, Peskind E, Petersen RC, Poon WW, Quinn JF, Rajbhandary RA, Raskind M, Reisberg B, Ringman JM, Roberson ED, Rosenberg RN, Sano M, Schneider LS, Seeley W, Shelanski ML, Slifer MA, Smith CD, Sonnen JA, Spina S, Stern RA, Tanzi RE, Trojanowski JQ, Troncoso JC, Van Deerlin VM, Vinters HV, Vonsattel JP, Weintraub S, Welsh-Bohmer KA, Williamson J, Woltjer RL, Cantwell LB, Dombroski BA, Beekly D, Lunetta KL, Martin ER, Kamboh MI, Saykin AJ, Reiman EM, Bennett DA, Morris JC, 175

Montine TJ, Goate AM, Blacker D, Tsuang DW, Hakonarson H, Kukull WA, Foroud TM, Haines JL, Mayeux R, Pericak-Vance MA, Farrer LA, Schellenberg GD (2011) Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet 43:436-441. Nakazawa M, Matsuno K, Mita S (1998) Activation of sigma1 receptor subtype leads to neuroprotection in the rat primary neuronal cultures. Neurochem Int 32:337-343. Nelson PT, Pious NM, Jicha GA, Wilcock DM, Fardo DW, Estus S, Rebeck GW (2013) APOE- epsilon2 and APOE-epsilon4 correlate with increased amyloid accumulation in cerebral vasculature. J Neuropathol Exp Neurol 72:708-715. Nery AA, Magdesian MH, Trujillo CA, Sathler LB, Juliano MA, Juliano L, Ulrich H, Ferreira ST (2013) Rescue of amyloid-Beta-induced inhibition of nicotinic acetylcholine receptors by a peptide homologous to the nicotine binding domain of the alpha 7 subtype. PLoS One 8:e67194. Octave JN, Pierrot N, Ferao Santos S, Nalivaeva NN, Turner AJ (2013) From synaptic spines to nuclear signaling: nuclear and synaptic actions of the amyloid precursor protein. J Neurochem 126:183-190. Oltvai ZN, Milliman CL, Korsmeyer SJ (1993) Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 74:609-619. Orellana JA, Shoji KF, Abudara V, Ezan P, Amigou E, Saez PJ, Jiang JX, Naus CC, Saez JC, Giaume C (2011) Amyloid beta-induced death in neurons involves glial and neuronal hemichannels. J Neurosci 31:4962-4977. Oseki KT, Monteforte PT, Pereira GJ, Hirata H, Ureshino RP, Bincoletto C, Hsu YT, Smaili SS (2014) Apoptosis induced by Abeta25-35 peptide is Ca(2+) -IP3 signaling-dependent in murine astrocytes. Eur J Neurosci 40:2471-2478. Pacheco-Quinto J, Eckman EA (2013) Endothelin-converting enzymes degrade intracellular beta-amyloid produced within the endosomal/lysosomal pathway and autophagosomes. J Biol Chem 288:5606-5615. Pal A, Fontanilla D, Gopalakrishnan A, Chae YK, Markley JL, Ruoho AE (2012) The sigma-1 receptor protects against cellular oxidative stress and activates antioxidant response elements. Eur J Pharmacol 682:12-20. Pal A, Hajipour AR, Fontanilla D, Ramachandran S, Chu UB, Mavlyutov T, Ruoho AE (2007) Identification of regions of the sigma-1 receptor ligand binding site using a novel photoprobe. Mol Pharmacol 72:921-933. Paradis E, Douillard H, Koutroumanis M, Goodyer C, LeBlanc A (1996) Amyloid beta peptide of Alzheimer's disease downregulates Bcl-2 and upregulates bax expression in human neurons. J Neurosci 16:7533-7539. Parks JK, Smith TS, Trimmer PA, Bennett JP, Jr., Parker WD, Jr. (2001) Neurotoxic Abeta peptides increase oxidative stress in vivo through NMDA-receptor and nitric-oxide- synthase mechanisms, and inhibit complex IV activity and induce a mitochondrial permeability transition in vitro. J Neurochem 76:1050-1056. Patel JR, Brewer GJ (2008) Age-related differences in NFkappaB translocation and Bcl-2/Bax ratio caused by TNFalpha and Abeta42 promote survival in middle-age neurons and death in old neurons. Exp Neurol 213:93-100. Pendlebury ST, Rothwell PM (2009) Prevalence, incidence, and factors associated with pre- stroke and post-stroke dementia: a systematic review and meta-analysis. Lancet Neurol 8:1006-1018. Pluta R, Furmaga-Jablonska W, Maciejewski R, Ulamek-Koziol M, Jablonski M (2013) Brain ischemia activates beta- and gamma-secretase cleavage of amyloid precursor protein: significance in sporadic Alzheimer's disease. Mol Neurobiol 47:425-434. 176

Pluta R, Ulamek M, Jablonski M (2009) Alzheimer's mechanisms in ischemic brain degeneration. Anat Rec (Hoboken) 292:1863-1881. Popa-Wagner A, Schroder E, Walker LC, Kessler C (1998) beta-Amyloid precursor protein and ss-amyloid peptide immunoreactivity in the rat brain after middle cerebral artery occlusion: effect of age. Stroke 29:2196-2202. Puzzo D, Arancio O (2013) Amyloid-beta peptide: Dr. Jekyll or Mr. Hyde? J Alzheimers Dis 33 Suppl 1:S111-120. Quirion R, Bowen WD, Itzhak Y, Junien JL, Musacchio JM, Rothman RB, Su TP, Tam SW, Taylor DP (1992) A proposal for the classification of sigma binding sites. Trends Pharmacol Sci 13:85-86. Rae J, Cooper K, Gates P, Watsky M (1991) Low access resistance perforated patch recordings using amphotericin B. J Neurosci Methods 37:15-26. Reddy NL, Hu LY, Cotter RE, Fischer JB, Wong WJ, McBurney RN, Weber E, Holmes DL, Wong ST, Prasad R, et al. (1994) Synthesis and structure-activity studies of N,N'- diarylguanidine derivatives. N-(1-naphthyl)-N'-(3-ethylphenyl)-N'-methylguanidine: a new, selective noncompetitive NMDA receptor antagonist. J Med Chem 37:260-267. Revett TJ, Baker GB, Jhamandas J, Kar S (2013) Glutamate system, amyloid ss peptides and tau protein: functional interrelationships and relevance to Alzheimer disease pathology. J Psychiatry Neurosci 38:6-23. Rezende TM, Vieira LQ, Cardoso FP, Oliveira RR, de Oliveira Mendes ST, Jorge ML, Ribeiro Sobrinho AP (2007) The effect of mineral trioxide aggregate on phagocytic activity and production of reactive oxygen, nitrogen species and arginase activity by M1 and M2 macrophages. Int Endod J 40:603-611. Rogove AD, Lu W, Tsirka SE (2002) Microglial activation and recruitment, but not proliferation, suffice to mediate neurodegeneration. Cell Death Differ 9:801-806. Ruscher K, Shamloo M, Rickhag M, Ladunga I, Soriano L, Gisselsson L, Toresson H, Ruslim- Litrus L, Oksenberg D, Urfer R, Johansson BB, Nikolich K, Wieloch T (2011) The sigma-1 receptor enhances brain plasticity and functional recovery after experimental stroke. Brain 134:732-746. Rybczynska AA, Dierckx RA, Ishiwata K, Elsinga PH, van Waarde A (2008) Cytotoxicity of sigma-receptor ligands is associated with major changes of cellular metabolism and complete occupancy of the sigma-2 subpopulation. J Nucl Med 49:2049-2056. Salamone A, Mura E, Zappettini S, Grilli M, Olivero G, Preda S, Govoni S, Marchi M (2014) Inhibitory effects of beta-amyloid on the nicotinic receptors which stimulate glutamate release in rat hippocampus: the glial contribution. Eur J Pharmacol 723:314-321. Samuelsson M, Soderfeldt B, Olsson GB (1996) Functional outcome in patients with lacunar infarction. Stroke 27:842-846. Satou T, Cummings BJ, Cotman CW (1995) Immunoreactivity for Bcl-2 protein within neurons in the Alzheimer's disease brain increases with disease severity. Brain Res 697:35-43. Schelman WR, Andres RD, Sipe KJ, Kang E, Weyhenmeyer JA (2004) Glutamate mediates cell death and increases the Bax to Bcl-2 ratio in a differentiated neuronal cell line. Brain Res Mol Brain Res 128:160-169. Scherz MW, Fialeix M, Fischer JB, Reddy NL, Server AC, Sonders MS, Tester BC, Weber E, Wong ST, Keana JF (1990) Synthesis and structure-activity relationships of N,N'-di-o- tolylguanidine analogues, high-affinity ligands for the haloperidol-sensitive sigma receptor. J Med Chem 33:2421-2429. Schetz JA, Perez E, Liu R, Chen S, Lee I, Simpkins JW (2007) A prototypical Sigma-1 receptor antagonist protects against brain ischemia. Brain Res 1181:1-9.

177

Schubert D, Behl C, Lesley R, Brack A, Dargusch R, Sagara Y, Kimura H (1995) Amyloid peptides are toxic via a common oxidative mechanism. Proc Natl Acad Sci U S A 92:1989-1993. Selkoe DJ (2001) Alzheimer's disease: genes, proteins, and therapy. Physiol Rev 81:741-766. Selznick LA, Holtzman DM, Han BH, Gokden M, Srinivasan AN, Johnson EM, Jr., Roth KA (1999) In situ immunodetection of neuronal caspase-3 activation in Alzheimer disease. J Neuropathol Exp Neurol 58:1020-1026. Seminerio MJ, Robson MJ, Abdelazeem AH, Mesangeau C, Jamalapuram S, Avery BA, McCurdy CR, Matsumoto RR (2012) Synthesis and pharmacological characterization of a novel sigma receptor ligand with improved metabolic stability and antagonistic effects against methamphetamine. AAPS J 14:43-51. Seredenin SB, Antipova TA, Voronin MV, Kurchashova SY, Kuimov AN (2009) Interaction of afobazole with sigma1-receptors. Bull Exp Biol Med 148:42-44. Seredenin SB, Romanova GA, Shakova FM (2008) Neuroprotective effect of afobazole on rats with bilateral local photothrombosis of vessels in the prefrontal cortex. Bull Exp Biol Med 145:207-209. Seredenin SB, Voronin MV (2009) [Neuroreceptor mechanisms of the afobazole effect]. Eksp Klin Farmakol 72:3-11. Seth P, Fei YJ, Li HW, Huang W, Leibach FH, Ganapathy V (1998) Cloning and functional characterization of a sigma receptor from rat brain. J Neurochem 70:922-931. Shang YC, Chong ZZ, Wang S, Maiese K (2012) Prevention of beta-amyloid degeneration of microglia by erythropoietin depends on Wnt1, the PI 3-K/mTOR pathway, Bad, and Bcl- xL. Aging (Albany NY) 4:187-201. Shen MY, Hsiao G, Fong TH, Chen HM, Chou DS, Lin CH, Sheu JR, Hsu CY (2008) Amyloid beta peptide-activated signal pathways in human platelets. Eur J Pharmacol 588:259- 266. Shih AY, Blinder P, Tsai PS, Friedman B, Stanley G, Lyden PD, Kleinfeld D (2013) The smallest stroke: occlusion of one penetrating vessel leads to infarction and a cognitive deficit. Nat Neurosci 16:55-63. Shiota S, Takekawa H, Matsumoto SE, Takeda K, Nurwidya F, Yoshioka Y, Takahashi F, Hattori N, Tabira T, Mochizuki H, Takahashi K (2013) Chronic Intermittent Hypoxia/Reoxygenation Facilitate Amyloid-beta Generation in Mice. J Alzheimers Dis 37:325-333. Silkina IV, Zenina TA, Seredenin SB, Mirzoian RS (2006) [Effect of afobazole on the accumulation of free radical oxidation products and the catalase activity in rats with cerebral ischemia]. Eksp Klin Farmakol 69:47-50. Small DH, Maksel D, Kerr ML, Ng J, Hou X, Chu C, Mehrani H, Unabia S, Azari MF, Loiacono R, Aguilar MI, Chebib M (2007) The beta-amyloid protein of Alzheimer's disease binds to membrane lipids but does not bind to the alpha7 nicotinic acetylcholine receptor. J Neurochem 101:1527-1538. Smith KJ, Butler TR, Self RL, Braden BB, Prendergast MA (2008) Potentiation of N-methyl-D- aspartate receptor-mediated neuronal injury during methamphetamine withdrawal in vitro requires co-activation of IP3 receptors. Brain Res 1187:67-73. Solito E, Sastre M (2012) Microglia function in Alzheimer's disease. Front Pharmacol 3:14. Stephenson DT, Rash K, Clemens JA (1992) Amyloid precursor protein accumulates in regions of neurodegeneration following focal cerebral ischemia in the rat. Brain Res 593:128-135. Streit WJ, Braak H, Xue QS, Bechmann I (2009) Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer's disease. Acta Neuropathol 118:475-485. 178

Streit WJ, Mrak RE, Griffin WS (2004) Microglia and neuroinflammation: a pathological perspective. J Neuroinflammation 1:14. Su JH, Deng G, Cotman CW (1997) Bax protein expression is increased in Alzheimer's brain: correlations with DNA damage, Bcl-2 expression, and brain pathology. J Neuropathol Exp Neurol 56:86-93. Su TP, Hayashi T (2003) Understanding the molecular mechanism of sigma-1 receptors: towards a hypothesis that sigma-1 receptors are intracellular amplifiers for signal transduction. Curr Med Chem 10:2073-2080. Su TP, Hayashi T, Maurice T, Buch S, Ruoho AE (2010) The sigma-1 receptor chaperone as an inter-organelle signaling modulator. Trends Pharmacol Sci 31:557-566. Su TP, Hayashi T, Vaupel DB (2009) When the endogenous hallucinogenic trace amine N,N- dimethyltryptamine meets the sigma-1 receptor. Sci Signal 2:pe12. Sudduth TL, Powell DK, Smith CD, Greenstein A, Wilcock DM (2013) Induction of hyperhomocysteinemia models vascular dementia by induction of cerebral microhemorrhages and neuroinflammation. J Cereb Blood Flow Metab 33:708-715. Sudduth TL, Weekman EM, Brothers HM, Braun K, Wilcock DM (2014) beta-amyloid deposition is shifted to the vasculature and memory impairment is exacerbated when hyperhomocysteinemia is induced in APP/PS1 transgenic mice. Alzheimers Res Ther 6:32. Takano T, Oberheim N, Cotrina ML, Nedergaard M (2009) Astrocytes and ischemic injury. Stroke 40:S8-12. Talantova M, Sanz-Blasco S, Zhang X, Xia P, Akhtar MW, Okamoto S, Dziewczapolski G, Nakamura T, Cao G, Pratt AE, Kang YJ, Tu S, Molokanova E, McKercher SR, Hires SA, Sason H, Stouffer DG, Buczynski MW, Solomon JP, Michael S, Powers ET, Kelly JW, Roberts A, Tong G, Fang-Newmeyer T, Parker J, Holland EA, Zhang D, Nakanishi N, Chen HS, Wolosker H, Wang Y, Parsons LH, Ambasudhan R, Masliah E, Heinemann SF, Pina-Crespo JC, Lipton SA (2013) Abeta induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc Natl Acad Sci U S A 110:E2518-2527. Tchedre KT, Huang RQ, Dibas A, Krishnamoorthy RR, Dillon GH, Yorio T (2008) Sigma-1 receptor regulation of voltage-gated calcium channels involves a direct interaction. Invest Ophthalmol Vis Sci 49:4993-5002. Tchedre KT, Yorio T (2008) sigma-1 receptors protect RGC-5 cells from apoptosis by regulating intracellular calcium, Bax levels, and caspase-3 activation. Invest Ophthalmol Vis Sci 49:2577-2588. Texido L, Martin-Satue M, Alberdi E, Solsona C, Matute C (2011) Amyloid beta peptide oligomers directly activate NMDA receptors. Cell Calcium 49:184-190. Torrence-Campbell C, Bowen WD (1996) Differential solubilization of rat liver sigma 1 and sigma 2 receptors: retention of sigma 2 sites in particulate fractions. Eur J Pharmacol 304:201- 210. Tsai SY, Hayashi T, Mori T, Su TP (2009) Sigma-1 receptor chaperones and diseases. Cent Nerv Syst Agents Med Chem 9:184-189. Uchida N, Ujike H, Tanaka Y, Sakai A, Yamamoto M, Fujisawa Y, Kanzaki A, Kuroda S (2005) A variant of the sigma receptor type-1 gene is a protective factor for Alzheimer disease. Am J Geriatr Psychiatry 13:1062-1066. Vagnerova K, Hurn PD, Bhardwaj A, Kirsch JR (2006) Sigma 1 receptor agonists act as neuroprotective drugs through inhibition of inducible nitric oxide synthase. Anesth Analg 103:430-434, table of contents.

179

Varadarajan S, Kanski J, Aksenova M, Lauderback C, Butterfield DA (2001) Different mechanisms of oxidative stress and neurotoxicity for Alzheimer's A beta(1--42) and A beta(25--35). J Am Chem Soc 123:5625-5631. Vilner BJ, Bowen WD (2000) Modulation of cellular calcium by sigma-2 receptors: release from intracellular stores in human SK-N-SH neuroblastoma cells. J Pharmacol Exp Ther 292:900-911. Walker JM, Bowen WD, Walker FO, Matsumoto RR, De Costa B, Rice KC (1990) Sigma receptors: biology and function. Pharmacol Rev 42:355-402. Weinstein JR, Koerner IP, Moller T (2010) Microglia in ischemic brain injury. Future Neurol 5:227-246. Weintraub MI (2006) Thrombolysis (tissue plasminogen activator) in stroke: a medicolegal quagmire. Stroke 37:1917-1922. Weldon DT, Rogers SD, Ghilardi JR, Finke MP, Cleary JP, O'Hare E, Esler WP, Maggio JE, Mantyh PW (1998) Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J Neurosci 18:2161-2173. Wen Y, Yang S, Liu R, Simpkins JW (2004) Transient cerebral ischemia induces site-specific hyperphosphorylation of tau protein. Brain Res 1022:30-38. Wilcock DM, Munireddy SK, Rosenthal A, Ugen KE, Gordon MN, Morgan D (2004) Microglial activation facilitates Abeta plaque removal following intracranial anti-Abeta antibody administration. Neurobiol Dis 15:11-20. Wilcock DM, Vitek MP, Colton CA (2009) Vascular amyloid alters astrocytic water and potassium channels in mouse models and humans with Alzheimer's disease. Neuroscience 159:1055-1069. Wolfe SA, Jr., De Souza EB (1993) Sigma and receptors in the brain-endocrine- immune axis. NIDA Res Monogr 133:95-123. Xiong ZG, Zhu XM, Chu XP, Minami M, Hey J, Wei WL, MacDonald JF, Wemmie JA, Price MP, Welsh MJ, Simon RP (2004) Neuroprotection in ischemia: blocking calcium-permeable acid-sensing ion channels. Cell 118:687-698. Xu J, Zeng C, Chu W, Pan F, Rothfuss JM, Zhang F, Tu Z, Zhou D, Zeng D, Vangveravong S, Johnston F, Spitzer D, Chang KC, Hotchkiss RS, Hawkins WG, Wheeler KT, Mach RH (2011) Identification of the PGRMC1 protein complex as the putative sigma-2 receptor binding site. Nat Commun 2:380. Yang F, Sun X, Beech W, Teter B, Wu S, Sigel J, Vinters HV, Frautschy SA, Cole GM (1998) Antibody to caspase-cleaved actin detects apoptosis in differentiated neuroblastoma and plaque-associated neurons and microglia in Alzheimer's disease. Am J Pathol 152:379- 389. Yang S, Bhardwaj A, Cheng J, Alkayed NJ, Hurn PD, Kirsch JR (2007) Sigma receptor agonists provide neuroprotection in vitro by preserving bcl-2. Anesth Analg 104:1179-1184, tables of contents. Yang ZJ, Carter EL, Torbey MT, Martin LJ, Koehler RC (2010) Sigma receptor ligand 4-phenyl-1- (4-phenylbutyl)-piperidine modulates neuronal nitric oxide synthase/postsynaptic density- 95 coupling mechanisms and protects against neonatal ischemic degeneration of striatal neurons. Exp Neurol 221:166-174. Yao M, Nguyen TV, Pike CJ (2005) Beta-amyloid-induced neuronal apoptosis involves c-Jun N- terminal kinase-dependent downregulation of Bcl-w. J Neurosci 25:1149-1158. Zeng C, Vangveravong S, Xu J, Chang KC, Hotchkiss RS, Wheeler KT, Shen D, Zhuang ZP, Kung HF, Mach RH (2007) Subcellular localization of sigma-2 receptors in breast cancer cells using two-photon and confocal microscopy. Cancer Res 67:6708-6716. 180

Zenina TA, Gavrish IV, Melkumyan DS, Seredenina TS, Seredenin SB (2005) Neuroprotective properties of afobazol in vitro. Bull Exp Biol Med 140:194-196. Zhang F, Eckman C, Younkin S, Hsiao KK, Iadecola C (1997) Increased susceptibility to ischemic brain damage in transgenic mice overexpressing the amyloid precursor protein. J Neurosci 17:7655-7661. Zhang H, Christopher K, Cuevas J (2009) Sigma Receptor Activation Inhibits Voltage-Gated Sodium Channels in Rat Intracardiac Ganglion Neurons. Int J Physiol Pathophysiol Pharmacol 2:1-11. Zhang H, Cuevas J (2002) Sigma receptors inhibit high-voltage-activated calcium channels in rat sympathetic and parasympathetic neurons. J Neurophysiol 87:2867-2879. Zhang H, Cuevas J (2005) sigma Receptor activation blocks potassium channels and depresses neuroexcitability in rat intracardiac neurons. J Pharmacol Exp Ther 313:1387-1396. Zhang Y, Shi Y, Qiao L, Sun Y, Ding W, Zhang H, Li N, Chen D (2012) Sigma-1 receptor agonists provide neuroprotection against gp120 via a change in bcl-2 expression in mouse neuronal cultures. Brain Res 1431:13-22. Zhang YW, Thompson R, Zhang H, Xu H (2011) APP processing in Alzheimer's disease. Mol Brain 4:3.

181

APPENDICES

Permissions to Reprint Previously Published Materials

182