+ All Categories
Home > Documents > Protein-Protein Interactions Provide a Platform for G ......Kai Ying Lai Doctoral of Philosophy...

Protein-Protein Interactions Provide a Platform for G ......Kai Ying Lai Doctoral of Philosophy...

Date post: 16-Feb-2021
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
212
Protein-Protein Interactions Provide a Platform for G- Protein-Coupled Receptors to Exert Physiological and Pathological Functions in Central Nervous System: Therapeutic Potential? By Kai Ying Lai A thesis submitted in conformity with the requirements for the degree of Doctoral of Philosophy Department of Physiology University of Toronto © Copyright by Kai Ying Lai 2019
Transcript
  • Protein-Protein Interactions Provide a Platform for G-Protein-Coupled Receptors to Exert Physiological and

    Pathological Functions in Central Nervous System: Therapeutic Potential?

    By

    Kai Ying Lai

    A thesis submitted in conformity with the requirements for the degree of Doctoral of Philosophy

    Department of Physiology University of Toronto

    © Copyright by Kai Ying Lai 2019

  • ii

    Protein-Protein Interactions Provide a Platform for G-Protein-

    Coupled Receptors to Exert Physiological and Pathological

    Functions in Central Nervous System: Therapeutic Potential?

    Kai Ying Lai

    Doctoral of Philosophy

    Department of Physiology

    University of Toronto

    2019

    Abstract

    In the central nervous system membrane receptors play an important role in neuronal

    communication. Since neurons mainly communicate through the release of neurotransmitters,

    membrane receptors are responsible for binding neurotransmitters and transducing their message

    into an intracellular response. In general, there are two types of membrane receptors: ionotropic

    and metabotropic. Ionotropic receptors are ligand-gated ion channels, which are responsible for

    mediating fast synaptic responses. On the other hand, metabotropic receptors act through their

    downstream signaling cascades to trigger intracellular responses.

  • iii

    A prominent example of metabotropic receptors is the G-protein-coupled receptor

    (GPCR) superfamily. In the human body this receptor superfamily consists of approximately 799

    receptors. Conventionally, these GPCRs are seven-transmembrane-domain receptors which act

    through their coupled G-proteins to elicit their physiological actions. There is also a growing

    body of evidence that these GPCRs can physically interact with each other as well as with other

    types of membrane receptors.

    This present study focuses on examining whether protein-protein interactions, other than

    coupled G-protein signaling cascades, are another platform for GPCRs to exert their effects or to

    regulate the functions of other proteins. We also investigate whether these protein-protein

    interactions involving GPCRs present any physiological or pathological implications.

  • iv

    Acknowledgments

    I would like to take this opportunity to thank the people who have helped me throughout

    my Ph.D. studies. Foremost, I would like to thank my supervisor, Dr. Fang Liu, for her support

    and guidance because without her, none of this work would be possible.

    I would also like to thank my advisory committee: Dr. Paul Fletcher, Dr. Sheena Josselyn

    and Dr. Albert Wong, who have consistently provided guidance and insightful inputs to me

    experiments and data analyses.

    I also want to take this opportunity to thank the people who I had the great honor to work

    closely with during my Ph.D. research:

    Dr. Ping Su --- for teaching me all the valuable biochemical techniques and giving me the

    previous opportunities to collaborate on several projects.

    Dr. Dongxu Zhai --- for not only helping me purchase the items my research required, but also

    performing the animal surgeries to make my research on mGluR1-NMDAR protein complex

    possible.

    Dr. Andrew Abela (from Dr. Paul Fletcher’s lab) --- for training me to use the touchscreen boxes

    and working with me to interpret the massive amount of data.

  • v

    Special thanks to the past and present lab members and members from other research labs:

    Dr. Shupeng Li, Dr. Hailong Zhang, Dr. Haiyin Li, Mr. Anlong Jiang, Mr. Jay Boychuk, Dr.

    Laura Feldcamp, Mr. Roger Raymond, and many more for making our research facility a fun

    studying and working place.

    Dr. Frankie Lee and Mr. Charlie Campbell for being the best people to share office with.

    Ms Lori Dixon, Ms Katrina Deverell and all the staff of the animal facility: for their support and

    assistance with my animal studies.

    Last but not least: I sincerely thank my parents, my sister and other family members for their

    unconditional love and support throughout my graduate studies.

  • vi

    Table of Contents

    Acknowledgments.......................................................................................................................... iv

    Table of Contents ........................................................................................................................... vi

    List of Tables ................................................................................................................................ xii

    List of Figures .............................................................................................................................. xiii

    List of Abbreviations .....................................................................................................................xv

    Chapter 1 Introduction .....................................................................................................................1

    Introduction .................................................................................................................................1

    1.1 Neuronal Communication within the Central Nervous System ...........................................1

    1.2 Types of Postsynaptic Membrane Receptors --- ionotropic vs metabotropic receptors ......2

    1.3 G-Protein-Coupled Receptors ..............................................................................................4

    1.4 Diversity of GPCR ...............................................................................................................5

    1.4.1 The Rhodopsin receptor family/Class A GPCR ......................................................6

    1.4.2 The Secretin receptor family/Class B GPCR ...........................................................7

    1.4.3 The Adhesion receptor family/Class B GPCR.........................................................8

    1.4.4 The Glutamate Receptor Family/ Class C GPCR ....................................................8

    1.4.5 The Frizzled Receptor Family/ Class F GPCR ........................................................9

    1.5 GPCR Dimerization .............................................................................................................9

    1.6 Protein-Protein Interaction on signaling cascades .............................................................10

    1.6.1 Interaction Between Members of the Same Receptor Subfamily ..........................10

    1.6.2 Interaction Between Members of Different Receptor Subfamilies ........................12

    1.6.3 Interaction between a GPCR and a non-GPCR .....................................................13

    1.7 Rationale and Hypothesis ..................................................................................................15

    Chapter 2 ........................................................................................................................................18

  • vii

    Metabotropic Glutamate Receptor 1 Modulates the Excitotoxic Functions of N-Methyl-D-

    Aspartate Receptor (NMDAR) through its Interaction with the GluN2A subunit of

    NMDAR ....................................................................................................................................18

    Metabotropic Glutamate Receptor 1 Modulates the Excitotoxic Functions of N-Methyl-D-Aspartate Receptor (NMDAR) through its Interaction with the GluN2A subunit of

    NMDAR ....................................................................................................................................19

    2.1 Introduction ........................................................................................................................19

    2.1.1 Glutamate Neurotransmission ................................................................................19

    2.1.2 The Implication of Glutamate in Ischemic Stroke .................................................20

    2.1.3 N-methyl-D-aspartate receptor (NMDAR) ............................................................21

    2.1.4 Metabotropic Glutamate Receptor 1 (mGluR1) ....................................................21

    2.1.5 Rationale and Hypothesis: Functional/Physical Cross-Talk between mGluR1 and NMDAR and Their Implications in Ischemic Stroke .....................................22

    2.2 Materials and Methods .......................................................................................................23

    2.2.1 Co-immunoprecipitation ........................................................................................23

    2.2.2 GST Protein Affinity Purification ..........................................................................24

    2.2.3 Western Blotting ....................................................................................................24

    2.2.4 GST Fusion Protein Constructs .............................................................................24

    2.2.5 GST Fusion Protein Expression and Purification ..................................................25

    2.2.6 Primary Mouse Hippocampal Culture ...................................................................26

    2.2.7 NMDA-Excitotoxicity in Primary Mouse Hippocampal Culture and Confocal Imaging ..................................................................................................................27

    2.2.8 Animals ..................................................................................................................28

    2.2.9 Transient Middle Cerebral Artery Occlusion (tMCAO) ........................................28

    2.2.10 Intracerebroventricular (ICV) Peptide Delivery ....................................................28

    2.2.11 Neurological Assessment .......................................................................................29

    2.2.12 Tetrazolium Chloride (TTC) Staining and Brain Infarction Analysis ...................29

    2.2.13 Acute Brain Slice Treatment ..................................................................................29

    2.2.14 NMDA-Stimulated ERK1/2 Phosphorylation .......................................................30

  • viii

    2.2.15 Membrane Expression of NMDA Receptors .........................................................30

    2.2.16 Statistical Analysis .................................................................................................31

    2.3 Results ................................................................................................................................31

    2.3.1 GluN2A subunit of NMDAR facilitates mGluR1-NMDAR complex formation ..31

    2.3.2 Identification of the amino acid sequence that enables the mGluR1 to form a complex with NMDAR ..........................................................................................31

    2.3.3 The development of interfering peptides to disrupt the mGluR1-NMDAR interaction ..............................................................................................................33

    2.3.4 The disruption of mGluR1-NMDAR interaction attenuated NMDA-mediated excitotoxicity..........................................................................................................37

    2.3.5 The neuroprotective effects of mGluR1-NMDAR disruption in animal model

    of ischemic stroke ..................................................................................................40

    2.3.6 The disruption of mGluR1-NMDAR complex abolished NMDA-mediated ERK1/2 Phosphorylation .......................................................................................47

    2.3.7 The mGluR1-NMDAR interaction does not regulate NMDAR membrane

    expression ..............................................................................................................49

    2.4 Discussion ..........................................................................................................................51

    Chapter 3 ........................................................................................................................................58

    Prenatal Disruption of D1R-SynGAP Complex Impairs GABAergic Interneuron Migration &

    Causes Behavioural Deficits in Adulthood ...............................................................................58

    Prenatal Disruption of D1R-SynGAP Complex Impairs GABAergic Interneuron Migration & Causes Behavioural Deficits in Adulthood ...........................................................................59

    3.1 Introduction ........................................................................................................................59

    3.1.1 γ-aminobutyric acid (GABA) ................................................................................59

    3.1.2 GABAergic Interneurons .......................................................................................59

    3.1.3 Morphology and Synaptic Connectivity of GABAergic Interneurons ..................60

    3.1.4 Biochemical Markers and Electrophysiological Properties of GABAergic Interneurons ...........................................................................................................62

    3.1.5 Pathological Significance of GABAergic Interneuron Dysfunction .....................64

    3.1.6 Dopaminergic System on GABAergic Interneurons .............................................65

  • ix

    3.1.7 Rationale and Hypothesis ......................................................................................66

    3.2 Materials and Methods .......................................................................................................67

    3.2.1 Drugs and Peptides ................................................................................................67

    3.2.2 Co-immunoprecipitation and GST affinity pull-down ..........................................67

    3.2.3 GST fusion protein constructs ...............................................................................68

    3.2.4 HEK293T Cell Culture and DNA Transfection.....................................................69

    3.2.5 cAMP accumulation assay .....................................................................................69

    3.2.6 Surface biotinylation assay ....................................................................................70

    3.2.7 Acute mouse brain slices .......................................................................................70

    3.2.8 Primary culture of MGE-derived neurons .............................................................71

    3.2.9 MGE Matrigel Explant ..........................................................................................71

    3.2.10 Immunohistochemistry and immunocytochemistry ...............................................71

    3.2.11 Analysis of immunohistochemistry .......................................................................73

    3.2.12 Animals ..................................................................................................................73

    3.2.13 Open-Field Locomotor Activity ............................................................................73

    3.2.14 Pre-pulse Inhibition ................................................................................................74

    3.2.15 Sociability Test ......................................................................................................74

    3.2.16 Touchscreen Discrimination Task .........................................................................75

    3.2.17 Responding on different schedules of reinforcement ............................................78

    3.2.18 Statistical analyses .................................................................................................79

    3.3 Results ................................................................................................................................79

    3.3.1 Activation of the D1R facilitates the D1R-SynGAP complex formation ...............79

    3.3.2 Identification of the amino acid sequence that enables the D1R to form a

    complex with SynGAP ..........................................................................................79

    3.3.3 SynGAP upregulates D1R-mediated signaling through the D1R-SynGAP

    interaction. .............................................................................................................83

  • x

    3.3.4 SynGAP enhances D1R cell surface localization through the D1R-SynGAP interaction. .............................................................................................................86

    3.3.5 Disruption of the D1R-SynGAP interaction inhibits GABAergic interneuron migration. ...............................................................................................................89

    3.3.6 Disruption of the D1R-SynGAP complex changes microtubule dynamics ...........95

    3.3.7 Prenatal disruption of the D1R-SynGAP complex leads to more parvalbumin- and calbindin-positive GABAergic interneurons located in the lateral

    neocortex of adult offspring. ................................................................................100

    3.3.8 Disruption of D1R-SynGAP on animal behaviours .............................................103

    3.4 Discussion ........................................................................................................................113

    Chapter 4 ......................................................................................................................................120

    Presynaptic Dopamine D2 Receptor Upregulates Dopamine Reuptake Actions of Dopamine

    Transporter Through Their Physical Interaction .....................................................................120

    Presynaptic Dopamine D2 Receptor Upregulates Dopamine Reuptake Actions of

    Dopamine Transporter Through Their Physical Interaction ...................................................121

    4.1 Introduction ......................................................................................................................121

    4.1.1 The Dopaminergic System ...................................................................................121

    4.1.2 The Four Major Pathways of the Dopaminergic System .....................................122

    4.1.3 Dopamine Receptors ............................................................................................123

    4.1.4 Dopamine Transporter .........................................................................................125

    4.1.5 Rationale and Hypothesis ....................................................................................127

    4.2 Materials and Methods .....................................................................................................131

    4.2.1 Experimental Animals .........................................................................................131

    4.2.2 Commercially-Synthesized Peptides ...................................................................132

    4.2.3 Animal Surgery ....................................................................................................132

    4.2.4 In vivo microdialysis............................................................................................132

    4.2.5 High Performance Liquid Chromatography (HPLC) ..........................................133

    4.2.6 Peptide-Induced Locomotor Activity in WKY and SHR Rats ............................133

    4.2.7 Y-Maze Test.........................................................................................................134

  • xi

    4.2.8 Co-immunoprecipitation and Western Blot .........................................................134

    4.2.9 Data Analysis .......................................................................................................135

    4.3 Results ..............................................................................................................................136

    4.3.1 The Effects of D2R-DAT Disruption on Extracellular Dopamine .......................136

    4.3.2 The Effects of TAT-DATNT on Hyperactivity in SHR rats .................................139

    4.3.3 The Effects of TAT-DATNT on Spontaneous Alternation Behaviour (SAB) in

    SHR Rats ..............................................................................................................143

    4.4 Discussion ........................................................................................................................145

    Chapter 5 Conclusion and Future Directions ...............................................................................152

    Conclusion and Future Directions ...........................................................................................152

    5.1 Conclusion .......................................................................................................................152

    5.2 Future Directions .............................................................................................................156

    5.2.1 mGluR1-NMDAR Interaction .............................................................................156

    5.2.2 D1R-SynGAP Interaction .....................................................................................159

    5.2.3 D2R-DAT Interaction ...........................................................................................161

    5.3 Final Thoughts .................................................................................................................162

    References ....................................................................................................................................163

    Copyright Acknowledgements.....................................................................................................194

  • xii

    List of Tables

    Table 3. 1 - Habituation and Pre-training phases prior to the visual touchscreen discrimination

    training. ....................................................................................................................................... 106

  • xiii

    List of Figures

    Figure 1.1 – Schematic Illustration of Synaptic Neurotransmission between Neurons ................. 2

    Figure 1.2 – Structural Representation of a G-Protein-Coupled Receptor (GPCR) ....................... 4

    Figure 2.1 – The mGluR1-NMDAR protein complex is mediated through two sites within the C-

    terminus of mGluR1. .................................................................................................................... 34

    Figure 2.2 – The pretreatment of TAT-mGluR1C1+C4

    protected hippocampal neurons against

    NMDA-excitotoxicity. .................................................................................................................. 39

    Figure 2.3 – The pre-treatment of TAT-mGluR1C1+C4

    reduced brain infarction and improved

    motor recovery in animals subjected to transient middle cerebral occlusion. .............................. 43

    Figure 2.4 – TAT-mGluR1C1+C4

    showed beneficial effects even when administered one hour

    after the onset of ischemia. ........................................................................................................... 46

    Figure 2.5 - The disruption of mGluR1-GluN2A protein complex hindered the NMDAR-

    mediated activation of ERK signaling. ......................................................................................... 48

    Figure 2.6 - The disruption of mGluR1-NMDAR protein complex did not alter the membrane

    expression of NMDAR. ................................................................................................................ 50

    Figure 3.1 - Regulation of D1R-SynGAP Complex Formation .................................................... 82

    Figure 3.2 - SynGAP modulates D1R-mediated signaling............................................................ 85

    Figure 3.3 - SynGAP enhances cell surface membrane localization of D1R ................................ 88

    Figure 3.4 - TAT-fusion peptides can cross both the placenta and blood-brain barrier, and TAT-

    D1Rpep is able to disrupt the D1R-SynGAP interaction. ................................................................ 91

    Figure 3.5 - Disrupted GABAergic interneuron tangential migration in TAT-D1Rpep-injected

    embryonic mice ............................................................................................................................. 94

  • xiv

    Figure 3.6 - TAT-D1Rpep changes neuronal cytoskeleton and decreases MAP2 phosphorylation.

    ....................................................................................................................................................... 99

    Figure 3.7 - Abnormal distribution of both PV- and CB-interneurons in adult offspring from

    TAT-D1Rpep-injected pregnant mice. .......................................................................................... 102

    Figure 3.8 - The disruption of D1R-SynGAP during neurodevelopment causes behavioural

    abnormalities. .............................................................................................................................. 105

    Figure 3.9 - The disruption of D1R-SynGAP during neurodevelopment impairs associative

    learning. ...................................................................................................................................... 110

    Figure 3.10 – The Disruption of D1R-SynGAP does not affect motivational behaviours .......... 112

    Figure 4.1 – The Disruption of D2R-DAT Protein Complex Stimulates Voluntary Movement in

    Sprague-Dawley Rats.................................................................................................................. 128

    Figure 4.2 – The Disruption of D2R-DAT by TAT-DATNT Rescues the Locomotor Impairment

    Imposed by AMPT-Mediated Dopamine Depletion. .................................................................. 131

    Figure 4.3 – The Stimulant Effect of D2R-DAT Disruption is due to the Rise of Extracellular

    Dopamine Level in SD Rats. ...................................................................................................... 138

    Figure 4.4 – TAT-DATNT Alleviates the Hyperactivity of Spontaneously Hypertensive Rats. . 142

    Figure 4.5 –TAT-DATNT at Low Dose Improved Spontaneous Alternation Behaviour in SHR

    Rats. ............................................................................................................................................ 144

  • xv

    List of Abbreviations

    3,5-DHPG 3,5-dihydroxyphenylglycine

    5-HT1AR serotonin 5-HT1A receptor

    5-HT2AR serotonin 5-HT2A receptor

    5-HT3AR serotonin 5-HT3A receptor

    aCSF artificial cerebrospinal fluid

    ADHD attention-deficit hyperactivity disorder

    AMPT α-methyl-p-tyrosine

    ASD autism spectrum disorder

    Ara-C cytarabine

    ATP adenosine triphosphate

    β1-AR β1-adrenoceptor

    BRET bioluminescence resonance energy transfer

    BSA bovine serum albumin

    CB calbindin

    CFP cyan fluorescent protein

    CNS central nervous system

    coIP coimmunoprecipitation

    CR calretinin

  • xvi

    CT carboxyl tail

    D1R dopamine D1 receptor

    D2R dopamine D2 receptor

    D3R dopamine D3 receptor

    D5R dopamine D5 receptor

    DAG diacylglyercol

    DAT dopamine transporter

    DMEM Dulbecco’s Modified Eagle Medium

    DMSO dimethyl sulfoxide

    EL extracellular loop

    ERK1/2 extracellular signal–regulated kinases1/2

    FR fixed ratio

    FRET fluorescence resonance energy transfer

    GABA γ-aminobutyric acid

    GABAAR GABAA receptor

    GAD glutamic acid decarboxylase

    GalR1 galanin receptor-1

    GDP guanosine diphosphate

    GPCR G-protein-coupled receptor

    GST glutathione-S-transferase

  • xvii

    GTP guanosine triphosphate

    HPLC high-performance liquid chromatography

    ICA internal carotid artery

    ICV intracerebroventricular delivery

    IL intracellular loop

    IP intraperitoneal

    IP3 inositol-1,4,5-triphosphate

    IPTG isopropyl β-D-1-thiogalactopyranoside

    LB lysogeny broth

    MAPK mitogen-activated protein kinase

    MGE medial ganglion eminence

    mGluR1 metabotropic glutamate receptor 1

    NAc nucleus accumbens core

    NET norepinephrine transporter

    NMDAR N-methyl-D-aspartate receptors

    P2RY purinergic receptor

    PAR protease-activated receptor

    PBS phosphate-buffered saline

    PFC prefrontal cortex

    PKA protein kinase A

  • xviii

    PPI prepulse inhibition

    PSD-95 postsynaptic density 95

    PR progressive ratio

    PV parvalbumin

    SAB spontaneous alternation behaviour

    SEM standard error of mean

    SERT serotonin transporter

    SHR Spontaneously Hypertensive rat

    SST somatostatin

    SynGAP synaptic Ras GTPase-activating protein

    TfR transferrin receptor

    TM transmembrane domain

    tMCAO transient middle cerebral artery occlusion

    TTC tetrazolium chloride

    VIP vasoactive intestinal peptide

    VTA ventral tegmental area

    WKY Wistar Kyoto rat

    YFP yellow fluorescent protein

  • 1

    Chapter 1 Introduction

    Introduction

    1.1 Neuronal Communication within the Central Nervous System

    Neurons, cells within in the central nervous system (CNS), are different from other cells

    in in the human body because they possess specialized thin branches knowns as dendrites and

    axons. Dendrites allow neurons to receive signals from other neurons, whereas neurons send

    information to neighboring neurons through their axons. This exact process of neurons

    communicating with each other is known as synaptic neurotransmission. Because the cell

    membrane acts as an insulator against direct electrical signaling, neuronal communication must

    be made through chemical signals like neurotransmitters. This exact process of neurons

    communicating with each other is known as synaptic neurotransmission.

    This chemical communication occurs when the dendrite of one neuron and the axon of

    another neuron are brought within 20 to 40 nm of each other, forming the synaptic cleft1. To

    initiate synaptic neurotransmission a neuron must have an action potential traveling down its

    axon terminal, creating an increase in the concentration of intracellular calcium ions. The

    increasing calcium ions cause vesicles filled with neurotransmitters to fuse with the cell

    membrane, triggering the release of neurotransmitters into the synaptic cleft. The

    neurotransmitters then travel through the synaptic cleft and bind to neurotransmitter receptors on

    the surface of adjacent neurons. The neuron releasing the chemical signal is called the

    presynaptic neuron, whereas the neuron that receives and reacts to the released neurotransmitter

    is called the postsynaptic neuron (Figure 1.1).

  • 2

    Figure 1.1 – Schematic Illustration of Synaptic Neurotransmission between Neurons

    The picture depicts the connection between both presynaptic and postsynaptic neurons. The

    presynaptic neuron communicates with the postsynaptic neuron by releasing neurotransmitters

    into the synaptic cleft. The neurotransmitters travel through the cleft and bind to membrane

    receptors on the surface of the postsynaptic neuron to trigger a response.

    1.2 Types of Postsynaptic Membrane Receptors --- ionotropic vs metabotropic receptors

    There are two major classes of neurotransmitter receptors sitting on the dendritic surface

    of postsynaptic neurons: ligand-gated ion channel receptors and metabotropic receptors.

    Ligand-gated ion channel receptors are also known as ionotropic receptors. These

    receptors are integral membrane proteins that contain a pore, which provides a pathway for

    specific ions to flow across the cell membrane. Upon the binding of neurotransmitters to their

  • 3

    orthosteric sites, ligand-gated ion channel receptors undergo conformational changes to open

    their pores for charged ions to pass through and enter the cell1, 2. These receptors are typically

    responsible for mediating fast synaptic neurotransmission on a millisecond time scale.

    Ligand-gated ion channel receptors are generally classified as either excitatory or

    inhibitory, depending on their effects on the postsynaptic neuron3. They are considered

    excitatory when their pores allow positively charged ions (i.e. cations, usually sodium or calcium

    ions). The influx of these positive ions causes the postsynaptic neuron to depolarize, making it

    more likely to fire an action potential. Inhibitory ligand-gated ion channel receptors are only

    permeable to negatively-charged ions (i.e. anions, usually chloride ions). Upon the binding of

    neurotransmitters, these channel receptors will also open their pores, but the influx of chloride

    ions causes postsynaptic neuron hyperpolarization, preventing the subsequent firing of an action

    potential.

    Metabotropic receptors elicit their effects on the postsynaptic neuron through their

    downstream signaling cascades1. Upon the binding of neurotransmitter, metabotropic receptors

    activate proteins that in turn activate “effector” proteins/enzymes. The activated effector

    enzymes subsequently generate second-messenger molecules that can diffuse within a cell to

    elicit effects on a variety of target proteins, greatly changing their activities. Since metabotropic

    receptors undergo several molecular steps to exert their actions rather than by directly opening an

    ion channel pore upon activation, the effects they produce are slower in onset and longer lasting

    than those produced by ionotropic receptors, and range from hundreds of milliseconds to

    minutes3. A classical example of metabotropic receptors is the G-protein-coupled receptor

    superfamily.

  • 4

    1.3 G-Protein-Coupled Receptors

    The G-protein-coupled receptors (GPCRs) are a receptor superfamily which all the

    members share several structural similarities. Every GPCR possesses seven transmembrane

    domains (TM1-7) composed of hydrophobic amino acids which are embedded in the membrane

    in a way that allows the receptor to undergo conformational changes, in addition to an

    extracellular amino terminus and an intracellular carboxyl tail (Figure 1.2)4.

    Figure 1.2 – Structural Representation of a G-Protein-Coupled Receptor (GPCR)

    The diagram illustrates the basic structural similarities shared by members of the GPCR

    superfamily. Along with an extracellular amino terminus and an intracellular carboxyl tail, each

    GPCR possesses seven membrane-spanning domains (TM1-7), which are connected by three

    intracellular loops (IL-3) and three extracellular loops (EL1-3).

  • 5

    G-protein-coupled receptors are coupled to a heterotrimeric guanine nucleotide-binding

    protein, also known as a G-protein. Each G-protein consists of three subunits: Gα, Gβ and Gγ.

    Based on sequence homology, four major G-protein families (Gs/olf, Gi/o, Gq/11 and G12/13) have

    been identified within the human genome5, 6.

    Upon the binding of a neurotransmitter, a GPCR undergoes conformational changes to

    expose its G-protein, promoting the exchange of the bound guanosine diphosphate (GDP) for

    guanosine triphosphate (GTP)7-9. When bound to GDP, the G-protein is in its inactive form as its

    three subunits (i.e. αβγ) remain associated. A neurotransmitter-induced conformational change of

    the GPCR accelerates the dissociation of GDP from its G-protein, which is the rate-limiting step

    in G-protein activation10. Due to the high concentration of GTP inside a cell, the nucleotide-free

    G-protein will quickly bind to any available GTP to facilitate its own activation. Subsequently,

    the displacement of GPD by GTP causes the G-protein to dissociate from the receptor and to

    separate into two parts (α- and βγ- subunits). Both the dissociated α- and βγ- subunits are then

    free to alter the activities of different downstream effector proteins inside the cell, leading to

    changes in cellular biochemistry, physiology and even gene expression. For instance, Gα subunits

    can modulate effectors such as adenylyl cyclase, cGMP phosphodiesterase, phospholipase C,

    etc.11, 12. Whereas, Gβγ subunits recruit G-protein-coupled receptor kinases to the cell membrane

    and regulate a variety of ion channels13, 14. These cellular responses are terminated when the Gα

    subunit reunites with Gβγ upon its complete hydrolysis of GTP.

    1.4 Diversity of GPCR

    More than three decades ago, the rhodopsin receptor was the first G-protein-coupled

    receptor (GPCR) to be structurally identified15, 16. The studies showed that the rhodopsin receptor

    transduces light energy into its transcellular signaling cascades. Since the structural revelation of

  • 6

    the rhodopsin receptor, significant technical advances have been made to unveil more members

    from the GPCR superfamily17. With the growing body of studies on the GPCR superfamily, it

    has become evident that this receptor superfamily can react to a broad range of ligands including

    small organic compounds18, 19, eicosandoids20, peptides21 and proteins22, in addition to light.

    By 2007, the human GPCR superfamily has been found to consist of at least 799 unique

    full-length receptors23. Fredriksson and colleagues further classified these 799 receptors into five

    main families based on phylogenetic criteria: Rhodopsin, Secretin, Adhesion, Glutamate and

    Frizzled24. To avoid possible confusion with specific GPCR receptors (e.g. rhodopsin receptor vs

    Rhodopsin receptor family), these five GPCR family names will be written in italics with an

    initial capital letter in this thesis.

    1.4.1 The Rhodopsin receptor family/Class A GPCR

    Within the GPCR superfamily, the Rhodopsin receptor family has the most members,

    containing approximately 670 full-length receptors23. Structurally, members of the Rhodospin

    receptor family possess short N-termini but diverse transmembrane domains. Given their short

    N-termini, the binding of ligands is primarily mediated by the transmembrane regions and

    extracellular loops 25.

    The Rhodopsin receptor family can respond to a diversity of ligands including peptides,

    amines and purines. Its diverse ligands involve this receptor family in a wide range of

    physiological functions including the senses of vision and olfaction, neuronal signaling, and

    immunological and cardiovascular functions, etc26. Consequently, the Rhodopsin receptor family

    can be further divided into four sub-groups: α, β, γ, and δ24, 27.

  • 7

    The α-group contains at least 18 physiologically important receptors, including the

    histamine receptors, the dopamine receptors, the serotonin receptors, the adrenoceptors, the

    muscarinic receptors, the prostanoid receptors, and the cannabinoid receptors28. These receptors

    are also widely studied for the development of antihistamines, antacid drugs, cardiovascular

    drugs and antipsychotics. In general, the ligand-binding pocket for members from the α-group is

    considered to be embedded in the transmembrane cavity, involving different transmembrane

    domains 29.

    The β-group mainly consists of peptide-binding receptors, notably the neuropeptide Y

    receptor. It is hypothesized that the binding pocket of this receptor group lies within the

    transmembrane cavity with the involvement of the extracellular loops and the N-terminus.

    The γ-group includes receptors for both peptides and lipid-like ligands24. The members of

    this group include opioid receptors, somatostatin receptor and angiotensin receptors. The δ-group

    mostly contains the purinergic receptors (P2RYs), the glycol-protein-binding receptors, the

    protease-activated receptors (PARs) and the olfactory receptors24.

    1.4.2 The Secretin receptor family/Class B GPCR

    Unlike the Rhodopsin family, the Secretin receptor family is a relatively small family,

    consisting of only 15 members. Secretin receptors share between 21% and 67% sequence

    homology, while most of the variation exists in the N-terminus region. The members of this

    family are the calcitonin and calcitonin-like receptors, the corticotropin-releasing hormone

    receptors, the glucagon receptor, the gastric inhibitory polypeptide receptor, the glucagon-like

    peptide receptors, the growth-hormone-releasing hormone receptors, the adenylate cyclase

    activating polypeptide receptor, parathyroid hormone receptors, the secretin receptor, and the

    vasoactive intestinal peptide receptors30. Given the identities of its members, the Secretin

  • 8

    receptor family is mostly involved in the endocrine and metabolic system of the mammalian

    body.

    1.4.3 The Adhesion receptor family/Class B GPCR

    The Adhesion family is the second largest receptor family within the GPCR superfamily,

    having 33 members24. Despite sharing some sequence similarities with the Secretin receptor

    family, there are striking differences within the N-terminal domain architecture between both

    families30. The Adhesion family possesses the GPCR proteolytic (GPS) domain in its

    extracellular regions, whereas the Secretin family does not. Adhesion receptors bind extracellular

    matrix molecules, while Secretin receptors bind peptide hormones. It is believed that this family

    likely plays a role in immunological functions and developmental biology of the central nervous

    system26.

    1.4.4 The Glutamate Receptor Family/ Class C GPCR

    The Glutamate receptor family has 22 members: eight metabotropic glutamate receptors,

    two GABAB receptors, the calcium-sensing receptor, the sweet and umami taste receptors,

    GPRC6A and seven orphan receptors31. Based on its members, it is evident that this family is

    important for neurobiological and gustatory functions26.

    In general, members from the Glutamate family interact with their respective endogenous

    ligands within their N-termini. Specifically, the crystallization of rat metabotropic glutamate

    receptor 1 (mGluR1) shed light into the mechanism by which the N-terminus interacts with a

    ligand. The crystallization study illustrated that the N-terminus of mGluR1 is folded into two

    lobes, fixed by intraprotomeric disulphide bridges32. Its ligand-binding mechanism has been

    compared to a Venus flytrap: the two lobes of its N-terminus form a cavity for the binding of

  • 9

    glutamate. Aside from their ligand-binding N-termini, metabotropic glutamate receptors can also

    interact with their allosteric ligands through their TM3, TM5, TM6 and TM733-35.

    1.4.5 The Frizzled Receptor Family/ Class F GPCR

    The Frizzled receptor family consists of 11 members: ten frizzled receptors and one

    smoothen receptor24. The frizzled receptors bind to Wnt glycoproteins36, whereas the smoothen

    receptor functions in a ligand-independent manner serving as the signaling unit in the sonic-

    hedgehog-and-smoothened-receptor complex37. The receptors from this family are implicated in

    developmental biology, cancer, etc.26.

    1.5 GPCR Dimerization

    Although GPCRs can function as monomers38, a growing body of research has suggested

    the possibility that GPCRs may also function as dimers (i.e. two GPCR come together to form a

    protein complex). Coimmunoprecipitation of differentially epitope-tagged receptors has been one

    of the commonly-used techniques to study GPCR dimerization. This technique was first used to

    study the dimerization of β2 adrenergic receptors (β2-AR). Hebert et al. co-expressed HA- and

    MYC-tagged β2-AR in cell lines, and detected HA immunoreactivity in samples

    immunoprecipitated with anti-MYC antibodies, demonstrating the existence β2-AR dimers39. To

    ensure the selectivity of the receptor-receptor interaction, the investigators repeated the study

    using MYC-tagged M2 muscarinic receptor, but did not coimmunoprecipitate it along with the

    HA-tagged β2-AR. Following this study, many research groups have used similar approaches and

    reported the homodimerization of different GPCRs including the dopamine receptor40, the

    metabotropic glutamate receptor 541, the δ-opioid receptor 42, the M3-muscarinic receptor 43, etc.

    Aside from coimmunoprecipitation, dimerization can be detected using resonance energy

    transfer approaches. This technique can detect dimerization in living cells as it bases on the non-

  • 10

    radiative transfer of energy between an energy donor and an energy acceptor44. With

    bioluminescence resonance energy transfer (BRET) technique, upon the dimerization of two

    receptors, the catalytic degradation of a luciferin molecule by a luciferase enzyme generates

    resonance energy that transfers to a green fluorescent protein and enables the fluorescent protein

    to emit at its characteristic wavelength45. By genetically linking GFP and Renilla luciferase to

    the carboxyl tail of β2-AR, a BRET study confirmed that β2-AR dimers do exist in living cells46.

    Furthermore, BRET was used to show dimerization in δ-opioid receptors47 and thyrotropin-

    releasing hormone receptors48.

    Another technique to detect receptor-receptor interactions in living cells is fluorescence

    resonance energy transfer (FRET). In this technique, a pair of fluorescent proteins such as cyan

    fluorescent protein (CFP) and yellow fluorescent protein (YFP) are used due to their unique

    emission-absorption relationship49. Upon the excitation of CFP, it generates resonance energy

    that is transferred to excite YFP if the two receptors are in close proximity. Stanasila et al. have

    used FRET to report the homo-dimerization of α1a-adrenoceptors and α1b-adrenoceptors,

    respectively50. In addition to this, the formation of heterodimer between α1a-adrenoceptor and

    α1b-adrenoceptor has also been documented using FRET50.

    1.6 Protein-Protein Interaction on signaling cascades

    1.6.1 Interaction Between Members of the Same Receptor Subfamily

    There is an increasing body of evidence demonstrating the existence of receptor-receptor

    interactions between closely related receptors such as GABAB receptor 1 and GABAB receptor 2

    51-53, the δ-opioid and µ-opioid receptors54, 55, and the SST3 and SST2a somatostatin receptors56.

  • 11

    GPCRs are known to exert their physiological functions through their interactions with

    G-proteins. Therefore, receptor-receptor interactions may present a new means for GPCRs to

    elicit their physiological actions. In the Rhodopsin receptor family, the physical interaction

    between dopamine D1 receptor (D1R) and dopamine D2 receptor (D2R) was first reported in rat

    striatal tissue using coimmunoprecipitation57. The D1R-D2R interaction was later confirmed in

    both primary rat striatal neuronal culture and in situ brain sections using confocal FRET

    studies58.

    To unveil the structures mediating the D1R-D2R interaction, Pei et al. generated GST-

    fusion proteins that encoded different fragments of each dopamine receptor and performed a

    series of affinity binding assays59. The experimental data concluded that the carboxyl tail of D1R

    interacts with the third intracellular loop of D2R to facilitate their receptor-receptor interaction59.

    A different research group further expanded that the D1R-D2R interaction was specifically

    mediated through discrete amino acids within those regions: two adjacent glutamic acid residues

    in the carboxyl tail of D1R and two adjacent arginine residues in the third intracellular loop of

    D2R60.

    In the central nervous system, five distinct dopamine receptors (i.e. D1 to D5 receptors)

    have been identified, and all of them belong to the GPCR superfamily. In general, D1 receptors

    activate adenylyl cyclase to increase cyclic AMP production through Gs/olf protein signaling,

    whereas D2 receptors exert the opposite effects by inhibiting adenylyl cyclase through their Gi/o

    protein coupling61, 62. Rather than Gs/olf or Gi/o G-proteins, the activated D1R-D2R heterodimer

    couple with a completely different G-protein signaling cascade (i.e. Gq/11), which enables the

    protein complex to activate phospholipase C and trigger different downstream pathways to

    release calcium from endoplasmic reticulum57, 58, 63.

  • 12

    The D1R-D2R protein complex is not the only example in which a physical interaction

    between two receptors can alter their signaling transduction. The physical interaction between

    dopamine D2 receptor (D2R) and D5 receptor (D5R) is another example, where the carboxyl tail

    of D5R interacts with the third intracellular of D2R 64, 65. The activation of D5R induces a robust

    increase in intracellular calcium level, and this ability is attenuated by its interaction with D2R64.

    Another example within the dopaminergic receptor family is the interaction between D2R and

    dopamine D3 receptor (D3R)66. Novi et al. co-transfected COS-7 cells with both D2R and D3R to

    study whether D3 receptor can alter D2 functions through their physical interaction, and revealed

    that in the presence of excess D3R, partial D2R agonists such as aripiprazole and S33592 lost

    their partial agonistic properties but rather produce antagonistic actions at D2R67.

    1.6.2 Interaction Between Members of Different Receptor Subfamilies

    Receptor-receptor interaction between members of different GPCR receptor subfamilies

    can also impact their respective signaling cascades. The interaction between dopamine D2

    receptor (D2R) and serotonin 5-HT2A receptor (5-HT2AR) is one example. This receptor-receptor

    interaction was detected in cells using FRET, BRET, and coimmunoprecipitation techniques68-70.

    The interaction is mediated by the carboxyl tail of 5-HT2AR and the third intracellular loop of the

    D2R68. Using FRET, Lukasiewicz et al. determined that ligand-binding can directly influence the

    formation of this heterodimer in cells: the presence of either 5-HT2AR agonists or D2R agonists

    decrease the level of interaction between the two receptors, whereas their respective antagonists

    increase it68. Regarding their G-protein signaling cascades, the two receptors are associated with

    different G-proteins and therefore distinct downstream signaling cascades: the D2R is linked to

    Gi/o protein, whereas 5-HT2AR is linked to Gq/11. Through their interaction, however, Borroto-

    Escuela et al. reported that activated D2R enhances serotonin 5-HT2AR-mediated Gq/11 signaling

    cascade (e.g. phospholipase C activation and intracellular Ca2+ mobilization)69. On the other

  • 13

    hand, the activation of 5-HT2AR attenuates the ability of D2R to inhibit adenylyl cyclase69. These

    findings together suggested an intriguing functional cross-talk between D2R and 5-HT2AR

    through their physical interaction.

    Another physical interaction leading to functional cross-talk occurs between two

    members from the Rhodopsin receptor family: the galanin receptor-1 (GalR1) and serotonin 5-

    HT1A receptor (5-HT1AR)71. GalR1, a receptor for the neuropeptide galanin, is a member of the γ-

    group in the Rhodopsin receptors family, whereas 5-HT1AR belongs to the α-group24. Both

    receptors transduce their signals through the activation of Gi/o proteins, inhibiting the activity of

    adenylyl cyclase72, 73. The receptor-receptor interaction between GalR1 and 5-HT1AR was first

    reported in HEK293 cells using FRET, whereas the co-activation of GalR1 and 5-HT1AR did not

    yield additive inhibition of adenylyl cyclase, despite both receptors being coupled to Gi/o

    proteins71. Furthermore, the presence of GalR1 reduces ligand-binding affinity for 5-HT1AR

    receptor, suggesting an antagonistic allosteric relationship between the receptors74.

    1.6.3 Interaction between a GPCR and a non-GPCR

    Aside from interacting with members from their own GPCR superfamily, GPCRs can

    also interact with membrane receptors outside the GPCR superfamily, with which they share no

    structural or functional similarities. One of the most intriguing examples of such GPCR/non-

    GPCR interactions is the interaction between dopamine D5 receptor (D5R) and the ligand-gated

    ion channel GABAA receptor (GABAAR)75. This interaction is mediated through the carboxyl

    tail of D5R and the second intracellular loop of the GABAAR γ2-subunit. It was revealed in

    HEK293 cells that the formation of the D5R-GABAAR protein complex requires the co-

    activation of both receptors. Functionally, the presence of GABA reduces the ability of D5R to

    stimulate adenylyl cyclase, but does not alter the affinity of dopamine for its receptor. The

  • 14

    activation of D5R also dampens GABAAR-mediated current in HEK293 cells and modifies the

    amplitude of GABAAR-medated miniature inhibiotry postsynaptic currents in cultured

    hippocampal neurons. Such antagonistic regulation is abolished when a peptide encoding the

    carboxyl tail of D5R is present or when the carboxyl-tail of D5R receptor is replaced with the

    carboxyl tail of dopamine D1 receptor. These data together emphasize the importance of the

    binding between the carboxyl tail of D5R and the γ2 subunit of GABAAR for the expression of

    functional cross-talk.

    Functional cross-talk through physical interaction is not exclusively limited to receptor-

    receptor interaction. Hu et al. showed that the β1-adrenoceptor (β1-AR), a GPCR, can also

    interact with cytoplasmic proteins such as the postsynaptic density scaffolding protein PSD-9576.

    The interaction is mediated by the carboxyl tail of β1-AR and the third PDZ domain of the PSD-

    95. Particularly, the last few amino acids of the β1-AR carboxyl tail are crucial for the formation

    of this interaction as mutations of these few amino acid residues abolish the binding and disrupt

    the co-localization of β1-AR and PSD-95 in HEK293 cells. Although this interaction does not

    alter the ability of β1-AR to induce the production of cylic AMP, the presence of PSD-95

    markedly attenuates the internalization of β1-AR in HEK293 cells.

  • 15

    1.7 Rationale and Hypothesis

    For many years, it is widely accepted that G-protein coupled receptors (GPCRs) function

    through their G-protein signaling cascades as their name suggests. This conventional mechanism

    of action involves the binding of an agonist, where GPCR activates its G-protein and triggers its

    downstream signaling cascades to exert its physiological effects.

    Beside G-protein signaling cascades described in Section 1.5 and 1.6, there are studies

    suggesting that members from the GPCR superfamily can physically interact with one another,

    and that it is perhaps their structural similarities that allow for their receptor-receptor

    interactions. Many researchers have suggested that these physical interactions can have impacts

    on the functionalities of the receptors involved. In other words, a GPCR can regulate/modulate

    the functions of another GPCR through their physical interaction.

    As protein-protein interactions become better understood within the GPCR superfamily,

    several aspects of protein-protein interactions involving GPCRs remain to be explored. For

    instance, it remains to be confirmed whether protein-protein interactions provide an additional

    platform for GPCRs to elicit their physiological functions in the central nervous system. Another

    question is whether GPCRs exclusively form protein complexes with membrane receptors within

    the GPCR superfamily. Although many studies discussed in Section 1.6 reported protein-protein

    interactions solely involving receptors from the GPCR superfamily, there are also reports of

    protein-protein interactions between GPCR and receptors outside the GPCR superfamily. In this

    thesis, we investigate whether GPCR protein-protein interactions exclusively exist at the post-

    synaptic membrane. Many reports discussed in this chapter focus on GPCR protein-protein

    interactions at the post-synaptic membrane, but the possibility of GPCR protein-protein

  • 16

    interactions occurring at the pre-synaptic membrane remains to be determined. To answer some

    of these questions about protein-protein interactions involving GPCRs, we have investigated

    three different protein-protein interactions in this dissertation:

    i) metabotropic glutamate receptor 1 and N-methyl-D-aspartate receptor (Chapter 2)

    ii) dopamine D1 receptor and synaptic Ras GTPase-activating protein (Chapter 3)

    iii) dopamine D2 receptor and dopamine transporter (Chapter 4)

    Regarding the potential protein-protein interaction between metabotropic glutamate

    receptor 1 and N-methyl-D-aspartate receptor, several studies have reported a functional

    connection between these two receptors, whereas metabotropic glutamate receptor 1 has been

    shown to modulate the effects of N-methyl-D-aspartate receptor on excitotoxicity. Structurally,

    both receptors have been reported to bind to inter-related scaffolding proteins at the postsynaptic

    membrane, prompting the possibility of a physical interaction between the receptors. In Chapter

    2, our hypothesis is that these two receptors physically do form a protein complex, and such

    protein complex is implicated in the excitotoxic actions of N-methyl-D-aspartate receptor.

    To date, many studies have reported that GPCRs can form protein complexes with

    membrane receptors, but few have focused on the possibility of GPCRs interacting with

    intracellular proteins. One objective explored in this thesis is to determine whether GPCRs can

    physically interact with intracellular proteins. To do so we report on the investigation of the

    potential interaction between dopamine D1 receptor and synaptic Ras GTPase-activating protein

    in Chapter 3. Although a growing body of research has suggested the important roles of both

    dopamine D1 receptor and synaptic Ras GTPase-activating protein in the development of

    GABAergic interneurons within the central nervous system, the exact molecular mechanism

  • 17

    remains elusive. Therefore, we hypothesize that dopamine D1 receptor interacts with synaptic

    Ras GTPase-activating protein, and more importantly, this interaction is crucially involved in the

    tangential migration of GABAergic interneurons in the developing mammalian brain.

    Unlike the two protein-protein interactions mentioned above, the interaction between

    dopamine D2 receptor and dopamine transporter is known to occur at the pre-synaptic membrane.

    This interaction is mediated through the N-terminus of dopamine transporter and the third

    intracellular loop of dopamine D2 receptor. Although the previous studies have revealed that this

    interaction is involved in the regulation of dopamine re-uptake in vitro, its physiological

    functions remain to be confirmed in vivo. Based on the previous in vitro data, we here

    hypothesize that the interaction between dopamine D2 receptor and dopamine transporter

    promotes the re-uptake of dopamine in vivo, and therefore the disruption of this interaction leads

    to an increase in extracellular dopamine and causes behavioural changes.

    By studying these three potential protein complexes, we have examined the prospects of

    GPCRs physically interacting with a membrane receptor (N-methyl-D-aspartate receptor) and an

    intracellular protein (synaptic Ras GTPase-activating protein) at the post-synaptic membrane as

    well as a neurotransmitter transporter (dopamine transporter) at the pre-synaptic membrane. By

    disrupting these protein complexes in vitro and in vivo, we strive to investigate whether protein-

    protein interactions can be another viable platform for GPCRs to fulfill their physiological roles.

    This investigation aims to provide direct evidence to answer the above questions regarding

    GPCR protein-protein interactions.

  • 18

    Chapter 2

    Metabotropic Glutamate Receptor 1 Modulates the Excitotoxic Functions of N-Methyl-D-Aspartate Receptor (NMDAR) through

    its Interaction with the GluN2A subunit of NMDAR

    Author contributions for this chapter:

    I performed all experiments described in this chapter with the following exceptions:

    • Dr. Dongxu Zhai performed the transient middle cerebral artery occlusion surgery • Dr. Ping Su conducted the co-immunoprecipitation experiment.

    • Mr. Jay Boychuk performed western blotting to examine the phosphorylation level of ERK1/2.

  • 19

    Metabotropic Glutamate Receptor 1 Modulates the Excitotoxic Functions of N-Methyl-D-Aspartate Receptor (NMDAR) through its Interaction with the GluN2A subunit of NMDAR

    2.1 Introduction

    2.1.1 Glutamate Neurotransmission

    Glutamate is the carboxylate anion form of glutamic acid, but more importantly, it is the

    major excitatory neurotransmitter in the mammalian central nervous system (CNS)77-79. In the

    CNS, glutamate is about 1,000 times higher in concentration than other important

    neurotransmitters such as dopamine or serotonin79.

    The biosynthesis of glutamate often takes place in neuronal terminals where it is largely

    synthesized from glutamine. Glial cells release glutamine into extracellular spaces, where

    neurons take up and transform it into glutamate via phosphate-activated glutaminase80. After

    being released into the synaptic cleft, the actions of glutamate can be terminated by reuptake into

    either neurons or glial cells through specific glutamate transporters. Once in glial cells, glutamate

    will be transformed back into glutamine by specific glutamine synthetase81.

    As a principal neurotransmitter, glutamate mediates synaptic transmission and plays a

    crucial role in synaptic plasticity, learning and memory, and cell death82-85. To exert its

    physiological functions, glutamate must bind to its receptors, which are divided into two main

    families ----- ionotropic and metabotropic receptors. Ionotropic glutamate receptors are ligand-

    gated ion channels, which include N-methyl-D-aspartate receptors (NMDAR), α-amino-3-

    hydroxy-5-methyl-4-isoxazolepropionic acid receptor and kainate receptors. These receptors

    mediate the fast-excitatory actions of glutamate by allowing the influx of cations, mostly

    Na+/Ca2+ or Na+/K+ ions, into cells. Metabotropic glutamate receptors mediate slower

  • 20

    neurotransmission through their downstream signaling cascades, and generally rather serve a

    modulatory role in neurotransmission.

    2.1.2 The Implication of Glutamate in Ischemic Stroke

    Although glutamate is essential for the normal functions of the CNS, it is also implicated

    in the pathophysiology of ischemic stroke. Stoke is one of the leading causes of mortality and

    long-term disability worldwide as it affects nearly 15 million people every year86, 87. In the

    United States alone, a stroke will take place on average every 40 seconds, and kills one person

    every four minutes88, 89. Most stroke cases (approximately 80%) are ischemic stroke, which

    arises when the systematic blood flow to the brain is severely impaired due to a thrombosis, an

    embolism or systemic hypo-perfusion86, 90. During an ischemic stroke, the brain is subsequently

    deprived of oxygen due to the lack of steady blood flow. As a result, neurons are unable to

    generate sufficient adenosine triphosphates (ATPs) through oxidative phosphorylation. In the

    lack of ATPs, Na+/K+ ATPases fail to maintain and restore the ionic gradient potential across the

    plasma membrane. Consequently, neurons will continue to depolarize and lead to a massive

    release of neurotransmitters, particularly glutamate91, 92. Since normal membrane potential is also

    required for the proper functioning of glutamate transporters, these transporters are disabled to

    overcome the massive release of glutamate, further contributing to the pathological

    progression93. The excessive glutamate release will then lead to over-stimulation of ionotropic

    glutamate receptors, which results in a constant influx of Ca2+ ions. The abnormal massive influx

    of Ca2+ ions triggers a variety of calcium-regulated processes such as the activations of

    proteases, endonucleases, nitric oxide synthase, the production of free radicals, and the

    disruption of mitochondrial membrane94. Within the ionotropic glutamate receptor family,

    NMDAR is the most potent in mediating Ca2+ ion influx and its antagonists have been widely

    considered potential neuroprotective agents95-97. Unfortunately, many NMDAR antagonists have

  • 21

    failed in clinical trials as they had unacceptable adverse effects in patients, likely due to the

    physiological significance of NMDAR95, 98, suggesting the need for an alternative approach.

    2.1.3 N-methyl-D-aspartate receptor (NMDAR)

    Like other ionotropic glutamate receptors, NMDAR possesses a tetrameric structure. As

    of now, seven NMDAR subunits have been identified: GluN1, GluN2A-GluN2D, and GluN3A-

    GluN3B99. Generally, a functional NMDAR contains two obligatory GluN1 subunits and two

    GluN2/GluN3 subunits. Although a thorough structural investigation has not been conducted on

    NMDAR, the X-ray crystallography of a closely-related GluA2 provides some valuable insights

    to the structural properties of NMDAR100, 101. It is widely accepted that all seven NMDAR

    subunits share several structural similarities: an extracellular amino terminus, a ligand-binding

    domain, four transmembrane domains (note: with the second “transmembrane” domain as an

    membrane re-entrant loop102), and an intracellular carboxyl tail103-107. Functionally, the GluN1

    subunits provide the binding site for glycine, a co-agonist, while the GluN2 subunits construct

    the binding site for glutamate/NMDA. The binding of glutamate and glycine in the ligand-

    binding domains causes the receptor to undergo conformational changes, opening the receptor

    pore to allow the influx of cations. Within the tetrameric NMDAR, the composition of the GluN2

    subunits plays an important role in determining the receptors properties such as activation and

    deactivation kinetics, ion conductance, and affinity for agonist108.

    2.1.4 Metabotropic Glutamate Receptor 1 (mGluR1)

    To date, there are eight metabotropic glutamate receptors (mGluR1-8) identified in the

    human genome109-116. These eight metabotropic glutamate receptor subtypes are divided into

    three groups based on their sequence homology, downstream signaling cascades, pharmacology,

    and phylogenetics117. Group I consists of mGluR1 and mGluR5, which are generally coupled

  • 22

    with Gq/11 proteins and are activated by 3,5-dihydroxyphenylglycine (3,5-DHPG). Group II has

    GluR2 and mGluR3, which are coupled with Gi/o proteins and are activated by 2R,4R-amin-

    opiperidindicarboxylic acid. Group III includes mGluR4, mGluR6, mGluR7 and mGluR8, which

    are also reported to couple to Gi/o in recombinant systems but are activated by 2-amino-4-

    phosphonobutyrate.

    As a member of the GPCR superfamily, mGluR1 shares the common seven-

    transmembrane-domain structure (refer to Figure 1.2) and relies on G-proteins to trigger its

    downstream signaling cascades. The second intracellular loop is the longest intracellular loop

    and has been implicated in G-protein coupling and selectivity118. Studies involving chimeric

    receptors have confirmed that the second intracellular loop is the essential component to mediate

    the coupling between mGluR1 and G-protein, whereas the first intracellular loop, the third

    intracellular loop and the carboxyl tail strengthen the coupling118, 119. Upon activation, mGluR1

    activates phospholipase C through its coupling with Gq/11. Phospholipase C will in turn cleave

    phosphatidylinositol-4,5-bisphosphate into inositol-1,4,5-triphosphate (IP3) and diacylglyercol

    (DAG). While DAG stays at the membrane to activate protein kinase C, the soluble IP3 can

    diffuse into the cell and bind to its receptors on the surface of the endoplasmic reticulum to

    release Ca2+ from the intracellular calcium store.

    2.1.5 Rationale and Hypothesis: Functional/Physical Cross-Talk between mGluR1 and NMDAR and Their Implications in Ischemic Stroke

    Located on the post-synaptic membrane, mGluR1 may influence the actions of NMDAR,

    and previous studies have suggested that mGluR1 contributes to NMDAR-mediated

    excitotoxicity. For instance, pharmacological activation of mGluR1 amplifies the neurotoxic

    effects of NMDAR stimulation in vitro120-122. Reports have also shown that mGluR1 antagonists

    prevent neuronal damage in animal models of stroke123, 124. Since both NMDAR and mGluR1 are

  • 23

    localized on the post-synaptic membrane, there is a possibility that they may form a physical

    interaction. The GluN2A subunit of NMDAR can bind scaffolding proteins such as PSD-95125,

    126, whereas mGluR1 through its carboxyl tail can form protein complexes with Homer1, another

    scaffolding protein127. Interestingly, the coupling to Homer1 enables mGluR1 to form protein

    complexes with intracellular proteins such as PSD-95 through Shank protein128. Therefore, we

    hypothesize that: 1) mGluR1 and NMDAR form a receptor-receptor interaction, and that 2) this

    receptor-receptor interaction influences the excitotoxic actions of NMDAR.

    2.2 Materials and Methods

    2.2.1 Co-immunoprecipitation

    Rat hippocampal brain tissue was lysed with 1 × RIPA lysis buffer (50 mM Tris (Bioshop

    Canada Inc.; Catalogue #: TRS001), 150 mM NaCl, 1% NP-40 (Bioshop Canada Inc.; Catalogue

    # NON505), 0.5% deoxycholate (Sigma-Aldrich Canada Co.; Catalogue # D6750), 2 mM

    EDTA, 1% Triton X-100, 0.1% SDS, 10% glycerol and 1:100 protease inhibitor cocktail (Sigma-

    Aldrich, catalogue# P8340)), and homogenized by a physical homogenizer. For co-

    immunoprecipitation, 500 – 1000 μg solubilized protein extracted from rat hippocampal brain

    tissue was incubated in the presence of primary antibodies or IgG (negative control) (1 – 2 μg)

    together with protein A/G plus agarose (Santa Cruz Biotechnology, Dallas, TX, USA; catalogue#

    sc-2003) at 4°C for 12 h. Pellets were washed with 1 × TBST (20 mM Tris, 150 mM NaCl, and

    0.1% Triton X-100 at pH 7.4), boiled for 5 min in 2x Laemmli sample buffer (Bio-Rad

    Laboratories, Mississauga, ON, Canada; catalogue# 161-0737) and subjected to SDS-PAGE. 50

    – 100 μg of protein extracted from rat hippocampal brain tissue was also boiled in 2x Laemmli

    sample buffer and used as a control in each experiment.

  • 24

    2.2.2 GST Protein Affinity Purification

    For GST protein affinity purification experiments, 500 – 1000 μg of protein was

    incubated with glutathione-sepharose beads (GE Healthcare Bio-Sciences, Mississauga, ON,

    Canada; catalogue# 17075601) bound to the indicated GST-fusion proteins (50 – 100 μg) at 4°C

    overnight. Beads were washed with PBS buffer (80mM Na2HPO4, 1.5M NaCl, 20mM

    KH2PO4, 30mM KCl, and 0.5% Triton X-100 at pH 7.4), boiled for 5 min in SDS sample buffer

    and subjected to SDS-PAGE.

    2.2.3 Western Blotting

    The SDS-PAGE was conducted in Tris-Glycine-SDS buffer (BioBasic Canada Inc.;

    catalogue# A0030) and was run at 90 – 120 V for approximately two hours. Subsequently,

    proteins were transferred onto nitrocellulose membrane at 400 mV for 2 hours before western

    blotting with primary antibodies. The primary antibodies used in this study include anti-mGluR1

    (Sigma Millipore, Etobicoke, ON, Canada; rabbit, catalogue# 07-617), ant-GluN2A (Santa Cruz

    Biotechnology, Dallas, TX, USA; goat, catalogue# sc1468), anti-GluN2A (Sigma Millipore,

    Etobicoke, ON, Canada; rabbit, catalogue# 07-632), anti-GluN1 (Sigma Millipore, Etobicoke,

    ON, Canada; mouse, catalogue# 05-432), anti-TfR (Novus Biologicals, Centennial, CO, USA;

    rabbit, catalogue# NB100-92243), anti-ERK1/2 (Cell Signaling Technology, Boston, MA, USA;

    rabbit, catalogue# 9102S), and anti-pERK1/2 (Cell Signaling Technology, Boston, MA, USA;

    rabbit, catalogue #9101S). The intensity of protein expression level was quantified by

    densitometry (software: Image Lab, Bio-Rad Laboratories, Mississauga, ON, Canada).

    2.2.4 GST Fusion Protein Constructs

    GST-fusion proteins encoding truncated mGluR1 fragments were amplified by PCR from

    full-length rat cDNA clones. GST-fusion proteins were prepared from bacterial lysates with

  • 25

    Glutathione Sepharose 4B beads as per the manufacturer (GE Healthcare Bio-Sciences,

    Mississauga, ON, Canada; catalogue# 17075601) as previously described. To construct GST-

    fusion proteins encoding truncated mGluR1, cDNA fragments were amplified by PCR with

    specific primers (Life Technologies, Waltham, MA, USA). All 5´ and 3´ oligonucleotides

    incorporated BamHI sites (GGATCC) and XhoI sites (CTCGAG), respectively, so that digestive

    enzymes BamHI and XhoI (New England Biolabs, Whitby, ON, Canada) were used to facilitate

    sub-cloning into the pGEX-4T3 vector. All constructs were sequenced to confirm appropriate

    splice fusion and the absence of spurious PCR generated nucleotide errors.

    2.2.5 GST Fusion Protein Expression and Purification

    Upon the confirmation of DNA sequences, the purified plasmids were transformed into

    BL21 competent cells (approximately 3 µL plasmids and 33 µL BL21 competent cells) and

    incubated on wet ice for 30 minutes. Subsequently, samples were heat-shocked at 42°C for 90

    seconds, followed by a two-minute incubation on wet ice. The samples were then cultured on

    ampicillin-containing lysogeny broth (LB) agar plates overnight at 37°C.

    Following the overnight incubation, bacterial colonies were collected from the

    ampicillin-containing LB agar plates. Picked colonies were incubated (at 250 rpm and 37°C) in

    25 mL ampicillin-containing LB medium overnight, and then the solution was transferred into a

    225 mL of fresh ampicillin-containing LB medium for further incubation (at 250 rpm and 37°C)

    until the culture solution reached an optical density value of 0.6 at 600 nm. To induce the

    production of GST-fusion proteins, isopropyl β-D-1-thiogalactopyranoside (IPTG) at 0.5 mM

    was added to the culture solution, and the incubation continued for three hours in an orbital

    shaker at 250 rpm and 37°C. The solution was then centrifuged at 3000 rpm for 20 minutes, and

    the resulting pellets were collected for protein extraction.

  • 26

    The pellets were re-suspended and lysed in 5 mL PBS containing 1% Triton and 1:100

    protease inhibitors. To break the cells, the samples were sonicated (Vibra Cell, Sonics &

    Materials Inc. Danbury, Connecticut, USA) on ice for 40 seconds, and was repeated for twice

    more for each sample. Following sonication, samples were incubated on a shaker at 4°C for one

    hour to allow further lysis. Subsequently, samples were centrifuged at 12, 000 rpm for 10 – 15

    minutes and the resulting supernatant was collected from each sample. Supernatants were mixed

    with glutathione agarose beads to purify the GST-fusion proteins and incubated on a shaker at

    4°C for two hours. Following the two-hour incubation, samples were centrifuged at 3000 rpm at

    4°C for one minute to remove the supernatant from each sample. Remaining beads were washed

    with 1× PBS. The GST-fusion proteins were eluted from the beads by the addition of 10 mM

    glutathione in 50 mM Tris-HCl, and samples were centrifuged at 14,000 rpm for 10 minutes to

    collect the supernatants containing purified GST-fusion proteins.

    2.2.6 Primary Mouse Hippocampal Culture

    Pregnant CD-1 mice were sacrificed, and embryos were collected on E16-18. The

    embryos were dissected to collect the hippocampal region. The hippocampal tissues were

    subsequently subjected to 0.25% Trypsin lysis for 15 minutes at 37°C. After, trypsin was

    inhibited by Dulbecco's Modified Eagle Medium containing10% fetal bovine serum. Samples

    were centrifuged at 100 × g for five minutes to remove supernatant. Fresh neurobasal medium

    containing 1:50 B27, 1:100 L-Glutamine and 1:100 Strepto-Penicllin (catalogue# 15140122,

    Gibco by Life Technologies) were added and the tissue pellet was then mechanically dissociated

    by gentle trituration through a fire-polished Pasteur pipette. The cultured neurons were then

    plated into poly-D-lysine (catalogue# P1024, Sigma-Aldrich, Oakville, ON, Canada) coated 24-

    well plate at a density of 1.0 × 105 cells per well. Cultures were incubated at 37°C in a 5% CO2

    incubator in Neurobasal (catalogue# 12349015, Gibco by Life Technologies) + B27 (catalogue#

  • 27

    17504044, Gibco by Life Technologies) + Glutamax (catalogue# 35050061, Gibco by Life

    Technologies) medium. Half of the medium were replaced by fresh neurobasal medium every

    three to four days until the end of experiments. Cytarabine (Ara-C) (Sigma-Aldrich, Oakville,

    ON, Canada) was added to the medium from four days in vitro (DIV 4) onwards to prevent glial

    cell differentiation.

    2.2.7 NMDA-Excitotoxicity in Primary Mouse Hippocampal Culture and Confocal Imaging

    On DIV 13-14, half the medium was collected from wells and replaced by fresh

    neurobasal medium. Neurons were treated with vehicle, treatment peptides (10 μM; TAT-

    mGluR1C1 YGRKKRRQRRRTFLNIFRRKKPGAGN and TAT-mGluR1C4 YGRKKRRQRRR-

    PGTPGNSLRSLYPPPPPPQHLQML) or TAT control peptide (10 μM; YGRKKRRQRRR) for

    one hour. Subsequently, neurons were exposed to 100 μM NMDA in the presence of 10 μM

    glycine for one hour. Following NMDA exposure, medium was removed and replaced with the

    original medium collected at the beginning of the experiment. 24 hours later, the neurons were

    double-labelled with 50 μg/mL propidium iodide (Sigma-Aldrich, catalogue# P4170) and 20

    μg/mL Hoechst 33342 (Life Technologies, Waltham, MA, USA; catalogue# H3570). Following

    the histological staining, the cells were washed with 1x PBS, and then fixed with 4%

    paraformaldehyde for 15 minutes. After fixation, cells were washed three times with 1x PBS,

    five minutes each. Subsequently, cells were imaged with confocal laser scanning microscope

    (Olympus FluoView FV1200, Center Valley, PA, USA) at 10× objective lens magnification for

    primary cultured neurons. The number of propidium iodide and Hoechst 33342-stained cells was

    quantified using ImageJ (National Institutes of Health, Bethesda, MD, USA) to assess the level

    of cell death.

  • 28

    2.2.8 Animals

    Male Sprague-Dawley rats weighing 200 – 225g were purchased from Charles River

    Laboratories (Montreal, Quebec, Canada), and were acclimatized to the animal facility for a

    week before being subject to any surgical and experimental procedures. All animal procedures

    were approved by the Animal Care Committee at the Center for Addiction and Mental Health

    and followed the relevant guidelines and regulations of the Canadian Council on Animal Care

    2.2.9 Transient Middle Cerebral Artery Occlusion (tMCAO)

    The intraluminal suture method was used to induce transient focal cerebral ischemia

    (tMCAO) in rats as described previously129, 130. Male Sprague Dawley rats, weighing 300 – 350

    grams, were anesthetized using 5% isoflurane and maintained at 2.5%, supplemented with

    compressed air. tMCAO was achieved by introducing a 3–0 monofilament suture into the middle

    cerebral artery via the internal carotid artery. Ischemia was maintained for 90 minutes. During

    the surgical procedure, body temperature was maintained at 36.5-37.5°C with a rectal feedback

    controlled homoeothermic blanket system and a heating lamp. Rats were deeply anesthetized

    with isoflurane and sacrificed 24 hours after the induction of tMCAO.

    2.2.10 Intracerebroventricular (ICV) Peptide Delivery

    Animals were mounted onto a stereotaxic frame under anesthesia. The scalp was shaved

    and swabbed with betadine, then a midline frontal incision was made in the scalp and the skin

    was retracted bilaterally. A burr hole (2 mm) was drilled into the skull using a hand-drill and an

    injector cannula was gently inserted into the following coordinate: 0.8 mm posterior to the

    bregma, 1.5 mm lateral to the midline, and 4.5 mm ventral to the surface of the skull. After the

    cannula reached its final dorsal/ventral coordinate, 3 µl of peptide solution (5 mM; 15 nmol per

    animal) was slowly administered. Following the injection, the injector cannula remained in

  • 29

    position for 1 minute before being slowly removed. The opening on the skull was then closed

    with bone wax, and the scalp incision was sutured with 3-0 silk suture. Animals were then

    removed from the stereotaxic frame and placed underneath a heat lamp for recovery.

    2.2.11 Neurological Assessment

    Neurological examinations were performed at two hours and at 24 hours after the onset of

    the occlusion. Neurological functions were scored on a five-point scale 131, 132: a score of 0

    indicated no neurological deficits, a score of 1 (failure to extend the left forepaw fully), a mild

    focal neurological deficit, a score of 2 (inconstant circling to the left) a moderate focal

    neurological deficit, a score of 3 (falling to the left) a severe focal deficit, and rats with a score of

    4 did not walk spontaneously and had a depressed level of consciousness.

    2.2.12 Tetrazolium Chloride (TTC) Staining and Brain Infarction Analysis

    TTC staining was performed as previously described with some modifications130.

    Following the 24-hr assessment, animals were fully anesthetized with 5% isoflurane. Upon

    achieving complete anesthesia, animals were perfused with PBS; brains were immediately

    collected, dissected and cut into 1-mm-thick coronal sections. The slices were incubated in

    0.25% 2,3,5-triphenyltetrazolium chloride (in PBS) at 37°C for 30 minutes, and subsequently

    fixed with 4% paraformaldehyde (in PBS) overnight at 4°C. The slices were then scanned and

    analyzed for infarction using ImageJ (National Institutes of Health, Bethesda, MD, USA).

    2.2.13 Acute Brain Slice Treatment

    Male Sprague Dawley rats, weighing 225 – 250 g, were purchased from Charles River

    Laboratories (Quebec, Canada). One week after arrival, animals were completely anesthetized

    with 5% isoflurane through inhalation; animals were then sacrificed, and the brains were

    collected. The brains were immediately submerged in ice-cold artificial cerebrospinal fluid

  • 30

    (aCSF: 125 mM NaCl, 2.5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 1.25 mM NaH2PO4, 26 mM

    NaHCO3 and 25 mM glucose at pH 7.4) with constant supply of 95% O2 and 5% CO2 for five

    minutes. After the five-minute incubation in aCSF, the hippocampus region was isolated and cut

    into 350-µm slices using a McIlwain Tissue Chopper. The slices were then incubated in 35°C

    aCSF with constant supply of 95% O2 and 5% CO2 for 60 minutes to allow tissue recovery

    2.2.14 NMDA-Stimulated ERK1/2 Phosphorylation

    Following the recovery and peptide treatment procedures described above, brain slices

    were subjected to 50 µM NMDA for 30 minutes, except for the control group. After NMDA

    exposure, slices were lysed with 1x RIPA. One PhosSTOP pellet (Roche Diagnostics,

    Indianapolis, IN, USA; catalogue# 4906845001) was dissolved in 10 mL 1x RIPA to inhibit

    phosphatases and preserve phosphorylation of extracellular signal–regulated kinases1/2

    (ERK1/2). The level of ERK1/2 phosphorylation was then determined through immunoblotting.

    2.2.15 Membrane Expression of NMDA Receptors

    Following the recovery procedures described above, the slices were divided into five

    treatment groups: i) control, ii) TAT peptide (20 µM TAT peptide), iii) TAT-mGluR1C1 (10 µM

    TAT and 10 µM TAT-mGluR1C1), iv) TAT-mGluR1C4 (10 µM TAT and 10 µM TAT-

    mGluR1C4), and v) TAT-mGluR1C1+C4 (10 µM TAT-mGluR1C1 and 10 µM TAT-mGluR1C4).

    Except for the control group, the slices were treated with their corresponding peptide treatments

    for 60 minutes. Following the peptide incubation, brain slices were lysed, and membrane

    proteins were extracted using a commercially available extraction kit (Product # BSP002,

    BioBasic Inc.). The solubilized proteins were subsequently subjected to western blotting to

    determine the membrane expression of GluN1 and GluN2A subunits of NMDAR.

  • 31

    2.2.16 Statistical Analysis

    All data were expressed as mean ± standard error of mean (SEM). The significance levels

    of *p < 0.05, **p < 0.01, or ***p < 0.001 were used for all an


Recommended