two novel photoactivatable phosphodiesterases to ...€¦ · two novel photoactivatable...

82
Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP by Fiona Bergin A thesis submitted in conformity with the requirements for the degree of Masters of Science Department of Molecular Genetics University of Toronto © Copyright by Fiona Bergin 2017

Upload: others

Post on 18-Jun-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP

by

Fiona Bergin

A thesis submitted in conformity with the requirements for the degree of Masters of Science Department of Molecular Genetics

University of Toronto

© Copyright by Fiona Bergin 2017

Page 2: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

ii

Two Novel Photoactivatable Phosphodiesterases to

Independently Hydrolyze cAMP and cGMP

Fiona Bergin

Master of Science

Department of Molecular Genetics University of Toronto

2017

Abstract cAMP and cGMP are universal second messengers and have been implicated in synaptic

plasticity underlying learning and memory. Pharmacological and genetic manipulations highlight

the role of postsynaptic cAMP/cGMP in synaptic plasticity; however these techniques alone lack

the precision to understand their rapid and dynamic spatiotemporal roles. To non-invasively

control the rapid molecular activity in living neurons, I developed genetically engineered

photoactivatable PDE4 (PhPDE4) and PDE5 (PhPDE5) to independently degrade cAMP/cGMP

by light. To optimize their light-dependent activities, I validated their single-photon and two-

photon photoactive properties in vitro. These enzymes showed substrate specificity and rapid

millisecond level photoactivation. For in vivo application, I demonstrated the suppression effect

of cAMP in synapse structural plasticity by two-photon photoactivation of PhPDE4. In

combination with bacterial photoactivatable enzymes (PAC, BLGC) to enhance cAMP/cGMP

signaling, these novel light sensitive enzymes will enable us to understand the rapid

spatiotemporal role of cAMP/cGMP in synaptic plasticity and brain function.

Page 3: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

iii

Acknowledgments I would like to first and foremost thank my supervisor Dr. Kenichi Okamoto. Your continued

guidance, support and laughter greatly enriched my graduate studies.

I am grateful for my committee advisors, Dr. Boulianne and Dr. Hui, for their helpful

suggestions and guidance throughout my Master’s degree.

I am thankful for all the members of Dr. Okamoto’s lab, Jelena Borovak, Thomas Luyben,

Megan Valencia and Hang Li for all of your technical guidance and friendly chats!

I sincerely thank my fiancé Garrett Cannella and my family for their constant love, support and

encouragement throughout my studies.

Page 4: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

iv

Table of Contents

Acknowledgments ................................................................................................................ iii

Table of Contents .................................................................................................................. iv

List of Tables ........................................................................................................................ vii

List of Figures ...................................................................................................................... viii

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

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

1.1 Overview ..............................................................................................................................1

1.1.1 Scientific method and the need for technological advancements ..........................................1

1.2 Optogenetic tools .................................................................................................................2

1.2.1 Introduction .............................................................................................................................2

1.2.2 Channelrhodopsin: inducing neuronal activity ........................................................................2

1.2.3 Halorhodopsin: suppressing neuronal activity ........................................................................3

1.2.4 Other optogenetic tools: manipulating protein function ........................................................4

1.3 Optogenetic Techniques offer Specific Experimental Control .................................................5

1.3.1 Temporal and Spatial Control of Optogenetic tools ................................................................5

1.3.2 Optogenetics enables manipulation of specific cell types .......................................................7

1.4 Neuroscience ........................................................................................................................8

1.4.1 Synaptic Plasticity .................................................................................................................. 88

1.4.2 Structural Synaptic Plasticity ................................................................................................. 10

1.4.3 LTP and memory ................................................................................................................... 10

1.4.4 Critical intracellular signaling of LTP ..................................................................................... 11

1.5 Optogenetic manipulation of cAMP and cGMP signaling ...................................................... 14

1.5.1 Photoactivatable adenylyl cyclase (PAC) & Blue light sensitive guanylyl cyclase (BLGC) ..... 14

1.5.2 Light Activatable Phosphodiesterase (LAPD) ........................................................................ 14

1.6 Rational and aims of this study ........................................................................................... 16

Chapter 2 Materials and Methods ........................................................................................ 17

Materials and Methods .................................................................................................. 17 2

2.1 Enzyme Design ................................................................................................................... 17

Page 5: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

v

2.1.1 PhPDE enzyme design ........................................................................................................... 17

2.1.2 Protein expression and purification ...................................................................................... 17

2.2 In Vitro photoactivation assay by ELISA ............................................................................... 18

2.2.1 One photon illumination ....................................................................................................... 18

2.2.2 Two photon illumination ....................................................................................................... 19

2.3 Expression and Validation of PhPDEs in Neurons ................................................................. 19

2.3.1 Slice Culture .......................................................................................................................... 19

2.3.2 Expression in Neurons ........................................................................................................... 20

2.3.3 Structural Long Term Potentiation (sLTP) experiments ........................................................ 20

2.4 Calculations and Statistical Analysis .................................................................................... 21

Chapter 3 Photoactivatable Phosphodiesterase 4 (PhPDE4) .................................................. 22

Photoactivatable Phosphodiesterase 4 (PhPDE4) ............................................................ 22 3

3.1 Introduction ....................................................................................................................... 22

3.2 Results ............................................................................................................................... 24

3.2.1 Design of Photoactivatable Phosphodiesterase 4 (PhPDE4) ................................................. 24

3.2.2 Optimization of the PhPDE4 light dependent photoactivation ............................................ 25

3.2.3 Characterization of PhPDE4 using one-photon light (LED) ................................................... 26

3.2.4 Two-photon characterization of PhPDE4 .............................................................................. 29

3.2.5 Establish soluble and membrane bound forms of PhPDE4................................................... 31

3.2.6 Validation of PhPDE4 activity in living neurons .................................................................... 33

3.2.7 Photoactivation of PhPDE4 for future mouse behavioral experiments ................................ 36

3.3 Summary of Chapter 4 ........................................................................................................ 37

Chapter 4 Photoactivatable Phosphodiesterase 5 (PhPDE5) .................................................. 38

Photoactivatable Phosphodiesterase 5 (PhPDE5) ............................................................ 38 4

4.1 Introduction ....................................................................................................................... 38

4.2 Results ............................................................................................................................... 40

4.2.1 PhPDE5 enzyme design ......................................................................................................... 40

4.2.2 Optimization of the PhPDE5 light dependent photoactivation ............................................ 41

4.2.3 Characterization of PhPDE5 using one-photon light (LED) ................................................... 42

4.2.4 Two-photon characterization of PhPDE5 .............................................................................. 45

4.2.5 Establish soluble and membrane bound forms of PhPDE5................................................... 47

Page 6: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

vi

4.2.6 Illumination technique of PhPDE5 for future mouse behavioral experiments ..................... 49

4.3 Summary of Chapter 4 ........................................................................................................ 49

Chapter 5 Comparison of photo activity between PhPDEs and LAPD ..................................... 50

Comparison of photo activity between PhPDEs and LAPD ............................................... 50 5

5.1 Introduction ....................................................................................................................... 50

5.2 Results ............................................................................................................................... 51

5.2.1 Comparison of the light sensitive activation between LAPD, PhPDE4 and PhPDE5 ............. 51

5.2.2 Two-photon activation of LAPD ............................................................................................ 52

5.2.3 Photoactivation properties of purified LAPD, PhPDE4 and PhPDE5 ..................................... 53

5.2.4 Establish soluble and membrane bound forms of LAPD ....................................................... 53

5.2.5 Preparation of LAPD lenti viral construct for in vivo applications ........................................ 55

5.3 Summary of Chapter 5 ........................................................................................................ 56

Chapter 6 Discussion ............................................................................................................ 57

Discussion ...................................................................................................................... 57 6

6.1 Summary of the project ...................................................................................................... 57

6.2 Photoactive properties of PhPDEs ....................................................................................... 58

6.3 In vivo validation of PhPDEs in living neurons ...................................................................... 60

6.4 Comparisons and limitations ............................................................................................... 60

6.5 Future Applications ............................................................................................................. 62

6.6 Conclusion .......................................................................................................................... 62

References ........................................................................................................................... 63

Page 7: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

vii

List of Tables Table 1 Photoactivation of purified LAPD, PhPDE4 and PhPDE5 using one-photon and two-photon

excitation light…………………………………………………………………………………….... 53

Page 8: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

viii

List of Figures

Chapter 1

Figure 1-1 One-Photon and two-photon excitation properties. …………………..…………………..….6

Figure 1-2 Two-photon laser excitation spatial resolution………………………..…………………..….7

Figure 1-3 The hippocampus is a critical structure for long term memory formation…….…………..….9

Figure 1-4 cAMP and cGMP postsynaptic signaling……………………………………………………13

Figure 1-5 Current optogenetic tools to manipulate cAMP and cGMP signaling with light……………15

Chapter 3

Figure 3-1 Protein sequence alignment of the C-terminal catalytic domains in PDE2A used for LAPD and PDE4B. ……………………………………………………………………………………..………...25

Figure 3-2 Design of photoactivatable PDE4 variants and their activity………………………..…..…....26

Figure 3-3 cAMP dependent photoactivity of PhPDE4 has a rapid on and off response in vitro.….……27

Figure 3-4 PhPDE4 is activated by various light intensities and wavelengths using one-photon light......28

Figure 3-5 PhPDE4 is more efficiently activated by longer two-photon excitation and shows rapid and light intensity dependent activation at 1,100 nm…………………………………………………...……..30

Figure 3-6 In vitro one-photon photoactivation between membrane bound and soluble forms of PhPDE4……..……………………………………………………………………………………..………31

Figure 3-7 Subcellular localization of membrane bound and soluble forms of PhPDE4 in living neurons……..……………………………………………………………………………….……..………32

Figure 3-8. PhPDE4 activation at single dendritic spines suppresses cAMP effect during structural LTP……..………………………………………………………………………………...………..………34

Figure 3-9 PhPDE4 activation at single dendritic spine causes little change during sLTP induction but significantly suppresses the cAMP dependent potentiation of sLTP.………..……………………………35

Figure 3-10 In vitro photoactivation of PhPDE4 using the implantable fiber optic LED light source for in vivo mouse experiments. ……..…………….……………………………………………………..………36

Page 9: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

ix

Chapter 4

Figure 4-1 Protein sequence alignment of the C-terminal catalytic domains in PDE2A used for LAPD and PDE5A. ……..………………………………………………………………………………..………41

Figure 4-2 Design of photoactivatable PDE5 variants and their activity.………………………..………42

Figure 4-3 cGMP dependent photoactivity of PhPDE5 has a rapid on and off response in vitro………..43

Figure 4-4 PhPDE5 is activated by various light intensities and wavelengths using one-photon light.……..………………………………………………………………………………………..…….…44

Figure 4-5 PhPDE5 is more efficiently activated by longer two-photon excitation and shows rapid and light intensity dependent activation at 1,100 nm………………………………………………………….46

Figure 4-6 In vitro one-photon photoactivation between membrane bound and soluble forms of PhPDE5. ……..…………………………………………………………………………………..…………….……47

Figure 4-7 Subcellular localization of membrane bound and soluble forms of PhPDE5 in living neurons. ……..…………………………………………………………………………………..………………….48

Figure 4-8 In vitro photoactivation of PhPDE5 using the implantable fiber optic LED light source for in vivo mouse experiments. ..………………………………………………………………………………..49

Chapter 5

Figure 5-1 In vitro photoactivation of PhPDE4, PhPDE5 and LAPD on cAMP and cGMP levels. ……51

Figure 5-2 Two-photon excitation efficiently induces the degradation of both cAMP and GMP ..……..52

Figure 5-3 Subcellular localization of membrane bound and soluble forms of LAPD in living neurons 54

Figure 5-4 Validate expression and light dependent activity of pLenti-LAPD in HEK293 cells….…..…55

Chapter 6

Figure 6-1 Schematic diagram of the photocycle of PhPDEs……………………………………………58

Page 10: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

x

List of Abbreviations AC Adenylyl Cyclase

BLGC Blue Light Sensitive Guanylyl Cyclase

bPAC Bacterial Photoactivatable Adenylyl Cyclase

CA1 Cornu Ammon1 (type of Hippocampal Pyramidal Neuron)

CaMKII Calcium calmodulin-dependent protein kinase II

cAMP Cyclic adenosine monophosphate

cGMP Cyclic guanosine monophosphate

ChR2 Channelrhodopsin 2

CRE CREB responsive element

Cre-LoxP Cre recombinase – Locus of X-over P1

CREB cAMP response element binding protein

DISC1 Disrupted in schizophrenia

E-LTP Early Long Term Potentiation

ELISA Enzyme Linked Immunosorbent Assay

GAP43 Growth Associated Protein – 43

GC Guanylyl Cyclase

GFP Green Fluorescent Protein

HEK293 Human Embryonic Kidney 293 cells

L-LTP Late Long Term Potentiation

LAPD Light activatable phosphodiesterase

LED Light emitting diode

LTD Long Term Depression

LTP Long Term Potentiation

MLS Membrane localization sequence

Page 11: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

xi

NA Numerical aperture

NMDAR N-methyl-D-aspartate- type glutamate receptor

NO Nitric Oxide

NOS Nitric Oxide Synthase

NpHR Halorhodopsin

PAS-GAF-PHY Per-arnt-sim - cGMPPDE/adenylyl cyclase/Fh1A - Phytochrome domain

PDE Phosphodiesterase

PhPDE4 Photoactivatable Phosphodiesterase 4

PhPDE5 Photoactivatable Phosphodiesterase 5

PKA Protein Kinase A

PKG Protein Kinase G

RFP Red Fluorescent Protein

sLTP Structural Long Term Potentiation

Page 12: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

1

Chapter 1 Introduction

Introduction

1.1 Overview

1.1.1 Scientific method and the need for technological advancements

To understand molecular mechanisms of learning and memory, pharmacological and genetic

manipulations of various molecules have been used in the brain. However, those traditional

approaches are irreversible and limited in their spatial and temporal manipulation (Dugue,

Akemann, & Knopfel, 2012). Pharmacological approach using specifically designed chemical

inhibitors or stimulators enables perturbations of signalling pathways at the molecular level. This

approach highlights the link between different molecules and their cellular function. However,

pharmacological manipulation is limited by its spatial specificity as administration in vivo causes

widespread cellular changes in the brain. Furthermore, depending on the chemical inhibitor or

stimulator as well as the mode of injection, pharmacological approaches are typically required to

manipulate molecular activity at the minutes to hours’ time scale (Toettcher, Voigt, Weiner, &

Lim, 2011). Another technique widely used is genetic manipulations. Genetic targeting and

transgenic approaches enable precise manipulation of single genes. Knocking out or

overexpressing single genes in vivo enables observation of associated behavioural changes.

While this technique has great molecular specificity, it lacks the ability to manipulate synapse

level function. Also, to control the temporal alteration of genetic manipulation is available using

inducible systems however this takes up to a few hours (Weber et al., 2012).

For manipulating with precise temporal control as well as spatial specificity including

subcellular and cellular targeting I established an optogenetic approach to suppress cAMP and

cGMP for studying their functions in the brain. This optogenetic manipulation is reversible with

rapid onset and offset with minimal invasion. In combination with pharmacological and genetic

techniques, this approach will be a critical advanced technique for neuroscience research.

Page 13: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

2

1.2 Optogenetic tools

1.2.1 Introduction

Optogenetics is defined as “the combination of genetic and optical methods to achieve

gain or loss of function of well-defined events in specific cells of living tissue” (Deisseroth,

2011). Various types of photoreceptors are found in nature and these light sensitive proteins

contain different chromophores, which can absorb specific wavelengths of light from ultra

violet-B to near-infrared light (Rockwell et al., 2014). Typically, the absorption of light by the

chromophore in the light sensitive domain induces a conformational change resulting in altered

function of the receptor (Ziegler & Moglich, 2015). The light sensitive activity causes light-

sensitive locomotion of organisms to move towards or away from light or functions to initiate a

biochemical cascade for energy production (Jenkins, 2017; Ueki et al., 2016). To apply this

property to neuroscience research these light sensitive molecules were characterized and

optimized for controlling neuron activity by light.

1.2.2 Channelrhodopsin: inducing neuronal activity

Channelrhodopsin (ChR2), a single component light activated cation channel was discovered

from algae Chlamydomonas reinhardtii (Nagel et al., 2002). This channel absorbs blue light,

causing a conformational change, opening the channel and enabling the passive diffusion of

cation ions down its concentration gradient. This channel enables Chlamydomonas reinhardtii to

find optimal conditions for photosynthesis as it stimulates swimming towards light in dim

conditions or moving away from excess amount of light (Erickson, Wakao, & Niyogi, 2015;

Nagel et al., 2002; Nagel et al., 2003). When it is expressed in living neurons, illumination with

blue light causes the channels to open within a millisecond, inducing rapid depolarization of the

membrane potential, which induces action potentials and thus neuronal excitability (Boyden,

Zhang, Bamberg, Nagel, & Deisseroth, 2005). This discovery accelerated the search for other

photoreceptors for the optogenetic application to control neuronal activity.

ChR2 was used to examine the properties of memory storage. Current knowledge in this

area suggests that during a distinct event, strong simultaneous activation of a discrete subset of

neurons creates a specific memory trace. Reactivation of this specific trace is believed to

stimulate memory recall and should be distinct and independent of other memories. Liu and

Page 14: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

3

colleagues specifically expressed ChR2 only in the neurons activated during a fear memory (Liu

et al., 2012). Illumination of ChR2 expressed only in this specific neural circuit, in a neutral

context elicited the same fear memory recall. Verifying that specific memories are stored in

discrete neural circuits. This experiment highlights the application of optogenetic tools for

neuroscience research.

1.2.3 Halorhodopsin: suppressing neuronal activity

A perfect tool to complement such experimental manipulation would be a light sensitive

channel that can silence neural activity with the same precision. The discovery of halorhodopsin,

a chloride pump from Natronomonas pharasonis (NpHR) provided this complementary tool for

ChR2, to turn neural activity off (Kolbe, Besir, Essen, & Oesterhelt, 2000). When expressed in

living neurons, NpHR protein was found to hyperpolarize the membrane potentials and silence

neural activity upon illumination within millisecond precision. Interestingly, NpHR is optimally

activated by yellow light instead of blue light used for ChR2. It was demonstrated that, ChR2

and NpHR can be co-expressed and independently activated using different wavelengths of light

such that blue light induces activity of neurons and yellow light suppresses activity (F. Zhang et

al., 2007).

These optogenetic tools work in vivo as demonstrated in Caenorhabditis elegans. The

illumination of blue and yellow light induced the activation and silencing of motor neuron

activity respectively in C. elegans (F. Zhang et al., 2007). Furthermore, the activation of ChR2 in

muscle cells of C. elegans caused an instant increase in swimming motion whereas activation of

NpHR by yellow light instantly blocked this movement. Together these optogenetic tools enable

scientists to study the effect of activating or silencing neuronal activity, within a biologically

relevant time scale to study the functions of specific neural circuits.

These optogenetic tools were also applied to address brain diseases such as the neural connection

to anxiety disorders, which encompass numerous psychiatric diseases such as posttraumatic

stress disorder, generalized anxiety disorder and panic disorder (Tye et al., 2011) (Lieb, 2005).

In a rodent anxiety model, specific stimulation of ChR2 in a discrete population of excitatory

neurons in the amygdala showed significantly reduced anxiety like behaviour. Furthermore,

Page 15: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

4

selective illumination and therefore inhibition of this same projection using NpHR to

hyperpolarize this pathway resulted in anxiety. This experiment was repeated on a different

neural projection in the brain composed of the same cell types but showed no effect on anxiety

levels when ChR2 or NpHR were activated (Tye et al., 2011). Therefore, using this optogenetic

approach a specific cellular pathway rather than cell type was isolated to be important for

anxiety. Thus, optogenetic approach offers superior specific experimental control and will have a

big impact on basic neuroscience research as well as understanding diseased states.

1.2.4 Other optogenetic tools: manipulating protein function

The field of optogenetics accelerated as optogenetic tools moved from manipulating whole cell

activities to perturbing intracellular molecular functions (Guglielmi, Falk, & De Renzis, 2016).

Critical cellular activities are regulated by kinase and phosphatase activity. Currently there is a

light sensitive Raf kinase, which induces reversible activity of the Raf/MEK/ERK pathway upon

illumination (Krishnamurthy et al., 2016). Conversely, there is a light sensitive

phosphatidylinositol 5-phosphatase, which is recruited to the cell membrane upon illumination

and causes rapid dephosphorylation of its downstream targets (Idevall-Hagren, Dickson, Hille,

Toomre, & De Camilli, 2012). Some other optogenetic enzymes recently developed include

Photoactivatable Rac1, which regulates actin cytoskeleton dynamics (Wu et al., 2009) or a light

inducible protein oligomerization (CRY2olig), which can be used to examine protein interactions

in living cells (Taslimi et al., 2014). All of these engineered optogenetic enzymes now enable the

perturbation of various intracellular activities with subcellular resolution and fast, seconds order

precision (Guglielmi et al., 2016).

Page 16: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

5

1.3 Optogenetic Techniques offer Specific Experimental Control These light sensitive tools are superior manipulators of biological activity by their activation

speed, spatial control and the ability to restrict their expression to specific neuronal populations

(Deisseroth, 2011).

1.3.1 Temporal and Spatial Control of Optogenetic tools

Optogenetic tools offer millisecond control of excitation and inhibition of neural activity in

specific populations. Both ChR2 and NpHR have confirmed millisecond activation and silencing

of living neurons. This light dependent response advances the study of rapid temporal functions

of specific neuronal populations in the brain functions and related diseases (F. Zhang, Wang,

Boyden, & Deisseroth, 2006).

Another benefit of optogenetics is spatial control of neuronal activation by light. For example,

one photon illumination can be used to activate specific brain region or neural circuit expressing

light sensitive proteins to understand their roles in the brain functions and related diseases.

Moreover two-photon laser excitation with a fine focal point (~1μm3) using a high numerical

aperture (NA) objective lens, can be employed to localize activation to subcellular structures

such as single synapses to understand the impact on the function of microcircuits with synapse

level precision (Boyden et al., 2005).

Maria Goppert-Mayer first theorized the concept of two-photon absorption in 1931. One-

photon excitation of a fluorophore occurs when fluorophores are excited from their electronic

ground state to an excited state. Maria believed that this same excitation of a fluorophore could

be achieved by the simultaneous absorption of two less energetic photons from the infrared

spectra range (Figure 1-1). After this effect was validated in 1963, two-photon microscopy was

established in the 1990s (Denk, Strickler, & Webb, 1990; Kaiser & Garrett, 1961). To increase

the probability of simultaneous absorption of two photons, a femtosecond pulse laser is used for

two-photon microscopy. This two-photon excitation uses longer wavelengths of infrared light

therefore enabling deeper tissue penetration and spatially restricts activation to only the focal

point where two photons of light meet (~1μm3) (Figure 1-2). These properties make two-photon

laser microscopy a valuable tool for restricting optogenetic stimulation to a subcellular level.

Page 17: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

6

Figure 1-1 One-Photon and two-photon excitation properties. (A) One-photon excitation

using the visible light spectrum produces a larger area of illumination compared to (C) Two-

photon excitation focal volume (1µm3) using infrared light (radius = 0.61 x wavelength / NA x

1.414) (B) Excitation occurs when one photon absorption causes excitation between the ground

and excited state. (D) Two photon excitation occurs through the simultaneous absorption of two

lower energy photons (infrared range) if their combined energy is greater than the energy

difference between ground and excited state.

Page 18: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

7

Figure 1-2 Two-photon laser excitation spatial resolution.

(A) Due to the use of two-photons of light from longer infrared wavelengths of light, two-photon

imaging of mouse brain produces less scattering than confocal imaging resulting in deeper tissue

penetration (~ a few mm). (B) Two-photon microscopy has optimal spatial resolution for

resolving or manipulating single synaptic structures such as the post synaptic dendritic spines

highlighted above by white line.

1.3.2 Optogenetics enables manipulation of specific cell types

In combination with specific promoters, genetically engineered optogenetic tools can be

expressed in specified neuronal populations in the brain. For example, lentivirus packaged with a

CaMKIIα promoter, limits the expression in excitatory neurons and maintains the expression of

the optogenetic tool for more than 6 months in the mouse brain (Wang, Zhang, Szábo, & Sun,

2013).

Cre-LoxP expression system also enables us to limit the expression of optogenetic tools

in specific cells. This system utilizes a transgenic mouse line containing Cre recombinase under

the expression of a cell type specific promoter and either viral vector or mouse line containing

the inverted light sensitive protein flanked by loxP sites. Injection of the virus into these

transgenic mice restricts expression of optogenetic tools to cells expressing Cre recombinase.

This approach has been successful using lentivirus or adeno- associated virus (AAV) to target

optogenetic tools to specific neural cell types (Zeng & Madisen, 2012).

Another targeting method is the neuronal activity-dependent expression of optogenetic

enzymes. The expression of immediate early genes such as c-fos can be used to identify the

Page 19: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

8

neurons, which were recently activated during a specific learning and behavioral task. These

immediate early genes are transcribed within minutes and associated with neural activity

(Guzowski, 2002). Using the activity-dependent inducible system, optogenetic protein such as

ChR2 can be expressed only in the neurons activated during a particular task. This expression

system requires expression of ChR2 to be driven by the tetracycline-responsive element (TRE)

and a transgenic mouse line where the tetracycline transactivator (tTA) is driven by c-fos

promoter (Ramirez, Tonegawa, & Liu, 2013). Therefore, neural activity induces expression of

immediate early genes such as c-fos driving the expression of tTA to activate ChR2 expression in

only those neurons. Now only the cells activated during that particular training contain the

expressed ChR2 permitting the activation of only that neural circuit using light (Ramirez et al.,

2013).

1.4 Neuroscience

1.4.1 Synaptic Plasticity

Ramon y Cajal first demonstrated that brain tissue is composed of a network of signaling units

called neurons, and these neurons are arranged in functional groups that connect to one another

in a precise circuit. He postulated that memory storage relies on changes in the strength of

connections between activated neurons (Ramón y Cajal, 1995). The activity dependent changes

occur at the synapse, the junction where the presynaptic bouton meets the postsynaptic structure,

called the dendritic spine of another neuron. In the 1940s Donald Hebb supported this hypothesis

coining the phrase “neurons that wire together fire together, neurons that wire apart, fire apart”

(Morris, 1999). To confirm this hypothesis, many researchers looked into the mechanism

underlying how the synapse is strengthened leading to the model of long-term potentiation

(LTP). In 1972 Timothy Bliss and Terje Lomo applied brief high frequency trains of action

potentials in rabbit hippocampal slices resulting in stronger synaptic transmission and the

observation of LTP (Bliss & Lomo, 1973). Long term depression (LTD) was also found in

synapses that undergo prolonged low-frequency stimulation therefore suppressing synaptic

activity (Lynch, Dunwiddie, & Gribkoff, 1977).

The hippocampus is a critical region in the brain enabling long-term memory formation of

declarative memories. There are two different forms of synaptic plasticity in the hippocampus,

post-synaptic dependent and pre-synaptic dependent which occur in different hippocampal neural

Page 20: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

9

circuits. The induction of post-synaptic dependent LTP requires activation of N-methyl-D-

aspartate (NMDA)- type glutamate receptor (Collingridge, Kehl, & McLennan, 1983). In order

for NMDA receptors to be activated, they need the pairing of glutamate release from the pre-

synaptic terminal and depolarization of the postsynaptic membrane, enabling an influx of

calcium into the postsynaptic spine. This type of LTP is observed in two of the three-

hippocampal pathways (Shaffer collaterals, performant path) initially described by Ramon Y

Cajal using the Golgi staining method (Figure 1-3). The third pathway is non-NMDA receptor

dependent where the mossy fibers synapse onto CA3 neurons and rely on presynaptic dependent

mechanisms for the induction of LTP. Importantly LTP was blocked when calcium signalling

was inhibited by EGTA and conversely, LTP was induced when calcium was added to the

presynaptic cell. Therefore indicating that presynaptic increase in intracellular calcium ions is

critical for LTP induction.

Figure 1-3 The hippocampus is a critical structure for long term memory formation.

(A) The location of the hippocampus in the brain. (B) Drawing of the neural circuitry of the

rodent hippocampus adapted from Ramon y Cajal (1911).

Page 21: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

10

1.4.2 Structural Synaptic Plasticity

Along with these functional changes structural alterations also occur during synaptic plasticity

and requires reorganization of the major postsynaptic cytoskeletal protein actin. Actin is

regulated by the binding and release of an abundant postsynaptic protein CaMKII (Calcium

calmodulin-dependent protein kinase II) suggesting its importance in regulating structural and

functional plasticity. Our lab found that the postsynaptic CaMKIIβ/actin regulates both

functional and structural synaptic plasticity (Kim et al., 2015; Okamoto, Nagai, Miyawaki, &

Hayashi, 2004).

1.4.3 LTP and memory

Pharmacological inhibition of NMDA receptors blocked LTP and also impaired spatial memory

in the mouse hippocampus using the morris water maze (Morris, Anderson, Lynch, & Baudry,

1986). These early findings highlight the link between LTP and learning and memory as well as

introduced critical intracellular signalling underlying LTP.

Interestingly, LTP shares a similar time scale as memory formation further highlighting the link

between LTP and memory. Early phase of LTP (E-LTP) is typically induced by weak

stimulation, a single train of action potentials, causing an increase in intracellular calcium

leading to short-term structural and functional potentiation. This plasticity enhancement lasts for

up to 3 hours and has been linked to short-term memory formation (Kandel, 2012). Late phase of

LTP (L-LTP) is induced by multiple trains of synaptic stimulation, and causes a more persistent

structural and function plasticity enhancement lasting over 3 hours to months. This plasticity

enhancement is due to stronger stimulation inducing a greater calcium influx leading to trigger

new protein synthesis pathway critical for long-term memory formation and consolidation (Abel

et al., 1997; Frey, Huang, & Kandel, 1993). Application of protein synthesis inhibitors disrupt

long-term memory formation but did not affect short-term memory formation, suggesting a

critical link between L-LTP and long term memory (Frey, Krug, Reymann, & Matthies, 1988).

Page 22: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

11

1.4.4 Critical intracellular signaling of LTP

cAMP

Earl Sutherland was awarded the Nobel Prize in Physiology and Medicine in 1971 for

discovering cyclic adenosine monophosphate (cAMP). He determined that cAMP was a critical

second messenger that amplifies signal transduction across the cellular membrane (Sutherland,

1972). Neurotransmitters such as serotonin increase cAMP concentration in the brain linking

cAMP signalling and neural activity (Brunelli, Castellucci, & Kandel, 1976). Cyclic AMP

signalling is now known to mediate many activities in the brain including developmental

properties, synaptic plasticity and learning and memory. Using the fruit fly, Drosophila

melanogaster, mutations to the dunce gene, a cAMP-phosphodiesterase (PDE) which degrades

cAMP, caused an increase in cAMP signalling and strong learning deficits (Dudai, Jan, Byers,

Quinn, & Benzer, 1976). However, flies with a mutant adenylyl cyclase which synthesizes

cAMP, rutabaga, resulted in decreased cAMP signalling and defective learning (Levin et al.,

1992). These results suggest a critical link between regulated cAMP signalling and learning and

memory functions. At the cellular level, Eric Kandel, found that short-term memory depicted in

one trial of the gill-withdrawal reflex in the marine slug Aplysia, produced cAMP, which

transiently enhanced neurotransmitter release (Kandel, 2001). Furthermore, multiple training

trials caused long-term memory of the gill-withdrawal reflex lasting days to weeks which

required a more persistent increase in cAMP signalling causing cAMP-dependent protein

synthesis of plasticity related genes critical for long term memory formation. This simple reflex

system demonstrated in the very large (up to 1mm) neuron of Aplysia demonstrated the

important link between cAMP signalling, protein synthesis and synaptic plasticity. The canonical

cAMP pathway involves the hormone signalling, which activates adenylyl cyclase (AC) to

produce cAMP. cAMP then activates protein kinase A (PKA) which translocates into the nucleus

and phosphorylates CREB which binds the CREB responsive element (CRE) leading to protein

synthesis (Kandel, 2012).

Page 23: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

12

cGMP

3’5’ cyclic guanosine monophosphate (cGMP) is also a crucial second messenger to regulate

synaptic plasticity. cGMP is synthesized by nitric oxide (NO) stimulation of guanylyl cyclase

(GC), which converts GTP to cGMP. The canonical cGMP-signaling pathway, widely studied in

the CA1 neurons of the Schaffer Collateral pathway of the hippocampus during LTP induction,

includes NMDA receptor mediated Ca2+ influx which stimulates Ca2+/calmodulin-activated

neuronal NO synthase (nNOS) causing NO production in the postsynaptic structure. NO acts as a

retrograde messenger and translocates into the presynaptic structure where it activates GC to

produce cGMP and initiates the activation of cGMP dependent protein kinases (PKG). These

kinases initiate the increase of neurotransmitter release aiding in the presynaptic-dependent

potentiation of the synapse. NO also stimulates postsynaptic GC to activate postsynaptic PKG

for enhancing protein synthesis pathways. Thus, both cAMP and cGMP intracellular signalling

are known to play critical roles in synaptic plasticity.

PDEs

Various neurodegenerative diseases such as Alzheimer’s cause dramatic cognitive deficits for

which treatment is limited. Drug development for these diseases has moved from its original

focus of directly targeting neurotransmitters such as acetylcholine, glutamate and dopamine, to

the downstream signalling pathways in long-term memory formation. One of the major drug

targets for neurodegenerative diseases are phosphodiesterases (PDEs). PDE inhibitors, such as

rolipram, sildenafil and tadalafil have been shown to improve cognitive functions in various

mouse models of Alzheimer’s disease (Garcia-Osta, Cuadrado-Tejedor, Garcia-Barroso,

Oyarzabal, & Franco, 2012). However, the molecular mechanisms are still elusive.

The signalling dynamics between cAMP and cGMP during synaptic plasticity is also still

elusive. One member of the PDE family, PDE2, degrades both cAMP and cGMP. However the

rate at which each cyclic nucleotide is degraded depends on the cellular concentration of each.

Specifically, if there is low concentration of cGMP, then it will bind the N-terminal regulatory

domain and heighten the hydrolysis of cAMP. But if there is a low concentration of cAMP, it can

bind the N-terminal regulatory domain and cause an increase in cGMP hydrolysis. This activity

enables one cyclic nucleotide to be the dominant messenger molecule within that subcellular

micro domain. However, when both cyclic nucleotides are present in high concentrations, they

Page 24: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

13

compete for the binding site (Zhao, Greenstein, & Winslow, 2016). This causes important spatial

regulation of cyclic nucleotide signalling as well as temporal regulation by maintaining or

supressing cAMP or cGMP signalling depending on the cellular environment. This interesting

dynamic suggest an important crosstalk mechanism between these cyclic nucleotides however

the functions for such interaction at the cellular and subcellular level is unknown. Due to the

high expression of PDE2 in limbic structures such as the cortex, amygdala and hippocampus,

which are all critical sites for memory formation, maintaining the dynamic signalling interplay

between cAMP and cGMP signalling could be important for synaptic plasticity underlying

learning and memory. Inhibition of PDE2, maintaining both cAMP and cGMP signalling

enhances LTP and showed improvement on various memory tasks (Boess et al., 2004). Two

hours of acute tryptophan depletion, a dietary precursor for serotonin, causes short-term memory

impairment observed using the object recognition test. However using BAY 60-7550, a PDE2

inhibitor, 30 minutes of application before the test improved the short-term memory of these

mice (van Donkelaar et al., 2008). While research using PDE inhibitors has greatly expanded our

knowledge surrounding cAMP and cGMP signalling their spatiotemporal role and crosstalk

interactions during synaptic plasticity are less understood.

Figure 1-4 cAMP and cGMP postsynaptic signalling. Summary of the post-synaptic cAMP

and GMP signalling and the central role of various PDEs.

Page 25: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

14

1.5 Optogenetic manipulation of cAMP and cGMP signaling

1.5.1 Photoactivatable adenylyl cyclase (PAC) & Blue light sensitive guanylyl cyclase (BLGC)

Due to the widespread role of cAMP signaling, it is not surprising that a photoactivatable

adenylyl cyclase was found in a bacterium Beggiatoa (Stierl et al., 2011). This single component

blue light sensitive adenylyl cyclase (bPAC) has little activity levels in the dark but rapidly

produces cAMP upon illumination. This optogenetic tool has confirmed light dependent activity

and expresses in hippocampal pyramidal neurons, generating an efficient light sensitive approach

to manipulate cAMP spatiotemporal dynamics during synaptic plasticity. Using this bPAC,

mutagenesis of various residues involved in substrate binding sites, enabled the binding of GTP

instead of ATP and therefore produced cGMP upon illumination with blue light called blue light

sensitive guanylyl cyclase (BLGC). These tools now enable the rapid, reversible and non-

invasive stimulation of cAMP and cGMP signaling independently. To complete this optogenetic

toolkit, I prepared new tools, which independently degrade cAMP or cGMP.

1.5.2 Light Activatable Phosphodiesterase (LAPD)

A new optogenetic enzyme was recently developed, Light Activated Phosphodiesterase (LAPD),

which degrades both cAMP and cGMP upon illumination (Gasser et al., 2014). This enzyme is

composed of the light sensitive phytochrome domain from Deinococcus radiodurans fused to the

catalytic domain of PDE2, which degrades both cAMP and cGMP. The N-terminal regulatory

domain of PDEs differs across variants and contains various sites for the binding of modulators

to regulate their activity or localization properties. The C-terminal domain contains the catalytic

domain where binding and hydrolysis of cyclic nucleotides occur. Major similarities were

observed between the bacterial phytochrome domain of Deinococcus radiodurans and the N-

terminal regulatory GAF domains of PDE2. The structural similarities between these regulatory

domains enabled the replacement of the PDE2 N-terminal regulatory domain with the bacterial

phytochrome domain. Successfully enabling the degradation of both cAMP and cGMP to be

controlled by illumination with blue (455 nm), red (690 nm) and even broadband white light

(Gasser et al., 2014). The development of LAPD which degrades both cyclic nucleotides

provides a promising framework for the development of two novel optogenetic enzymes to

independently degrade cAMP or cGMP enabling the detailed understanding of activating or

blocking these second messengers during synaptic plasticity.

Page 26: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

15

Figure 1-5 Current optogenetic tools to manipulate cAMP and cGMP signaling with light.

(A) The current optogenetic toolkit is composed of photoactivatable adenylyl cyclase (PAC) to

produce cAMP, a blue light sensitive guanylyl cyclase that produces cGMP and a light

activatable phosphodiesterase LAPD (PDE2), which degrades both cAMP and cGMP. (B) Venn

diagram dividing the different families of PDEs by their substrate specificity. The red boxes

highlight a phosphodiesterase from each group, which will be used to generate the final

optogenetic tools to specifically degrade cAMP (PDE4) or cGMP (PDE5) independently using

the same strategy employed for LAPD.

Page 27: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

16

1.6 Rational and aims of this study In our lab we have established two-photon optogenetic microscopy techniques to light

dependently activate specific enzymes such as bPAC and BLGC at a single synapse resolution in

living hippocampal tissues to produce cAMP and cGMP. To compliment these tools I will create

two new optogenetic enzymes to light dependently degrade cAMP or cGMP. There are 11

families of PDEs encoded by 21 genes, each with unique tissue expressions (Bender & Beavo,

2006). Mammalian PDEs are divided into three types: PDEs that hydrolyze cAMP and cGMP

(PDE1, 2, 3, 10, 11), cAMP specific (PDE4, 7, 8) and cGMP specific (PDE5, 6, 9). By replacing

the PDE2 catalytic domain from LAPD with a cAMP-specific PDE (PDE4) catalytic domain or

cGMP-specific PDE (PDE5) catalytic domain, I prepared novel optogenetic enzymes to suppress

endogenous cAMP and cGMP signaling independently at the single synapse level. These

complementary tools will enable us to study their roles in synaptic plasticity by producing cAMP

or cGMP, or degrading cAMP or cGMP with spatial and temporal control of light.

Goal: To create and validate novel optogenetic enzymes to degrade cAMP and cGMP

independently enabling the spatiotemporal control of intracellular signaling processes mediated

by these second messengers.

Aim1: Develop and validate optogenetic tools to independently degrade cAMP or cGMP upon

illumination

Aim2: Validate and characterize two-photon photoactivation of PhPDE4 and PhPDE5 in vitro

Aim3: Demonstrate the functionality of PhPDEs in living neurons

Page 28: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

17

Chapter 2 Materials and Methods

Materials and Methods 2

2.1 Enzyme Design

2.1.1 PhPDE enzyme design

The DNA encoding LAPD (Gasser et al., 2014) was synthesized with codon usage optimized for

Homo sapiens (GenScript, New Jersey, USA). The gene encoding the catalytic domain of Homo

sapiens cAMP-specific phosphodiesterase 4 (HsPDE4B; UniProt PDE4B_HUMAN: amino acids

630-1971) was synthesized (GenScript, NJ, USA) and cGMP-specific phosphodiesterase 5

(HsPDE5A; UniProt PDE5A_HUMAN: amino acids 1531-2638) was obtained from a PDE5A

cDNA clone (ORIGENE, MD, USA). Photoactivatable phosphodiesterase 4 (PhPDE4) and 5

(PhPDE5) constructs were generated by fusing the PDE4B (amino acids 630-1971) or PDE5A

(amino acids 1531-2638) with N-terminus of LAPD phytochrome (DrBPhy; UniProt

BPHY_DEIRA: amino acids 1-506). To optimize the enzymatic photoactivation, we generated a

series of variants by fusing the different length of catalytic domain of PDE4B (amino acids from

627-, 628-, 629-, 630-, 631-, 632- or 633- to 1971) or PDE5A (amino acids from 1527-, 1528-,

1529-, 1530-, 1531-, 1532- or 1533- to 2638) by site-directed mutagenesis. For protein

purification, LAPD, LAPD4 and LAPD5 were subcloned into the pCAGGS plasmid vector

(Niwa, Yamamura, & Miyazaki, 1991) with a N-terminal 6 histidine tag. For in vitro cell lysate

assay and expression in neurons, PhPDE4 and PhPDE5 were subcloned into the pCAGGS

plasmid vector with a C-terminal EGFP tag (Clontech, CA, USA). For the membrane localized

forms, a GAP43 membrane localization signal (MEM: TaKaRa Clontech, CA, USA) was

subcloned into the N-terminal PhPDE4-EGFP and PhPDE5-EGFP.

2.1.2 Protein expression and purification

HEK293 cells were transfected with plasmids using Lipofectamine 2000. The HEK293 cells

were harvested after 48 hours of transfection, and homogenized in a buffer (40 mM HEPES/Na,

pH 8.0, 0.1 mM EGTA, 5 mM magnesium acetate, 1mM DTT, and 0.01% Tween-20) by

sonication, and centrifuged at 16,000g for 15 min to clear large cell debris. The supernatant was

then isolated and used for further in vitro analysis. Using the expressed GFP, enzyme

Page 29: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

18

concentrations were measured by GFP ELISA kit (Cell Biolabs, Inc, CA, USA). For purification,

the 6 histidine tag (6-His) constructs were expressed in HEK293 cells as above. After harvesting

and binding to Ni-nitrilotriacetic acid resin, the 6-His tag enzymes were eluted with 500 mM

imidazole according to the supplier’s instructions (Capturem His-tagged purification kit, TaKaRa

Clontech, CA, USA). The fractions that contained the enzymes were desalted and concentrated

in the above assay buffer (Amicon Ultra 30 kD, Milllipore, MA, USA). The protein

concentration of 6-His tag enzymes were determined by the Bradford assay (Bradford, 1976).

2.2 In Vitro photoactivation assay by ELISA

2.2.1 One photon illumination

In vitro photoactivation of HEK293 cell lysate containing PhPDE4 or PhPDE5 was performed

on paraffin film (Parafilm M, Sigma-Aldrich, MO, USA) covered glass slides at room

temperature in the dark. The lysate buffer contained 40 mM HEPES/Na, pH 8.0, 0.1 mM EGTA,

5 mM magnesium acetate, 1 mM DTT, 0.01% Tween-20. For each experiment, 2 µM cAMP or

cGMP was added to 100 µl of the cell lysate, then photoactivated with a 455 nm LED (5

mW/mm2; ThorLabs, NJ, USA). 60 µl of the samples were immediately added to 540 µl 0.1M

HCl to stop the reaction and the level of cAMP or cGMP was measured in triplicates for each

sample using direct ELISA biochemical assay (Enzo Life Sciences, NY, USA). All ELIZA data

was normalized to the starting amount of cAMP or cGMP at time zero and subtracted by the

empty vector control to remove any endogenous PDE activity in HEK293 cell lysate. Then the

amount of cAMP or cGMP degraded was divided by the concentration of PhPDE4 or PhPDE5 in

the lysate. Initial velocities of the enzymatic activity of PhPDEs were further divided by the

illumination time and the power of light used per surface area (mW/cm2). The duration of LED

light (0,5,15,30,60 seconds) and (0, 0.25, 0.5, 1.0, 1.5, 2.0 millisecond) was controlled by a LED

driver (DC2100, Thorlabs, NJ, USA). To examine the excitation wavelength-dependency for

LED light, we used LEDs which have various excitation peak (385, 455, 590, 660 nm, 10

mW/cm2, 30 sec, Thorlabs, UJ, USA).

Page 30: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

19

2.2.2 Two photon illumination

For in vitro LAPD cell lysate, photoactivation of the samples were excited with focal light under

60X objective lens with two-photon laser light (30 seconds, 1,040 nm; 10 mW). For PhPDE4 and

PhPDE5 cell lysate and LAPD, PhPDE4 and PhPDE5 purified protein photoactivation, a two-

photon microscope (Custom made FV1000 MPE; Olympus, Tokyo, Japan, 60x objective lens,

NA 1.0 equipped with Spectra-Physics MaiTai HP DeepSee and InSight DeepSee) was used with

two-photon laser light (800 – 1,300 nm, 4 mW). Two-photon light dependent activity of PhPDE4

and PhPDE5 was measured at various time points (0, 2.5, 5.0, 15, 30 seconds) in the dark or after

illumination (1,100nm; 4mW). Two-photon wavelength dependency was analyzed by

illuminating lysate for 30 seconds with different wavelengths of light (800 nm, 900 nm, 1000

nm, 1,100 nm, 1,200 nm, 1,300 nm; 4 mW) (Multiphoton FVMPE-RS, 40x objective, 0.8 NA

WD 3.3mm Spectra-Physics InSight DeepSee at Scarborough imaging facility, University of

Toronto). For light intensity dependence different intensities were verified by the photodiode

power sensor (Thor Labs, S132C, Ge 700-1,800 nm, 500 mW) (5 seconds at 1, 100 nm; 0, 0.06,

0.25, 0.5, 1.0, 2.0, 4.0 mW).

2.3 Expression and Validation of PhPDEs in Neurons

2.3.1 Slice Culture

Slice culture: 1 ml of slice culture media (80 % Minimal Essential Media with 20 % horse serum

(sigma), 1 ug/ml insulin, 30 mM HEPES, 1 mM CaCl2, 2 mM MgSO4, 5 mM NaHCO3, 16.5

mM D-glucose and 0.5 mM ascorbate; pH to 7.3, 320-330 osmolarity) and one 0.4 𝜇m millicell

insert was added to each well of a 6-well plate. The plate was placed in a 37 °C, 95 % O2, 5 %

CO2 incubator. Organotypic slice cultures of rat hippocampi were prepared from postnatal day 6-

7 Sprague-Dawley rats. Rats were decapitated, following the guidelines of the university network

animal care committee approved protocol and oversight, and the whole brain removed and

placed on a petri dish containing filter paper covered with frozen, oxygenated cutting solution.

First, the brain was separated along the midline. Then the cerebellum was gently removed along

with the brainstem, midbrain and thalamus revealing the hippocampus. Using a spatula, the

hippocampus was gently scoped out and placed in ice cold dissection medium until the second

hippocampus was removed. Then hippocampi were cut into 400 𝜇m thick transverse sections.

Slices were separated in cold dissection medium and transferred onto each well of the 6-well

Page 31: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

20

plate and incubated at 37 °C, 95 % O2, 5 % CO2. Slice culture media was refreshed every other

day (Gogolla, Galimberti, DePaola, & Caroni, 2006).

Gene gun bullet preparation: Hard tubing was dried (15 mins) using nitrogen (< 10 PSI). 100 μl

of spermidine was mixed with 12 mg of gold followed by the addition of 200 μM of DNA and

100 μL of 0.5 M cold CaCl2. Mixture was then vortexed for 6 seconds and left at room

temperature for 10 minutes. Once precipitated, tube was spun down and gold particles were

washed with 1mL of 100% ethanol 3 times. Gold particles were suspended in 1 x

polyvinylpyrrolidone (PVP). The gold-DNA-PVP mixture was drawn up with a syringe and

attached to the gene gun prep station by connecting a piece of soft tubing on the syringe to the

hard tubing on the gene gun preparation station. Gold-DNA-PVP solution was injected into the

tubing and left at room temperature for 2 minutes before the liquid was removed. Then tubing

was rotated 180° for 1 minute. Then nitrogen was turned on for 6 minutes at (0.4 pressure).

Tubing was then removed and cut into cartridges and stored in a 20 mL scintillator vial

containing Drierite (CaSO4) at -20°C (Woods & Zito, 2008).

2.3.2 Expression in Neurons

Neuron transfection: Neurons in the organotypic hippocampal slice were biolistically

cotransfected using a gene gun (120 psi helium) with DsRed and GFP-PhPDE4 or GFP-PhPDE5

or GFP-LAPD at a ratio of 1:3. Imaging and experimentation occurred 3-5 days after transfection

using an Olympus FV1000MPE based custom-made two-photon microscope (60x objective

LUMPLFLN NA 1.0, Spectra-Physics MaiTai HP DeepSee and InSight DeepSee) with 900 nm

InSight DeepSee laser excitation. Post processing of images and line scan measurements were

acquired using Metamorph software.

2.3.3 Structural Long Term Potentiation (sLTP) experiments

For sLTP experiments: slices were perfused in regular ACSF solution containing: 119 mM NaCl,

2.5 mM KCl, 4 mM CaCl2, 4 mM MgCl2, 26.2 mM NaHCO3, 1 mM NaH2PO4 and 11 mM

glucose at 30 °C equilibrated with 5 % CO2/ 95 % O2. For glutamate uncaging experiments,

ACSF was the same as above including 2.5 mM 4-methoxy-7-nitroindolinyl (MNI)-L-glutamate

with or without the addition of 50 μM forskolin.

Page 32: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

21

sLTP was induced in the targeted dendritic spine by two photon uncaging of MNI glutamate

(720 nm laser, 4 ms duration pulses repeated at 1Hz for 60 times) the focal light point was ~1 µm

from the tip of the spine head (Kim et al., 2015). 50 µM forskolin was added into the uncaging

ACSF and applied during sLTP induction for cAMP-dependent sLTP induction (forskolin

ACSF). After uncaging the forskolin contained ACSF was returned to regular ACSF. To

photoactivate postsynaptic PhPDE4, the target spines were excited at 1,100 nm (continuous

point-scan 30 sec, 4 mW) immediately before uncaging for sLTP induction with forskolin.

Spine size change was measured by RFP (DsRed) fluorescence intensity of dendritic spines in

the three-dimensional z-stack (0.5 µm intervals) images of the dendrites typically composed of

20-30 sections using Imaris software. The average basal spine fluorescence intensity before

uncaging was defined as 100%. The spine intensity over time was subtracted from the

background intensity as well as corrected for any changes in fluorescence intensity over time of

the dendrite (arbitrary units).

2.4 Calculations and Statistical Analysis The number of replications for each experiment and statistical methods are indicated in the figure

legend. Data are presented as mean ± SEM. Statistical significance is indicated in figures as * p

< 0.05, ** p < 0.01, *** p < 0.005, and **** p < 0.001.

Page 33: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

22

Chapter 3 Photoactivatable Phosphodiesterase 4 (PhPDE4)

Photoactivatable Phosphodiesterase 4 (PhPDE4) 3

3.1 Introduction cAMP is a critical second messenger involved in a myriad of biological processes. cAMP

is synthesized by adenylyl cyclase (AC) and degraded by a family of phosphodiesterases (PDEs).

In the brain the regulators of cAMP signaling, AC and cAMP specific PDE4 are both expressed

in regions critical for learning and memory such as the hippocampus and cortex (Kelly et al.,

2014; Xia, Choi, Wang, Blazynski, & Storm, 1993).

cAMP signaling plays an important role in synaptic plasticity that underlies learning and

memory (Brunelli et al., 1976). Learning and memory occurs through activity dependent

structural and functional changes at the synapse level known as long-term potentiation (LTP).

LTP is separated into two phase’s early-LTP (E-LTP) and late LTP (L-LTP) which coincide with

short term and long-term memory formation (Huang & Kandel, 1994; Nguyen, Abel, & Kandel,

1994). cAMP signaling during L-LTP induces critical cAMP-dependent protein synthesis

inducing long lasting functional plasticity changes. Furthermore, the application of forskolin, a

potent activator of AC, during LTP induction resulted in an enhanced enlargement of the

dendritic spine (Govindarajan, Israely, Huang, & Tonegawa, 2011). This suggests a link between

cAMP signaling and CaMKII, a major postsynaptic protein that triggers LTP and detaches from

actin enabling actin polymerization before re-associating with actin and maintaining the

enlargement of the cytoskeleton and potentiating the dendritic spine (Kim et al., 2015; Okamoto

et al., 2004). However, how cAMP is linked to this pathway inducing dendritic spine structural

changes is not clear, although cAMP signaling is important for both the structural and functional

synaptic plasticity changes, which are critical for learning and memory.

Improper regulation of cAMP signaling has been associated with numerous neurological

or neurodegenerative diseases such as Schizophrenia or Alzheimer’s disease (Gamo et al., 2013).

Specifically, a reduction in AC activity and subsequently cAMP production was found in the

brains of individuals with Alzheimer’s disease. Furthermore, the inhibition of PDE4 causing

prolonged cAMP signaling, greatly improved cognition in numerous mouse models of

Page 34: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

23

Alzheimer’s disease (Cheng et al., 2010; Sierksma et al., 2014). Moreover, there is a common

mutation found in individuals with Schizophrenia called disrupted in schizophrenia (DISC1),

which impairs the binding of PDE4 to DISC1, which is located in dendritic spines, inhibiting

PDE4 activity in this subcellular domain (McGirr et al., 2016). This particular mutation is

associated with deficits in both working memory and long-term memory processes (Brandon,

2016; Gamo et al., 2013). This further highlights the critical role of cAMP signaling at the

synapse level and its effect on learning and memory.

Pharmacological application of forskolin or various PDE inhibitors results in widespread

alterations of cAMP signaling in the brain. This limits our understanding the role of cAMP in

LTP and learning and memory. Furthermore, these methods cannot address how cAMP alters

structural plasticity changes at a single dendritic spine level. Optogenetic approaches to produce

or suppress cAMP with light enable the precise temporal and spatial control of intracellular

signaling in living neurons.

Using our established two-photon optogenetic microscopy techniques with a modified

photoactivatable adenylyl cyclase (PAC) which produces cAMP (Stierl et al., 2011), our lab

showed that increasing post-synaptic cAMP signaling within a single dendritic spine during E-

LTP enhanced the structural plasticity and functions to regulate plasticity at nearby structures

(unpublished data). Thus, revealing a novel rapid role of cAMP signaling in structural plasticity

at the single spine level. To compliment these results and understand cAMP pathway for both

structural and functional plasticity, I will develop a novel two-photon optogenetic tool to

specifically degrade cAMP upon light illumination.

In order to generate a new optogenetic enzyme to specifically degrade cAMP, I employed

the light sensitive domain structure for LAPD but replaced the catalytic domain to the cAMP

specific PDE4 (Gasser et al., 2014). To validate the function of this optogenetic enzyme I

analyzed its photochemical properties including specificity for cAMP, light dependent activation

in vitro, wavelength dependency and light sensitivity using both one-photon and two-photon

illumination. I then applied this in living neurons for manipulating cAMP at the single dendritic

spine.

Page 35: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

24

3.2 Results

3.2.1 Design of Photoactivatable Phosphodiesterase 4 (PhPDE4) The previously reported LAPD was developed by fusing the light sensitive phytochrome domain

from Deinococcus radiodurans to the catalytic domain of PDE2A which can bind and degrade

both cAMP and cGMP (Gasser et al., 2014). To obtain a photoactivatable PDE, which

specifically degrades only cAMP I replaced the catalytic domain of LAPD with the cAMP

specific PDE4 catalytic domain. First, I compared the catalytic domain sequences of PDE4B

with PDE2A and found the corresponding region of PDE4B where the PDE2A catalytic domain

was fused to the light sensitive bacterial phytochrome domain (PAS-GAF-PHY) for LAPD

(Gasser et al., 2014). This comparison narrowed down a potential region of the PDE4 catalytic

domain which could be used as the fusion site to join to the PAS GAF-PHY domain to create a

cAMP specific light sensitive PDE (Figure 3-1).

α-helical coiled coil structures have been commonly identified across proteins, including

PDEs, which form the linker between the regulatory domain and the catalytic domain (Pandit,

Forman, Fennell, Dillman, & Menniti, 2009). The length of this coiled coil region effects the

torsional strain between domains critical for the enzyme structure as well as activity induced

conformation changes (Hartmann, 2017). Therefore, to align the catalytic domain of PDE4 to the

coiled coil domain terminal of the phytochrome in LAPD, I chose the α-helical rich region that

can integrate with the coiled coil structure. Interestingly, the first LAPD construct (named

LAPD+2) that connected the PAS-GAF-PHY domain to the PDE2A catalytic domain had no

detectable catalytic activity upon illumination; however deletion of 2 residues created the fully

functioning LAPD (Gasser et al., 2014). Since the length of the coiled coil region connecting the

two domains is critical for optimized activity I created a series of single amino acid deletions

from the α-helical region of the PDE4 catalytic domain and fused to the PAS-GAF-PHY domain.

The fusion of this light sensitive phytochrome domain with each variant of the PDE4 catalytic

domain were named Photoactivatable PDE4 or PhPDE4. The variant number after each PhPDE4

refers to the difference of the amino acid position from the corresponding catalytic domain of

LAPD (Figure 3-2).

Page 36: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

25

Figure 3-1. Protein sequence alignment of the C-terminal catalytic domains in PDE2A used

for LAPD and PDE4B. The fusion site for the previously reported LAPD is where the end of

the light sensitive phytochrome domain from Deinococcus radiodurans (highlighted in green)

meets the yellow highlighted region of PDE2A catalytic domain. To determine the appropriate

site of the PDE4 catalytic domain to generate a new light sensitive PDE4, I aligned the catalytic

domains of PDE4B and PDE2A. The sequence alignment provided a starting point for which

region of the PDE4B catalytic domain should be used for the new light sensitive PDE4. I also

confirmed the coiled coil domain structure using protein prediction software.

3.2.2 Optimization of the PhPDE4 light dependent photoactivation To determine if any of these fusion protein variants were successful in generating light sensitive

phosphodiesterase activity, I transfected each variant into HEK293 cells and used the cell extract

to compare the photoactivation by measuring the amount of cAMP hydrolysis using an ELISA

assay (Figure 3-2). The light stimulation induced their photo activation and PhPDE4 variant -13

demonstrated the highest light-dependent activity and was used for further characterization

experiments as PhPDE4 (Photoactivatable PDE4).

Page 37: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

26

Figure 3-2. Design of photoactivatable PDE4 variants and their activity. (A) Depiction of the

fusion between the light sensitive phytochrome domain from Deinococcus radiodurans to the

conserved catalytic domain of PDE4 to specifically degrade cAMP. The amino acid sequences

are shown (green-segment of the phytochrome domain, yellow – portion of the PDE4 catalytic

domain). The variant numbers reflect the difference of the amino acid position from the

corresponding catalytic domain of LAPD. (B) Comparison of photoactivity between the variants

of PhPDE4. The light and dark dependent activity were measured by exposing each variant to 15

seconds in the dark or after blue light illumination (455 nm) and measuring the amount of

degraded cAMP using an ELISA biochemical assay. Data are mean ± SEM n = 9 (3 biological

replicates and 3 experimental replicates)

3.2.3 Characterization of PhPDE4 using one-photon light (LED) To examine PhPDE4 substrate specificity, I measured cAMP or cGMP dependent

photoactivation of PhPDE4. Using 455 nm blue light, PhPDE4 induced significant and rapid

degradation of cAMP but showed little effect on the level of applied cGMP, indicating rapid

activation upon illumination and cAMP specificity. The activity in the dark condition was very

limited, demonstrating light dependent activation of PhPDE4. Next I examined the response time

of PhPDE4 to light on and off. PhPDE4 photoactivation occurred in less than a millisecond

duration of light indicating rapid activation. To examine the off response of enzymatic activity, I

measured any subsequent PhPDE4 activity after the removal of light and found little additional

PhPDE4 activity after light-off, indicating rapid inactivation of PhPDE4. Along with rapid

activation, rapid deactivation upon removal of light is critical for temporal precision of this

enzyme (Figure 3-3).

Page 38: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

27

Figure 3-3. cAMP dependent photoactivity of PhPDE4 has a rapid on and off response in

vitro. (A) Schematic of one-photon activation of PhPDE4 in vitro (B) Time course of PhPDE4

activity in vitro. The photoactivity was measured in the dark (black), or after illumination

measured by the amount of cAMP (magenta) or cGMP (blue) degraded. Data are mean ± SEM

(light+cAMP n = 4, dark+cAMP n = 3, light+cGMP n = 3). (C) Rapid time course of

photoactivation of PhPDE4. PhPDE4 was stimulated by various short millisecond durations of

light. (D) Time course of off response of PhPDE4. The off response was measured immediately

after turning off the light, after 5 seconds of illumination (plotted PhPDE4 activity to 0) and

measured any subsequent activity. Data for (C) and (D) are mean ± SEM n = 12 (4 biological

replicates, 3 experimental).

Page 39: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

28

To examine the sensitivity of PhPDE4 to the light intensity for photoactivation, I next measured

the activity among different light intensities. The PhPDE4 activity was detected from 0.01

W/cm2 and reached a plateau by more than 0.05 W/cm2 light. The power requirement suggests

the possibility of sufficient photoactivation of PhPDE4 in the deep brain region using a LED

coupled fiber optics, which has a maximum output power of 5mW. Also, I examined the

photoactivation light wavelength-dependency of the PhPDE4. By measuring PhPDE4 activation

after exposure of various LED lights from 385 nm – 660 nm, PhPDE4 showed the broad

photoactivation of this enzyme, which was similar to the wavelength dependency with LAPD

containing the same phytochrome light sensitive domain (Figure 3-4) (Gasser et al., 2014).

According to this result, I will use 455 nm LED light for PhPDE4 photoactivation for further in

vivo studies.

Figure 3-4 PhPDE4 is activated by various light intensities and wavelengths using one-

photon light. (A) Schematic diagram representing the use of one photon blue light to measure

the intensity dependence of PhPDE4 as well as different LED to measure wavelength-

dependency of PhPDE4. (B) Light (455 nm) intensity dependence of PhPDE4 activation. The

intensity dependence of PhPDE4 was measured in vitro by exposing PhPDE4 for 15 seconds

(0.001 W/cm2 - 0.618W/cm2) (C) Wavelength dependent photo activation of PhPDE4.

Wavelength dependency was measured by activating PhPDE4 for 15 seconds in vitro using

different wavelengths of light (385, 455, 590, 660 nm; 10 mW). Data are mean ± SEM n = 9 (3

biological replicates, 3 experimental).

Page 40: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

29

3.2.4 Two-photon characterization of PhPDE4

In order to activate PhPDE4 at the single synapse level in the brain tissue, two-photon laser

excitation is necessary as its focal volume is limited to the same size of a synapse with high NA

objective lens (~1µm3) (Denk et al., 1990). The properties of two-photon activation for LAPD,

which contains the same light sensitive phytochrome domain, have not been tested. Therefore, to

examine the two-photon wavelength dependency of PhPDE4, I measured the two-photon

excitation spectra of PhPDE4 in vitro. As the light sensitive phytochrome domain has far red

light sensitivity around 700 nm one photon excitation (Gasser et al., 2014), I used a two-photon

microscope, which covers wavelengths from 800 nm to 1,300 nm. I found the longer two-photon

excitation wavelengths 1,100-1,300nm efficiently activated PhPDE4 in vitro more than shorter

wavelengths, corresponding to the predicted two-photon excitation from the one-photon

excitation of the phytochrome domain. I used 1,100 nm two-photon excitation for further

experiments. PhPDE4 degraded cAMP in seconds using 1,100 nm illumination indicating rapid

seconds order manipulation of cAMP by two photon light. Next, to determine the necessary two-

photon excitation light intensity for the photoactivation of PhPDE4, I measured the activity in

response to various light intensities in vitro. The enzymatic activity was detected from 0.06 mW

two-photon excitation light and reached the plateau at 0.1 mW indicating sufficient two-photon

power for manipulating cAMP at synapses in the brain tissue. These results demonstrate rapid

and wavelength dependent two-photon photoactivation of PhPDE4 in vitro.

Page 41: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

30

Figure 3-5. PhPDE4 is more efficiently activated by longer two-photon excitation and

shows rapid and light intensity dependent activation at 1,100 nm. (A) Schematic drawing of

two-photon excitation of PhPDE4 in vitro. (B) Two-photon excitation spectra for PhPDE4. 100ul

of PhPDE4 cell extract was stimulated by various wavelengths of two-photon excitation light for

30 seconds (800, 900, 1000, 1100, 1200, 1300 nm; 4 mW). (C) Time course of two-photon

photoactivation of PhPDE4 in vitro. PhPDE4 strongly degraded cAMP at 1,100 nm

(2.96±0.5μM/min/nMPhPDE4) (D) Two-photon excitation intensity dependency of PhPDE4

activation. PhPDE4 was photoactivated at 1,100 nm for 5 seconds by various two-photon

intensities (0.01 - 4.0 mW). For (B), (C), (D) data are mean ± SEM, n = 9 (3 biological

replicates, 3 experimental).

Page 42: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

31

3.2.5 Establish soluble and membrane bound forms of PhPDE4

Endogenous PDE4 has membrane bound and soluble forms which result in important regulation

of cAMP signaling in neurons (Maurice et al., 2014). To mimic the intracellular distribution of

PDE4, I prepared a membrane bound form of PhPDE4 in addition to the soluble form by

incorporating the GAP-43 membrane localization sequence (Gauthier-Kemper et al., 2014) at the

N-terminus of the light sensitive phytochrome domain. After the photoactivation of both

membrane bound and soluble forms of PhPDE4 in vitro I found similar enzymatic activation

(455 nm, 15 seconds) of membrane bound form of PhPDE4 compared to the soluble form

(Figure 3-6). Next to observe the subcellular localization of the soluble and membrane bound

forms of PhPDE4, I biolistically co-transfected GFP-PhPDE4 with RFP (DsRed2) into

organotypic hippocampal cultured slices. The two-photon RFP fluorescence images show CA1

pyramidal neurons with dendritic spines. The GFP signal for the PhPDE4 in the dendrite was

similar to the RFP, but in the membrane bound form, GFP showed strong fluorescence intensity

at the membrane, demonstrating the expression of soluble and membrane bound PhPDE4 in CA1

hippocampal neurons (Figure 3-7).

Figure 3-6. In vitro one-photon photoactivation between membrane bound and soluble

forms of PhPDE4. (A) Schematics of domain formation of soluble and membrane bound forms

of PhPDE4. A GAP-43 membrane localization signal was inserted into the N-terminal of the

light sensitive domain. (B) Photoactivation of soluble and membrane bounds forms of PhPDE4

in vitro (455 nm, 15 seconds, 0.5 W/cm2) Data are mean ± SEM, n = 9 (3 biological replicates, 3

experimental).

Page 43: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

32

Figure 3-7. Subcellular localization of membrane bound and soluble forms of PhPDE4 in

living neurons (A) Schematic drawing of biolistic transfection of soluble and membrane bound

forms of PhPDE4 with DsRed2. (B) Depiction of two-photon imaging of transfected CA1

pyramidal neurons in organotypic hippocampal slices for observation of subcellular localization

between soluble and membrane bound PhPDE4. (C) Representative two-photon fluorescence

live imaging of the membrane bound or soluble forms of PhPDE4. GFP (green) signal indicates

subcellular localization of PhPDE4. RFP (red) signal used as a control. (D) Soluble PhPDE4 and

(E) membrane bound PhPDE4 fluorescence profile of the white line on (C).

Page 44: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

33

3.2.6 Validation of PhPDE4 activity in living neurons

Pharmacological application of an adenylate cyclase activator forskolin indicates that increasing

cAMP signaling enhances structural enlargement of the dendritic spine during strong synaptic

activation (Govindarajan et al., 2011) (Matsuzaki et al., 2001). Using this property, I tested

whether photo-activation of PhPDE4 can suppress the cAMP-dependent structural enhancement

of dendritic spines in living neurons. To induce cAMP dependent structural plasticity (sLTP) I

used forskolin application and two photon caged glutamate uncaging (Matsuzaki et al., 2001). 4-

Methoxy-7-nitroindolinyk-caged-L-glutamate (MNI glutamate) is a form of glutamate that is

inactive in the dark due to a photosensitive linker region blocking the glutamate function. Using

two-photon laser excitation (720nm), caged glutamate can be photolysed nearby the target spine

enabling single spine functional and structural LTP induction (10mW, 4 msec, 1 Hz, 60 times)

(Pettit D. L. et al., 1997). Using this protocol on a single targeted dendritic spines of CA1

hippocampal neurons expressing GFP-PhPDE4 and DsRed2 (volume filler), I induced structural

enlargement of the dendritic spine, called structural potentiation (sLTP) (Figure 3-8B).

Photoactivation of PhPDE4 (1, 100nm, 30sec) with the glutamate uncaging did not alter sLTP.

(Figure 3-8B)(Matsuzaki, Honkura, Ellis-Davies, & Kasai, 2004).

Next, I induced cAMP-dependent sLTP by glutamate uncaging in the presence of forskolin. The

induction of LTP with increased cAMP signaling caused further potentiated spine structure

measured at 30 minutes (Figure 3-8D). In our lab we found that postsynaptic cAMP is necessary

for structural enhancement within 30 seconds of uncaging (unpublished data). Therefore, next I

examined whether activation of PhPDE4 blocks the cAMP structural effect. To do this I photo

activated PhPDE4 using 1,100 nm for 30 seconds immediately before uncaging in the presence

of forskolin. The activation of PhPDE4 at the target spine blocked the cAMP dependent

structural enhancement, but was similar to the uncaging only sLTP (Figure 3-8D). These results

indicate that, PhPDE4 photoactivation blocked the cAMP dependent enhancement of sLTP but

not the induction of sLTP by uncaging (Figure 3-9).

Page 45: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

34

Figure 3-8. PhPDE4 activation at single dendritic spines suppresses cAMP effect during

structural LTP (A) Depiction of sLTP induction protocol using glutamate uncaging (720 nm

excitation) and two-photon imaging (860 nm) with and without stimulation of PhPDE4. (B)

Time course imaging of baseline sLTP (black) and PhPDE4 stimulation sLTP (blue). Structural

plasticity changes at target dendritic spines were observed by measuring RFP fluorescence

intensity change over time after glutamate uncaging (720 nm, 10 mW; 4 msec, 1 Hz, 60 times)

(black) n = 11(11 spines, 9 neurons), and after PhPDE4 stimulation + glutamate uncaging (blue)

n = 10 (10 spines, 8 neurons). (C) Depiction of sLTP induction in the presence of forskolin with

and without PhPDE4 photoactivation. (D) Time course imaging of glutamate uncaging with 50

μM forskolin (magenta) n = 13 (13 spines, 8 neurons) (magenta) and the effect of PhPDE4

photoactivation in cAMP-dependent sLTP (yellow). PhPDE4 was stimulated for 30 seconds

using 1,100 nm two-photon excitation prior to glutamate uncaging with forskolin (yellow) n = 13

(13 spines, 9 neurons).

Page 46: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

35

Figure 3-9. PhPDE4 activation at single dendritic spine causes little change during sLTP

induction but significantly suppresses the cAMP effect during cAMP dependent sLTP. (A)

Two-photon fluorescent (RFP) image of a CA1 pyramidal neuron from organotypic hippocampal

slice showing a dendrite with dendritic spines expressing GFP-PhPDE4 + RFP (DsRed2). The

while dot represents the position of glutamate uncaging at a target spine. Blue circle represents a

target spine for PhPDE4 stimulation. (B) Summary of time course imaging of sLTP without

PhPDE4 stimulation (black) and with PhPDE4 stimulation (Blue) as well as cAMP dependent

sLTP with PhPDE4 stimulation (yellow) and without (magenta). (C) Comparison of the

induction of sLTP. Structural plasticity changes between experimental groups were observed by

measuring RFP fluorescence intensity of target dendritic spines immediately after sLTP

induction (1 minute). N.S denotes not significant (P>0.05); 1 way Anova. (D) Comparison of the

potentiation of target spines. Potentiation differences between experimental groups were

observed by measuring RFP fluorescence intensity of target dendritic spines at 30minutes after

sLTP induction. **** denotes significant differences (P<0.0001) Data are mean ± SEM, n = 11

(black), n = 10 (blue) 13 (magenta), 12 (yellow).

Page 47: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

36

3.2.7 Photoactivation of PhPDE4 for future mouse behavioral experiments

To examine whether our custom-made LED which will be implanted in the brains of freely

moving mice is sufficient to photoactivate PhPDE4, I measured the activity over various time

points. The photoactivation of PhPDE4 showed the activation in the seconds order indicating

possible suppression of cAMP signal in the brain for learning and memory experiments (Figure

3-10).

Figure 3-10. In vitro photoactivation of PhPDE4 using the implantable fiber optic LED

light source for in vivo mouse experiments (A) Schematic drawing of the implant LED in the

mouse brain and a picture of the unilateral LED light. These LEDs can also have bilateral fibers

to illuminate the PhPDE4 enzyme in both brain hemispheres. (B) In vitro time course experiment

of PhPDE4 with the custom made LED light (455 nm). Data are mean ± SEM, n = 9 (3

biological replicates, 3 experimental).

Page 48: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

37

3.3 Summary of Chapter 3 This chapter described the development and characterization of a novel photoactivatable enzyme,

PhPDE4. This is the first optogenetic enzyme to specifically degrade the second messenger

cAMP by light. I validated the properties of photoactivation including the time-course and

substrate specificity as well as its rapid on and off response. Also, for the possibility of PhPDE4

activation in living neurons, specifically at the single dendritic spine level, I validated the

photoactivation properties with two-photon excitation light in vitro and demonstrated an

application to suppress cAMP-dependent structural plasticity (sLTP) in CA1 hippocampal

neurons. The generation of PhPDE4 will enable the investigation of cAMP signaling from the

single dendritic spine, to neural circuit and even whole brain level.

Page 49: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

38

Chapter 4 Photoactivatable Phosphodiesterase 5 (PhPDE5)

Photoactivatable Phosphodiesterase 5 (PhPDE5) 4

4.1 Introduction The cyclic nucleotide cyclic guanosine 3’, 5’-monophosphate (cGMP) is a critical second

messenger involved in many biological processes ranging from cardiovascular, gastrointestinal

to the nervous system (Puzzo, Sapienza, Arancio, & Palmeri, 2008). cGMP is synthesized by

guanylyl cyclase (GC) and degraded by a family of phosphodiesterases (PDEs). In the brain, the

hippocampus contains a large expression of GC and cGMP specific-PDEs (Burette et al. 2002;

Szabaditis et al. 2007).

The canonical cGMP-signaling pathway, widely studied in the CA1 neurons of the

Schaffer Collateral pathway of the hippocampus during LTP induction includes NMDA receptor

mediated Ca2+ influx which stimulates Ca2+/calmodulin-activated neuronal NO synthase (nNOS)

causing NO production in the postsynaptic structure (Haley, Wilcox, & Chapman, 1992). NO

acts as a retrograde messenger and translocates into the presynaptic structure where it activates

GC to produce cGMP and initiates the increase of neurotransmitter release aiding in the

potentiation of the synapse (Hawkins, Son, & Arancio, 1998). Also, the NO activates post-

synaptic GC to produce cGMP and trigger PKG to regulate the protein synthesis, highlighting

the role of cGMP in functional plasticity and long-term memory formation (Bollen et al., 2014).

Bath application of pharmacological inhibitors of GC (LY83583), PKG (KT5823) or nNOS (3-

Br 7-Ni) blocked LTP induction in cultured rat hippocampal slices (Komsuoglu-Celikyurt et al.,

2011; Zhuo, Hu, Schultz, Kandel, & Hawkins, 1994). Furthermore, weak tetanic stimulation

does not produce LTP however the pairing of weak tetanic stimulation with injected cGMP

analogues can (Haley et al., 1992).

cGMP signaling is also involved in learning and memory processes. Application of an

nNOS inhibitor (3-Br 7-NI) or GC inhibitor (ODQ) significantly impaired both reference and

working memory (Komsuoglu-Celikyurt et al., 2011). Also, inhibition of GC and PKG impaired

memory retention in the passive avoidance task in chicks (Edwards, Rickard, & Ng, 2002).

Administration of PDE5 inhibitor, vardenafil, improved object recognition memory in different

Page 50: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

39

memory deficit rodent models (Reneerkens et al., 2012) Furthermore, mice with PKG knockout

show deficient spatial learning using the morris water maze; a hippocampal dependent memory

process (Wincott et al., 2013). cGMP pathway has also been implicated in various brain diseases

such as Alzheimer’s disease. In Alzheimer’s disease patients, there is a strong decrease in cGMP

levels in the cerebral spinal fluid (CSF) as well as an up-regulation of PDE5 expression in the

temporal cortex (Ugarte et al., 2015) suggesting a link between cGMP signaling and disease

progression (Domek-Lopacinska, van de Waarenburg, Markerink-van Ittersum, Steinbusch, & de

Vente, 2005). Inhibition of PDE5 with sildenafil or tadalafil produced cognitive benefits in

numerous mouse models of Alzheimer’s disease (Garcia-Osta et al., 2012). Current knowledge

of cGMP signaling stems from pharmacological or genetic manipulations of cGMP-hydrolyzing

PDEs or GC activators to manipulate cGMP signaling in the brain. These approaches lack the

temporal control of local cGMP levels, which could results in off target effects or unrelated

behavioral modifications for studying the role of cGMP.

Recently we established two-photon activation of a light sensitive GC (BLGC) to

increase cGMP signaling at target dendritic spines on CA1 pyramidal neuron (Ryu, Moskvin,

Siltberg-Liberles, & Gomelsky, 2010). Utilizing this optogenetic approach, we found that an

increase in cGMP signaling suppressed the cAMP-dependent structural enhancement of dendritic

spines (sLTP) (unpublished data). This experiment revealed a novel role for cGMP signaling in

regulating structural plasticity at dendritic spines. To complement the optogenetic approach, I

developed a novel optogenetic tool to independently degrade cGMP by generating a light

sensitive PDE5. PDE5 is structurally similar to PDE2 as both contain an N-terminal tandem

GAF domain, which is similar to the bacterial phytochrome domain in LAPD (Gasser et al.,

2014; Maurice et al., 2014). The main difference between PDE5 catalytic domain and PDE2

catalytic domain is its cGMP-specific binding site (Francis, Blount, & Corbin, 2011). In order to

generate a light sensitive cGMP-specific PDE, I replaced and optimized the catalytic domain of

PDE5 into LAPD. This optogenetic tool, with BLGC, enables us to increase and decrease cGMP

signaling during LTP induction at the single dendritic spine level to understand its role during

structural and functional plasticity that underlies LTP and learning and memory.

Page 51: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

40

4.2 Results

4.2.1 PhPDE5 enzyme design To obtain a photoactivatable PDE that specifically degrades only cGMP I replaced the catalytic

domain of LAPD (Gasser et al., 2014) with the cGMP-specific PDE5 catalytic domain. I used the

same strategy described for PhPDE4 (page 24), to determine the appropriate fusion site of the

bacterial phytochrome domain (PAS-GAF-PHY) to the catalytic domain of PDE5. To summarize

briefly, I compared the protein sequences of PDE5A with PDE2A to find the corresponding

region of PDE5 where the PDE2 catalytic domain was fused to the bacterial phytochrome

domain for LAPD (Figure 4-1). Then I used protein prediction software to determine which

residues in this region created α helices which would facilitate the formation of an α helix coiled

coil structure between the phytochrome domain and PDE5 catalytic domain (Hartmann, 2017).

Then, similar to PhPDE4 enzyme design I created a series of variants where a single amino acid

was deleted from the beginning of the PDE5 catalytic domain to optimize the length of the α-

helical region. The fusions of this light sensitive domain with each variant of the PDE5 catalytic

domain were named Photoactivatable PDE5 or PhPDE5. The variant number refers to the

difference of the amino acid position form the corresponding catalytic domain of LAPD (Figure

4-2).

Page 52: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

41

Figure 4-1. Protein sequence alignment of the C-terminal catalytic domains in PDE2A used

for LAPD and PDE5A. The fusion site for the previously published LAPD is where the end of

the light sensitive phytochrome domain from Deinococcus radiodurans (highlighted in green)

meets the yellow highlighted region of PDE2A catalytic domain (Gasser et al., 2014). To

determine the appropriate start site of the PDE5 catalytic domain to generate a new light

sensitive PDE5 I aligned PDE5A and PDE2A. The sequence alignment provided a starting point

for which region of the PDE5A catalytic domain should be used for the new light sensitive

PDE5. Then I used protein prediction software to determine which residues of PDE5A contained

α helical structures.

4.2.2 Optimization of PhPDE5 light dependent photoactivation To determine if any of these PhPDE5 fusion protein variants were successful in generating light

sensitive phosphodiesterase activity, I transfected each variant into HEK293 cell lysate and used

the cell extract to compare the photoactivation by measuring the amount of cGMP degraded

using an ELISA assay (Figure 4-2). PhPDE5 variant -3 demonstrated the highest light-dependent

activity and was used for further characterization experiments as PhPDE5.

Page 53: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

42

Figure 4-2. Design of photoactivatable PDE5 variants and their activity. (A) Depiction of the

fusion between the light sensitive phytochrome domain from Deinococcus radiodurans to the

conserved catalytic domain of PDE5 to specifically degrade cGMP. The amino acid sequences

are shown (green-segment of the phytochrome domain, yellow – portion of the PDE5 catalytic

domain). The variant numbers reflect the difference of the amino acid position from the

corresponding catalytic domain of LAPD. (B) Comparison of photoactivity between the variants

of PhPDE5. The light and dark dependent activity were measured by exposing each variant to 15

seconds in the dark or after one-photon illumination (455 nm) and measuring the amount of

added cGMP degraded using an ELISA biochemical assay. Data are mean ± SEM n = 9 (3

biological replicates and 3 experimental replicates)

4.2.3 Characterization of PhPDE5 using one-photon light (LED) To examine the substrate specificity of PhPDE5, I measured the amount of cGMP or cAMP

degraded after photoactivation of PhPDE5. Using 455 nm blue light, PhPDE5 induced rapid

degradation of cGMP with little effect on cAMP concentration, indicating rapid activation and

cGMP specificity. The activity in the dark condition was limited, demonstrating light dependent

activation of PhPDE5. I next examined the response time of PhPDE5 to light on and off

conditions. PhPDE5 was activated by less than a millisecond duration of light indicating rapid

activation. To examine the off response of enzymatic activity, I measured any subsequent

PhPDE5 activity after the removal of light and found little additional PhPDE5. Therefore, along

with rapid activation, I validated the rapid deactivation of PhPDE5 upon removal of light, which

is critical for the temporal precision of this enzyme (Figure 4-3).

Page 54: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

43

Figure 4-3. cGMP dependent photoactivity of PhPDE5 has a rapid on and off response in

vitro. (A) Schematic of one-photon activation of PhPDE5 in vitro (B) Time course of PhPDE5

activity in vitro. The photoactivity was measured in the dark (black), or after illumination

measured by the amount of cAMP (magenta) or cGMP (blue) degraded. Data are mean ± SEM

(light+cAMP n = 12, dark+cAMP n = 9, light+cGMP n = 9). (C) Rapid time course of

photoactivation of PhPDE5. PhPDE5 was stimulated by various short millisecond durations of

light. (D) Time course of off response of PhPDE5. The off response was measured immediately

after turning off the light, after 5 seconds of illumination (plotted PhPDE5 activity to 0) and

measured any subsequent activity. Data for (C) and (D) are mean ± SEM n = 12 (4 biological

replicates, 3 experimental).

Page 55: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

44

The sensitivity of PhPDE5 to different light intensities for photoactivation will provide necessary

details to determine appropriate light sources to activate PhPDE4 deep in the brains of living

animals. I measured the PhPDE5 activity in vitro in response to increasing light intensities.

PhPDE5 was activated by 0.01 mW/cm2 and reached a plateau around 0.05 mW/cm2. The low

power requirements suggest the potential to activate PhPDE5 expressed deep within brain tissue

using fiber optic LED implants. Also, I examined the wavelength-dependency for activating

PhPDE5. By measuring PhPDE5 activity after exposure to different wavelengths of LED light

(385, 455, 590, 660 nm; 10 mW) I found that PhPDE5 has broad photoactivation similar to

PhPDE4 and LAPD as they all contain the same light sensitive phytochrome domain (Figure 4-4)

(Gasser et al., 2014). Furthermore, like PhPDE4, I will use 455 nm light to activate PhPDE5 in

vivo using customized fiber optic LED implants. These results indicate that PhPDE5 is

successfully activated by one-photon light in vitro.

Figure 4-4 PhPDE5 is activated by various light intensities and wavelengths using one-

photon light. (A) Schematic diagram representing the use of one photon blue light to measure

the intensity dependence of PhPDE5 as well as different LED to measure wavelength-

dependency of PhPDE5. (B) Light (455 nm) intensity dependence of PhPDE5 activation. The

intensity dependence of PhPDE5 was measured in vitro by exposing PhPDE5 for 15 seconds

(0.001- 0.618 mW/cm2) (C) Wavelength dependent photo activation of PhPDE5. Wavelength

dependency was measured by activating PhPDE5 for 15 seconds in vitro using different

wavelengths of light (385, 455, 590, 660 nm; 10 mW). Data are mean ± SEM n = 9 (3 biological

replicates, 3 experimental).

Page 56: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

45

4.2.4 Two-photon characterization of PhPDE5

Two-photon laser excitation has a focal volume around ~1µm3 with high NA objective lens

which can localize activation of PhPDE5 at a single targeted dendritic spine in deep brain tissue

(Denk et al., 1990). Therefore, similar to PhPDE4, I determined the two-photon wavelength

dependency of PhPDE5 to determine best excitation spectra for photoactivation. As expected,

PhPDE5 two-photon excitation spectra closely resembles the PhPDE4 excitation spectra as they

both contain the same light sensitive phytochrome domain (Gasser et al., 2014). Specifically,

PhPDE5 was strongly activated by longer two photon wavelengths of light (1,100 nm -1,300 nm)

in vitro. I used 1,100nm excitation for further experiments and found that PhPDE5 degraded

cGMP in seconds, indicating the ability to manipulate by two-photon light. Next to determine the

necessary two-photon excitation light intensity for photoactivation of PhPDE5, I measured the

activity in response to various light intensities in vitro. The enzymatic activity was detected from

0.06mW two-photon excitation light and reached the plateau at 0.1 mW indicating sufficient

two-photon power for manipulating cGMP at synapses in the brain tissue. These results indicate

that PhPDE5 is successfully activated by two-photon light in vitro.

Page 57: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

46

Figure 4-5. PhPDE5 is more efficiently activated by longer two-photon excitation and

shows rapid and light intensity dependent activation at 1,100 nm. (A) Schematic drawing of

two-photon excitation of PhPDE5 in vitro. (B) Two-photon excitation spectra for PhPDE5. 100

ul of PhPDE5 cell extract was stimulated by various wavelengths of two-photon excitation light

for 30 seconds (800, 900, 1000, 1100, 1200, 1300 nm; 4 mW). (C) Time course of two-photon

photoactivation of PhPDE5 in vitro. PhPDE5 strongly degraded cGMP at 1,100 nm (D) Two-

photon excitation intensity dependency of PhPDE5 activation. PhPDE5 was photo activated at

1,100 nm for 5 seconds by various two-photon intensities (0.01 mW - 4 mW). For (B), (C), (D)

data are mean ± SEM, n = 9 (3 biological replicates, 3 experimental).

Page 58: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

47

4.2.5 Establish soluble and membrane bound forms of PhPDE5

cGMP signaling is regulated by both soluble and membrane bound PDEs which results in

important regulation of cGMP signaling in neurons (Menniti, Faraci, & Schmidt, 2006). Using

the same strategy described for PhPDE4 (page 40) I generated a membrane bound version of

PhPDE5 using the same GAP-43 membrane localization sequence. I found similar enzymatic

activation in vitro (455 nm, 15 seconds) of membrane bound form of PhPDE5 compared to the

soluble form (Figure 4-6). Next to observe the subcellular localization of the soluble and

membrane bound forms of PhPDE5, I biolistically co-transfected GFP-PhPDE5 with RFP

(DsRed2) into organotypic hippocampal cultured slices. The two-photon RFP fluorescence

images show CA1 pyramidal neurons with dendritic spines. The GFP signal for PhPDE5 in the

dendrite was similar to the volume filler RFP, but for the membrane bound form, GFP showed

strong fluorescence intensity just at the membrane, demonstrating the expression of soluble and

membrane bound PhPDE5 in CA1 hippocampal neurons (Figure 4-7).

Figure 4-6. In vitro one-photon photoactivation between membrane bound and soluble

forms of PhPDE5. (A) Schematics of domain formation of soluble and membrane bound forms

of PhPDE5. A GAP-43 membrane localization signal was inserted into the N-terminal of the

light sensitive domain. (B) Photoactivation of soluble and membrane bounds forms of PhPDE5

in vitro (455 nm, 15 seconds). Data are mean ± SEM, n = 9 (3 biological replicates, 3

experimental).

Page 59: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

48

Figure 4-7. Subcellular localization of membrane bound and soluble forms of PhPDE5 in

living neurons (A) Schematic drawing of biolistic gene gun transfection of soluble and

membrane bound forms of PhPDE5 with DsRed2. (B) Depiction of two-photon imaging of CA1

pyramidal neurons in organotypic hippocampal slices expressing PhPDE5 for observation of

subcellular localization between soluble and membrane bound forms. (C) Representative two-

photon fluorescence live imaging of the membrane bound or soluble forms of PhPDE5. GFP

(green) signal indicates subcellular localization of PhPDE5. RFP (red) signal used as a control.

(D) Soluble PhPDE5 and (E) membrane bound PhPDE5 fluorescence profile of the white line on

(C).

Page 60: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

49

4.2.6 Photoactivation of PhPDE5 for future mouse behavioral experiments

To examine whether PhPDE5 can be sufficiently activated in the brains of freely moving mice

using our custom-made implantable LEDs, I first measured the activity of PhPDE5 in vitro over

various time points using this light source. The photoactivation of PhPDE5 occurred in the

seconds order indicating possible suppression of cGMP signal in the brain for future learning and

memory experiments (Figure 4-8).

Figure 4-8. In vitro photoactivation of PhPDE5 using the implantable fiber optic LED light

source for in vivo mouse experiments (A) Schematic drawing of the implant LED in the mouse

brain and a picture of the unilateral LED light. These LEDs can also have bilateral fibers to

illuminate the PhPDE5 enzyme in both brain hemispheres. (B) In vitro time course experiment of

PhPDE5 with the custom made LED light (455 nm). Data are mean ± SEM, n = 9 (3 biological

replicates, 3 experimental).

4.3 Summary of Chapter 4

This chapter paralleled Chapter 3, describing the development and characterization of a novel

photoactivatable enzyme PhPDE5. This is the first optogenetic enzyme to specifically degrade

the second messenger cGMP. Similar to PhPDE4, I validated the properties of PhPDE5

photoactivation including the time course and substrate specificity and its rapid on and off

response. I also validated the two-photon photoactivation excitation properties in vitro and

examined the diffuse and membrane bound expression in vivo. The generation of PhPDE5 now

enables us to suppress cGMP signaling at a targeted dendritic spine as well as independently

block cGMP signaling in living animals during learning and memory tasks.

Page 61: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

50

Chapter 5 Comparison of photo activity between PhPDEs and LAPD

Comparison of photo activity between PhPDEs and 5LAPD

5.1 Introduction

All three light sensitive enzymes including a previously reported LAPD (Gasser et al.,

2014) to degrade cAMP and/or cGMP are composed of the light sensitive phytochrome domain

from Deinococcus radiodurans fused to the catalytic domain of PDE2, PDE4 or PDE5 (Gasser et

al., 2014). Due to the substrate specificity of the catalytic domain, this strategy was successful

for engineering three different light sensitive PDEs which all retain their cAMP/cGMP

specificity. There are 11 families of PDEs, which are classified according to their substrate

specificity, amino acid sequences, and regulatory processes (Bender & Beavo, 2006). PDEs are

modular enzymes containing an N-terminal regulatory domain and a C-terminal catalytic domain

to hydrolyze cAMP or cGMP or both.

This chapter will compare the in vitro light sensitive activity of LAPD, PhPDE4 and

PhPDE5 with regular PDE activity. Also, I further characterized LAPD using two-photon

excitation to verify in vitro activation as well as the diffuse expression of this enzyme in vivo.

Page 62: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

51

5.2 Results

5.2.1 Comparison of the light sensitive activation between LAPD, PhPDE4 and PhPDE5.

To compare the previously reported light sensitive activity of LAPD (Gasser et al., 2014),

alongside PhPDE4 and PhPDE5, using our in vitro techniques, I illuminated cell lysate

containing LAPD using blue light (455 nm), which caused rapid degradation of both cAMP and

cGMP (Figure 5-1). The light dependent cAMP and cGMP hydrolysis are relatively lower than

PhPDE4 and PhPDE5.

Figure 5-1. In vitro photoactivation of PhPDE4, PhPDE5 and LAPD on cAMP and cGMP

levels. Summary of optogenetic toolkit, which degrades cAMP and/or cGMP using 455 nm light.

(B) Comparison of the initial activity in vitro of PhPDE4, PhPDE5 and LAPD with cAMP

(magenta) or cGMP (blue) plotted per min per W/cm2. The initial velocity of PDE activity is

plotted per minute per W/cm2. PhPDE4 activity for cAMP vs cGMP (p = 0.0004 ****), PhPDE5

activity for cAMP and cGMP (p = 0.0066**), and LAPD activity cAMP vs cGMP (p = 0.3, ns).

Statistical analysis: Mann Whitney test, one tailed.

Page 63: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

52

5.2.2 Two-photon activation of LAPD

In order to activate LAPD specifically at the single synapse level of living neurons in the brain

tissue, two-photon laser illumination is necessary. Due to the different properties of single

photon and two-photon light, I validated LAPD activation using two-photon illumination. I

found that LAPD degraded cAMP and cGMP after 15 seconds of 1,040 nm illumination,

indicating strong photoactivation by two-photon laser light (Figure 5-2).

Figure 5-2. Two-photon photoactivation of LAPD efficiently induces the degradation of

both cAMP and cGMP. (A) Schematic drawing of two-photon excitation of LAPD in vitro. (B)

Comparison of LAPD activity for cAMP and cGMP in vitro. The photoactivity of LAPD with

1,040 nm two-photon light. Data are mean ± SEM, n = 9 (3 biological replicates, 3

experimental).

Page 64: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

53

5.2.3 Photoactivation properties of purified LAPD, PhPDE4 and PhPDE5

To determine if the kinetics of PhPDE4 and PhPDE5 optogenetic enzymes are similar to the

previously reported LAPD under one-photon (660 nm) and two-photon excitation (1, 100 nm), I

measured their photochemical properties using purified proteins and compared the

photoactivation efficiency among these optogenetic tools. PhPDE5 has slightly higher

photoactivation compared to PhPDE4 and LAPD using both one photon and two photon

illumination (Table 1).

Table 1. Photoactivation of purified LAPD, PhPDE4 and PhPDE5 using one-photon and two-photon excitation light.

5.2.4 Establish soluble and membrane bound forms of LAPD

PDE2 has membrane bound and soluble forms which results in important regulation of cAMP

and cGMP signaling in neurons (Bender & Beavo, 2006). Therefore, along with soluble LAPD,

to prepare a membrane bound form of LAPD, I examined the addition of two different

membrane localization sequences to the N terminal of the phytochrome domain: the PDE2

membrane localization sequence (MLS) and the GAP-43 membrane localization sequence. I

biolistically co-transfected GFP-LAPD with RFP (DsRed2) into organotypic hippocampal

cultured slices. The GFP signal for the soluble LAPD in the dendrite showed similar distribution

to the RFP, but for the membrane bound forms, GFP showed strong fluorescence intensity at the

membrane for GAP-43 and aggregated expression for MLS membrane bound form (Figure 5-3).

Page 65: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

54

Figure 5-3. Subcellular localization of membrane bound and soluble forms of LAPD in

living neurons (A) Schematic drawing of gene gun transfection of soluble and membrane bound

forms of LAPD with DsRed2. (B) Depiction of two-photon imaging of CA1 pyramidal neurons

in organotypic hippocampal slices expressing LAPD for observation of subcellular localization

between soluble and membrane bound forms. (C) Representative two-photon fluorescence live

imaging of the LAPD-mem, LAPD-MLS or LAPD-soluble. GFP (green) signal indicates

subcellular localization of LAPD. RFP (red) signal used as a control. (D) Soluble LAPD and (E)

LAPD-mem fluorescence profile of the white line on (C).

Page 66: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

55

5.2.5 Preparation of LAPD lenti viral construct for in vivo applications

In order to use this optogenetic enzyme as well as PhPDE4 or PhPDE5 in vivo to study the effect

of degrading cAMP and/or cGMP level, I used lenti virus system with CaMKII promoter, which

restricts expression to the excitatory neurons (Wang et al., 2013; Feng Zhang et al., 2010). I

expressed and validated the CaMKII-GFP-LAPD plasmid in HEK293 cells by analyzing the

expression of GFP using confocal microscopy. Next I confirmed the photoactivity of this

construct in HEK293 cells (Figure 5-4).

Figure 5-4. Validate expression and light dependent activity of pLenti-LAPD in HEK293

cells. (A) Lentivirus under the CaMKII promoter will be used to limit expression of LAPD to

excitatory neurons in the hippocampus. (B) HEK293 cells were transfected with CaMKII-GFP-

LAPD plasmid. Fluorescent images verified transfection efficiency. Images were obtained using

a confocal microscope (10X objective, 475 nm excitation). Scale bar represents 50 μm. (C)

Photoactivity of CaMKII-GFP-LAPD. Using 455 nm light, LAPD photoactivation was

confirmed in the presence of 4 μM of cAMP after 2 minutes of illumination (p<0.05, n = 3). Data

(n = 3). Statistical significance was calculated using paired t-test.

Page 67: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

56

5.3 Summary of Chapter 5 This chapter described the in vitro activation of LAPD to compare with PhPDE4 and PhPDE5. I

confirmed the one-photon photoactivation of LAPD as well as the two-photon photoactivation.

To compare their photochemical properties, I purified LAPD, PhPDE4 and PhPDE5 and

measured their photoactivation kinetics under one-photon and two-photon light stimulation in

vitro. I also expressed the soluble form of LAPD and the membrane bound form of LAPD in

CA1 hippocampal neurons and validated their localization in vivo. Furthermore, for future mouse

behavioral experiments I validated a lentiviral construct in vitro to restrict LAPD expression to

hippocampal neurons. The generation of PhPDE4 and PhPDE5 along with LAPD will enables us

to study cAMP/cGMP signaling at a targeted dendritic spine as well as deep in the brain tissue of

living animals during learning and memory tasks.

Page 68: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

57

Chapter 6 Discussion

This chapter is divided into 6 sections. Section 6.1 is a summary of the project. Section 6.2

discusses photoactivation properties of PhPDEs. Section 6.3 discusses in vivo application of

PhPDE4 in living neurons. 6.4 Highlights the comparisons of kinetics and limitations of these

new optogenetic tools. Section 6.5 discusses future work and important neuroscience questions

that can be addressed using these tools to conclude the thesis.

Discussion 6

6.1 Summary of the project We established the optogenetic toolkit for manipulating cAMP and cGMP with the necessary

spatiotemporal precision to study their roles in neuronal activity underlying learning and

memory. This cyclic nucleotide optogenetic toolkit contains the previously reported bacterial

photoactivatable adenylyl cyclase (bPAC) which produces cAMP in response to blue light (Stierl

et al., 2011), the blue light sensitive guanylyl cyclase which produces cGMP (Ryu et al., 2010)

and a far red light sensitive, light activatable phosphodiesterase (LAPD) which degrades both

cAMP and cGMP (Gasser et al., 2014).

In this thesis, I established two optogenetic enzymes, PhPDE4 and PhPDE5 that degrade second

messenger molecules, cAMP and cGMP, independently upon light illumination. These

optogenetic tools provide us with great spatial and temporal control of cAMP and cGMP

signaling in an unprecedented way. By controlling the light-dependent activation, this enables us

to study endogenous cAMP/cGMP functions from millisecond to minutes order to understand

their rapid role in synaptic functions and related learning and memory. Furthermore, I optimized

two-photon laser light-dependent activation of PhPDE4 and PhPDE5 in vitro and validated the

function at single synapse level in living neurons. Thus, PhPDE4 and PhPDE5 will provide novel

powerful tools to study cAMP/cGMP signaling from the synapse level to the brain’s cognitive

functions.

Page 69: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

58

6.2 Photoactive properties of PhPDEs By fusing the light sensitive domain from Deinococcus radiodurans to the catalytic domain of

PDE4 or PDE5 I generated two new light sensitive enzymes named photoactivatable PDE4

(PhPDE4) and photoactivatable PDE5 (PhPDE5) that independently degrade cAMP and cGMP

respectively. This basic design stemmed from LAPD where illumination caused the degradation

of both cAMP and cGMP (Gasser et al., 2014). Utilizing the similarity of a regulatory domain of

PDEs (GAF-B) and a light sensitive phytochrome domain (PHY), the phytochrome domain was

tightly connected to the catalytic domain of PDEs by linking their helical structures (Gasser et

al., 2014). Therefore, the predicted light-dependent conformational changes for photoactivation

are similar to the activation of PDEs (Figure 6-1).

Figure 6-1. Schematic diagram of the predicted photo cycle of PhPDEs. (A) Closed state

when no light is present. (B) Illumination induces conformational changes enabling (C) cAMP or

cGMP to bind to the catalytic domain causing their degradation. (D) Removal of light then

rapidly reverts the enzyme back into its closed conformational state impairing further

degradation of cAMP or cGMP.

Page 70: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

59

Under dark conditions, the conformation of the enzyme renders the binding site for cAMP or

cGMP on the catalytic domain blocked (Figure 6-1;A). Once illuminated, the light-sensitive

phytochrome domain absorbs photons of light inducing a rotation of the chromophore resulting

in a conformational change and catalytic activity (Figure 6-1;B). It is believed that this rotation

causes rearrangement of hydrogen bonds enabling the evolutionarily conserved PHY domain to

convert from B-sheet confirmation into an alpha helix (Ziegler & Moglich, 2015). This change in

phytochrome domain causes conformational changes in the attached catalytic domain resulting in

the opening of the enzyme and presenting the binding site for cAMP or cGMP (Figure 6-1; C).

As the catalytic domain of phosphodiesterases are highly conserved each contains a glutamine

binding pocket specific for either cAMP or cGMP, I hypothesized that the substrate specificity

should be maintained without the presence of their regulatory domain (Zhu, Yang, Dai, & Huang,

2013). In in vitro photoactivation experiments, I detected that PhPDE4 rapidly degrades cAMP

but not cGMP, and PhPDE5 rapidly degrades cGMP but not cAMP (Figure 6-1;C) (see figure 3-

3, 4-3). I also validated that PhPDE activity can be induced broadly by various one photon light

sources (385-660 nm) and is sensitive to low light intensities (0.01-0.05mW/cm2) (see Figure 3-

4, 4-4). Moreover this activation can occur within 0.25 milliseconds of illumination suggesting

the application for studying rapid cAMP/cGMP functions (see figure 3-3, 4-3).

PhPDEs are also sensitive to two-photon laser light illumination. This is critical to

degrade cAMP and/or cGMP at a single dendritic spine to study their role in synaptic plasticity

as two-photon focal points form within a micron excitation volume, similar to the size of a

dendritic spine, when using high numerical aperture (NA) objective lenses, making this

technique suitable for manipulating light-sensitive enzymes at the single synapse level deep

within living brain tissue (Denk et al., 1990). I found that the longer two-photon excitation

wavelength, which corresponds to the single photon excitation wavelength efficiently, activated

PhPDE4, PhPDE5 and LAPD (see figures 3-5, 4-5, 5-2). I also confirmed that after the removal

of light, catalytic activity of these light sensitive enzymes ceased (Figure 6-1;D) (see figure 3-3,

4-3). Thus, PhPDE4 and PhPDE5 serve as the first optogenetic enzymes to independently

degrade cAMP or cGMP by light.

Page 71: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

60

6.3 In vivo validation of PhPDEs in living neurons To verify that these tools can be applied for neuroscience research, at first I expressed PhPDE4

and PhPDE5 as well as LAPD in living neurons (see figures 3-7, 4-7 and 5-3). All three light

sensitive PDEs showed homogenous expression in CA1 pyramidal neurons of hippocampal

slices. As PDEs have membrane bound forms in addition to soluble forms, I prepared new

membrane bound forms of light sensitive PDEs. Endogenous PDE2A localizes to synaptosomal

membranes, therefore to generate membrane bound forms I first inserted the membrane

localization sequence from this PDE to LAPD. However, this construct caused aggregated

expression throughout the hippocampal neurons. I next tried a commonly used membrane

localization sequence from Growth Associated Protein 43 (GAP-43), which attaches to the

neuron membrane by dual palmitoylation targeting GAP-43 to lipid rafts (Gauthier-Kemper et

al., 2014). Using GAP-43 membrane localization sequence, LAPD was successfully localized to

the membrane and the same strategy was applied for PhPDE4 and PhPDE5 (see figures 3-7, 4-7

and 5-3).

To validate the functionality of these optogenetic enzymes in vivo, I applied PhPDE4 to validate

the role of cAMP signaling on structural plasticity. During LTP induction, there is an increase in

the size of the dendritic spine, which persists over 30 mins called structural LTP (sLTP). We

found that post-synaptic cAMP enhances sLTP. By reducing the cAMP signaling by

photoactivation of PhPDE4 at the target spine during cAMP-dependent sLTP, PhPDE4

successfully blocked this cAMP-dependent enhancement (See figure 3-8, 3-9). Since the

difference between PhPDE4, PhPDE5 and LAPD is the catalytic domain containing the substrate

specific site for cyclic nucleotide-binding, these results suggest the functions of PhPDEs in living

systems.

6.4 Comparisons and limitations of PhPDE4, 5 and LAPD

To compare the optogenetic enzymes to the previously published LAPD, I measured

photoactivation of LAPD, PhPDE4 and PhPDE5 under one-photon and two-photon illumination

using the purified proteins (see table 1). Comparing the reported Vmax of endogenous PDE2,

PDE4 and PDE5 activity to the activity of LAPD, PhPDE4 and PhPDE5 some major differences

Page 72: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

61

arise. Strikingly, the activity of LAPD was dramatically lower for both cAMP and cGMP

compared to the PDE2 activity of 123 µM cGMP/min/mg and 120 µM cAMP/min/mg,

suggesting that photoactivation of LAPD does not fully activate the PDE2 catalytic domain

(Rosman et al., 1997). While, PhPDE4 activity was slightly higher than the known PDE4 range

(0.0078-0.4 µM/min/mg) (Huston et al., 1997), and PhPDE5 activity was only slightly lower

than the known PDE5 range (1.0-1.3 µM/min/mg) (Loughney et al., 1998). This similarity

suggests that photoactivation of PhPDEs produces an almost maximally activated enzymatic

state. Moreover, it has been reported that endogenous PDE5 has higher enzymatic activity than

PDE4, which is a trend that I saw with PhPDE4 and PhPDE5. Also, PDE2 has higher activity for

cGMP degradation than cAMP degradation that is also retained in the light sensitive activity of

LAPD. Therefore, the optogenetic enzymes, despite being engineered to only turn on upon

illumination still retain similar enzymatic properties compared to endogenous form. These results

suggest that the photo activation of PhPDE4 and PhPDE5 in living neurons has the potential to

mimic the same level of activity as endogenous PDEs.

One limitation to PhPDEs is their broad spectrum of activation by one-photon illumination.

Although the phytochrome domain from Deinococcus radiodurans is known as a far red light

sensitive domain, I found that PhPDE4 and PhPDE5 were sensitive to a broad range of one-

photon wavelengths (see figures 3-4, 4-4) (Gasser et al., 2014). For example during

electrophysiology experiments on hippocampal slices, one could co-transfect PhPDE4 and PAC

(blue light sensitive adenylyl cyclase) with the intent to increase and decrease cAMP signaling in

the same cells using different wavelengths of light. Using this example, while PhPDE4 could be

independently activated using 660 nm light without stimulating PAC, but activation of PAC

using 455 nm blue light will also activate PhPDE4. In order to achieve this type of

experimentation, optogenetic enzymes with narrow wavelength specificity and far enough

spectral separation will be required. In contrast, spectral differences using two-photon

illumination such that PhPDE4 and 5 were best activated by more than 1,100 nm two-photon

wavelengths, whereas PAC and BLGC are activated by 700-800 nm two-photon excitation. This

could be a possibility for independent activation of PhPDE4 with PAC for regulation of cAMP

and PhPDE5 with BLGC for cGMP using two-photon illumination. Co-expressing these pairs

would enable the activation or suppression of cAMP or cGMP signaling in a single dendritic

spine during synaptic plasticity.

Page 73: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

62

6.5 Future Applications It is well established that both cAMP and cGMP are important for long term memory through the

activation of CREB and generation of plasticity related proteins. PhPDE4 or PhPDE5 will enable

us to suppress endogenous cAMP/cGMP functions to study the roles of cAMP/cGMP in

cognitive function. To validate the functionality and correct optogenetic setup to activate

PhPDE4 and PhPDE5 in living animals for future behavioral experiments, the novel object

recognition test will be used to observe the effect of blocking cAMP or cGMP on memory

formation. After this validation, PhPDE4 or PhPDE5 can be stimulated with varying spatial and

temporal dimensions during novel object recognition for short or long term memory, or 5 choice

serial reaction time task for attentional memory to understand the effect of suppressing the

spatiotemporal cAMP or cGMP signaling on cognitive output. Furthermore, these enzymes can

be used to suppress the rapid cAMP or cGMP functions during LTP or LTD induction using

electrophysiology on rodent hippocampal slices. Finally, using our established two-photon

optogenetic approach, I will monitor the effect of suppressing cAMP or cGMP signaling at

single targeted dendritic spines on structural plasticity.

6.6 Conclusion cAMP and cGMP are universal second messengers for intracellular signaling critical for a

myriad of biological functions. cAMP and cGMP have been implicated in synaptic plasticity and

their misregulation has been associated with a variety of neurodegenerative disorders.

Pharmacological, genetic and electrophysiology techniques have shown the correlative role of

cAMP and cGMP in learning and memory. To further examine the role of cAMP or cGMP

during this activity dependent process requires a method to manipulate their signaling within a

biologically relevant timescale while restricting such activation spatially as to leave neighboring

cells unaffected. Optogenetic tools enable perturbations to each of the three levels of neural

activity with this spatiotemporal accuracy. To complement previously characterized PAC and

BLGC, which produce cAMP and cGMP respectively, I have created and validated two new

optogenetic tools to independently suppress cAMP or cGMP signaling for the first time using

light. The completion of this optogenetic toolkit will enable the accurate dissection of cAMP and

cGMP during activity dependent synaptic plasticity.

Page 74: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

63

References Abel, T., Nguyen, P. V., Barad, M., Deuel, T. A., Kandel, E. R., & Bourtchouladze, R. (1997).

Genetic demonstration of a role for PKA in the late phase of LTP and in hippocampus-based long-term memory. Cell, 88(5), 615-626.

Bender, A. T., & Beavo, J. A. (2006). Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev, 58(3), 488-520. doi: 10.1124/pr.58.3.5

Bliss, T. V., & Lomo, T. (1973). Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit following stimulation of the perforant path. J Physiol, 232(2), 331-356.

Boess, F. G., Hendrix, M., van der Staay, F.-J., Erb, C., Schreiber, R., van Staveren, W., . . . Koenig, G. (2004). Inhibition of phosphodiesterase 2 increases neuronal cGMP, synaptic plasticity and memory performance. Neuropharmacology, 47(7), 1081-1092. doi: 10.1016/j.neuropharm.2004.07.040

Bollen, E., Puzzo, D., Rutten, K., Privitera, L., De Vry, J., Vanmierlo, T., . . . Prickaerts, J. (2014). Improved long-term memory via enhancing cGMP-PKG signaling requires cAMP-PKA signaling. Neuropsychopharmacology, 39(11), 2497-2505. doi: 10.1038/npp.2014.106

Boyden, E. S., Zhang, F., Bamberg, E., Nagel, G., & Deisseroth, K. (2005). Millisecond-timescale, genetically targeted optical control of neural activity. Nat Neurosci, 8(9), 1263-1268. doi: 10.1038/nn1525

Bradford, M. M. (1976). A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem, 72, 248-254.

Brandon, N. J. (2016). Uncovering the function of Disrupted in Schizophrenia 1 through interactions with the cAMP phosphodiesterase PDE4: Contributions of the Houslay lab to molecular psychiatry. Cellular Signalling, 28(7), 749-752. doi: http://dx.doi.org/10.1016/j.cellsig.2015.09.019

Brunelli, M., Castellucci, V., & Kandel, E. R. (1976). Synaptic facilitation and behavioral sensitization in Aplysia: possible role of serotonin and cyclic AMP. Science, 194(4270), 1178-1181.

Cheng, Y. F., Wang, C., Lin, H. B., Li, Y. F., Huang, Y., Xu, J. P., & Zhang, H. T. (2010). Inhibition of phosphodiesterase-4 reverses memory deficits produced by Abeta25-35 or Abeta1-40 peptide in rats. Psychopharmacology (Berl), 212(2), 181-191. doi: 10.1007/s00213-010-1943-3

Collingridge, G. L., Kehl, S. J., & McLennan, H. (1983). Excitatory amino acids in synaptic transmission in the Schaffer collateral-commissural pathway of the rat hippocampus. J Physiol, 334, 33-46.

Page 75: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

64

Deisseroth, K. (2011). Optogenetics. Nature Methods, 8(1), 26-29. doi: 10.1038/nmeth.f.324

Denk, W., Strickler, J. H., & Webb, W. W. (1990). Two-photon laser scanning fluorescence microscopy. Science, 248(4951), 73-76.

Domek-Lopacinska, K., van de Waarenburg, M., Markerink-van Ittersum, M., Steinbusch, H. W., & de Vente, J. (2005). Nitric oxide-induced cGMP synthesis in the cholinergic system during the development and aging of the rat brain. Brain Res Dev Brain Res, 158(1-2), 72-81. doi: 10.1016/j.devbrainres.2005.06.003

Dudai, Y., Jan, Y. N., Byers, D., Quinn, W. G., & Benzer, S. (1976). dunce, a mutant of Drosophila deficient in learning. Proc Natl Acad Sci U S A, 73(5), 1684-1688.

Dugue, G. P., Akemann, W., & Knopfel, T. (2012). A comprehensive concept of optogenetics. Prog Brain Res, 196, 1-28. doi: 10.1016/b978-0-444-59426-6.00001-x

Edwards, T. M., Rickard, N. S., & Ng, K. T. (2002). Inhibition of guanylate cyclase and protein kinase G impairs retention for the passive avoidance task in the day-old chick. Neurobiol Learn Mem, 77(3), 313-326. doi: 10.1006/nlme.2001.4021

Erickson, E., Wakao, S., & Niyogi, K. K. (2015). Light stress and photoprotection in Chlamydomonas reinhardtii. Plant J, 82(3), 449-465. doi: 10.1111/tpj.12825

Francis, S. H., Blount, M. A., & Corbin, J. D. (2011). Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions. Physiol Rev, 91(2), 651-690. doi: 10.1152/physrev.00030.2010

Frey, U., Huang, Y. Y., & Kandel, E. R. (1993). Effects of cAMP simulate a late stage of LTP in hippocampal CA1 neurons. Science, 260(5114), 1661-1664.

Frey, U., Krug, M., Reymann, K. G., & Matthies, H. (1988). Anisomycin, an inhibitor of protein synthesis, blocks late phases of LTP phenomena in the hippocampal CA1 region in vitro. Brain Res, 452(1-2), 57-65.

Gamo, N. J., Duque, A., Paspalas, C. D., Kata, A., Fine, R., Boven, L., . . . Arnsten, A. F. (2013). Role of disrupted in schizophrenia 1 (DISC1) in stress-induced prefrontal cognitive dysfunction. Transl Psychiatry, 3, e328. doi: 10.1038/tp.2013.104

Garcia-Osta, A., Cuadrado-Tejedor, M., Garcia-Barroso, C., Oyarzabal, J., & Franco, R. (2012). Phosphodiesterases as therapeutic targets for Alzheimer's disease. ACS Chem Neurosci, 3(11), 832-844. doi: 10.1021/cn3000907

Gasser, C., Taiber, S., Yeh, C.-M., Wittig, C. H., Hegemann, P., Ryu, S., . . . Möglich, A. (2014). Engineering of a red-light–activated human cAMP/cGMP-specific phosphodiesterase. Proceedings of the National Academy of Sciences, 111(24), 8803-8808. doi: 10.1073/pnas.1321600111

Gauthier-Kemper, A., Igaev, M., Sundermann, F., Janning, D., Bruhmann, J., Moschner, K., . . . Brandt, R. (2014). Interplay between phosphorylation and palmitoylation mediates

Page 76: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

65

plasma membrane targeting and sorting of GAP43. Mol Biol Cell, 25(21), 3284-3299. doi: 10.1091/mbc.E13-12-0737

Gogolla, N., Galimberti, I., DePaola, V., & Caroni, P. (2006). Preparation of organotypic hippocampal slice cultures for long-term live imaging. Nat Protoc, 1(3), 1165-1171. doi: 10.1038/nprot.2006.168

Govindarajan, A., Israely, I., Huang, S.-Y., & Tonegawa, S. (2011). The Dendritic Branch Is the Preferred Integrative Unit for Protein Synthesis-Dependent LTP. Neuron, 69(1), 132-146. doi: http://dx.doi.org/10.1016/j.neuron.2010.12.008

Guglielmi, G., Falk, H. J., & De Renzis, S. (2016). Optogenetic Control of Protein Function: From Intracellular Processes to Tissue Morphogenesis. Trends in Cell Biology, 26(11), 864-874. doi: 10.1016/j.tcb.2016.09.006

Guzowski, J. F. (2002). Insights into immediate-early gene function in hippocampal memory consolidation using antisense oligonucleotide and fluorescent imaging approaches. Hippocampus, 12(1), 86-104. doi: 10.1002/hipo.10010

Haley, J. E., Wilcox, G. L., & Chapman, P. F. (1992). The role of nitric oxide in hippocampal long-term potentiation. Neuron, 8(2), 211-216.

Hartmann, M. D. (2017). Functional and Structural Roles of Coiled Coils. Subcell Biochem, 82, 63-93. doi: 10.1007/978-3-319-49674-0_3

Hawkins, R. D., Son, H., & Arancio, O. (1998). Chapter 11 Nitric oxide as a retrograde messenger during long-term potentiation in hippocampus. In T. M. D. V. L. D. R. Ranney Mize & J. F. Michael (Eds.), Progress in Brain Research (Vol. Volume 118, pp. 155-172): Elsevier.

Huang, Y. Y., & Kandel, E. R. (1994). Recruitment of long-lasting and protein kinase A-dependent long-term potentiation in the CA1 region of hippocampus requires repeated tetanization. Learn Mem, 1(1), 74-82.

Huston, E., Lumb, S., Russell, A., Catterall, C., Ross, A. H., Steele, M. R., . . . Houslay, M. D. (1997). Molecular cloning and transient expression in COS7 cells of a novel human PDE4B cAMP-specific phosphodiesterase, HSPDE4B3. Biochem J, 328 ( Pt 2), 549-558.

Idevall-Hagren, O., Dickson, E. J., Hille, B., Toomre, D. K., & De Camilli, P. (2012). Optogenetic control of phosphoinositide metabolism. Proc Natl Acad Sci U S A, 109(35), E2316-2323. doi: 10.1073/pnas.1211305109

Jenkins, G. I. (2017). Photomorphogenic Responses to Ultraviolet-B light. Plant Cell Environ. doi: 10.1111/pce.12934

Kaiser, W., & Garrett, C. G. B. (1961). Two-Photon Excitation in Ca${\mathrm{F}}_{2}$: ${\mathrm{Eu}}^{2+}$. Physical Review Letters, 7(6), 229-231.

Page 77: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

66

Kandel, E. R. (2001). The molecular biology of memory storage: a dialogue between genes and synapses. Science, 294(5544), 1030-1038. doi: 10.1126/science.1067020

Kandel, E. R. (2012). The molecular biology of memory: cAMP, PKA, CRE, CREB-1, CREB-2, and CPEB. Mol Brain, 5, 14. doi: 10.1186/1756-6606-5-14

Kelly, M. P., Adamowicz, W., Bove, S., Hartman, A. J., Mariga, A., Pathak, G., . . . Kleiman, R. J. (2014). Select 3',5'-cyclic nucleotide phosphodiesterases exhibit altered expression in the aged rodent brain. Cell Signal, 26(2), 383-397. doi: 10.1016/j.cellsig.2013.10.007

Kim, K., Lakhanpal, G., Lu, H. E., Khan, M., Suzuki, A., Hayashi, M. K., . . . Okamoto, K. (2015). A Temporary Gating of Actin Remodeling during Synaptic Plasticity Consists of the Interplay between the Kinase and Structural Functions of CaMKII. Neuron, 87(4), 813-826. doi: 10.1016/j.neuron.2015.07.023

Kolbe, M., Besir, H., Essen, L. O., & Oesterhelt, D. (2000). Structure of the light-driven chloride pump halorhodopsin at 1.8 A resolution. Science, 288(5470), 1390-1396.

Komsuoglu-Celikyurt, I., Gocmez, S. S., Mutlu, O., Gacar, N., Aricioglu, F., & Utkan, T. (2011). Evidence for the involvement of neuronal nitric oxide synthase and soluble guanylate cyclase on cognitive functions in rats. Life Sciences, 89(23–24), 905-910. doi: http://dx.doi.org/10.1016/j.lfs.2011.09.017

Krishnamurthy, V. V., Khamo, J. S., Mei, W., Turgeon, A. J., Ashraf, H. M., Mondal, P., . . . Zhang, K. (2016). Reversible optogenetic control of kinase activity during differentiation and embryonic development. Development, 143(21), 4085-4094. doi: 10.1242/dev.140889

Levin, L. R., Han, P. L., Hwang, P. M., Feinstein, P. G., Davis, R. L., & Reed, R. R. (1992). The Drosophila learning and memory gene rutabaga encodes a Ca2+/Calmodulin-responsive adenylyl cyclase. Cell, 68(3), 479-489.

Lieb, R. (2005). Anxiety disorders: clinical presentation and epidemiology. Handb Exp Pharmacol(169), 405-432.

Liu, X., Ramirez, S., Pang, P. T., Puryear, C. B., Govindarajan, A., Deisseroth, K., & Tonegawa, S. (2012). Optogenetic stimulation of a hippocampal engram activates fear memory recall. Nature, 484(7394), 381-385. doi: 10.1038/nature11028

Loughney, K., Hill, T. R., Florio, V. A., Uher, L., Rosman, G. J., Wolda, S. L., . . . Ferguson, K. (1998). Isolation and characterization of cDNAs encoding PDE5A, a human cGMP-binding, cGMP-specific 3',5'-cyclic nucleotide phosphodiesterase. Gene, 216(1), 139-147.

Lynch, G. S., Dunwiddie, T., & Gribkoff, V. (1977). Heterosynaptic depression: a postsynaptic correlate of long-term potentiation. Nature, 266(5604), 737-739.

Page 78: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

67

Matsuzaki, M., Ellis-Davies, G. C., Nemoto, T., Miyashita, Y., Iino, M., & Kasai, H. (2001). Dendritic spine geometry is critical for AMPA receptor expression in hippocampal CA1 pyramidal neurons. Nat Neurosci, 4(11), 1086-1092. doi: 10.1038/nn736

Matsuzaki, M., Honkura, N., Ellis-Davies, G. C., & Kasai, H. (2004). Structural basis of long-term potentiation in single dendritic spines. Nature, 429(6993), 761-766. doi: 10.1038/nature02617

Maurice, D. H., Ke, H., Ahmad, F., Wang, Y., Chung, J., & Manganiello, V. C. (2014). Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov, 13(4), 290-314. doi: 10.1038/nrd4228

McGirr, A., Lipina, T. V., Mun, H. S., Georgiou, J., Al-Amri, A. H., Ng, E., . . . Roder, J. C. (2016). Specific Inhibition of Phosphodiesterase-4B Results in Anxiolysis and Facilitates Memory Acquisition. Neuropsychopharmacology, 41(4), 1080-1092. doi: 10.1038/npp.2015.240

Menniti, F. S., Faraci, W. S., & Schmidt, C. J. (2006). Phosphodiesterases in the CNS: targets for drug development. Nat Rev Drug Discov, 5(8), 660-670. doi: 10.1038/nrd2058

Morris, R. G. (1999). D.O. Hebb: The Organization of Behavior, Wiley: New York; 1949. Brain Res Bull, 50(5-6), 437.

Morris, R. G., Anderson, E., Lynch, G. S., & Baudry, M. (1986). Selective impairment of learning and blockade of long-term potentiation by an N-methyl-D-aspartate receptor antagonist, AP5. Nature, 319(6056), 774-776. doi: 10.1038/319774a0

Nagel, G., Ollig, D., Fuhrmann, M., Kateriya, S., Musti, A. M., Bamberg, E., & Hegemann, P. (2002). Channelrhodopsin-1: a light-gated proton channel in green algae. Science, 296(5577), 2395-2398. doi: 10.1126/science.1072068

Nagel, G., Szellas, T., Huhn, W., Kateriya, S., Adeishvili, N., Berthold, P., . . . Bamberg, E. (2003). Channelrhodopsin-2, a directly light-gated cation-selective membrane channel. Proc Natl Acad Sci U S A, 100(24), 13940-13945. doi: 10.1073/pnas.1936192100

Nguyen, P. V., Abel, T., & Kandel, E. R. (1994). Requirement of a critical period of transcription for induction of a late phase of LTP. Science, 265(5175), 1104-1107.

Niwa, H., Yamamura, K., & Miyazaki, J. (1991). Efficient selection for high-expression transfectants with a novel eukaryotic vector. Gene, 108(2), 193-199.

Okamoto, K., Nagai, T., Miyawaki, A., & Hayashi, Y. (2004). Rapid and persistent modulation of actin dynamics regulates postsynaptic reorganization underlying bidirectional plasticity. Nat Neurosci, 7(10), 1104-1112. doi: 10.1038/nn1311

Pandit, J., Forman, M. D., Fennell, K. F., Dillman, K. S., & Menniti, F. S. (2009). Mechanism for the allosteric regulation of phosphodiesterase 2A deduced from the X-ray structure of a near full-length construct. Proc Natl Acad Sci U S A, 106(43), 18225-18230. doi: 10.1073/pnas.0907635106

Page 79: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

68

Puzzo, D., Sapienza, S., Arancio, O., & Palmeri, A. (2008). Role of phosphodiesterase 5 in synaptic plasticity and memory. Neuropsychiatric Disease and Treatment, 4(2), 371-387.

Ramirez, S., Tonegawa, S., & Liu, X. (2013). Identification and optogenetic manipulation of memory engrams in the hippocampus. Front Behav Neurosci, 7, 226. doi: 10.3389/fnbeh.2013.00226

Ramón y Cajal, S. (1995). Histology of the nervous system of man and vertebrates. New York :: Oxford University Press.

Reneerkens, O. A., Rutten, K., Akkerman, S., Blokland, A., Shaffer, C. L., Menniti, F. S., . . . Prickaerts, J. (2012). Phosphodiesterase type 5 (PDE5) inhibition improves object recognition memory: indications for central and peripheral mechanisms. Neurobiol Learn Mem, 97(4), 370-379. doi: 10.1016/j.nlm.2012.02.008

Rockwell, N. C., Duanmu, D., Martin, S. S., Bachy, C., Price, D. C., Bhattacharya, D., . . . Lagarias, J. C. (2014). Eukaryotic algal phytochromes span the visible spectrum. Proc Natl Acad Sci U S A, 111(10), 3871-3876. doi: 10.1073/pnas.1401871111

Rosman, G. J., Martins, T. J., Sonnenburg, W. K., Beavo, J. A., Ferguson, K., & Loughney, K. (1997). Isolation and characterization of human cDNAs encoding a cGMP-stimulated 3',5'-cyclic nucleotide phosphodiesterase. Gene, 191(1), 89-95.

Ryu, M.-H., Moskvin, O. V., Siltberg-Liberles, J., & Gomelsky, M. (2010). Natural and engineered photoactivated nucleotidyl cyclases for optogenetic applications. The Journal of Biological Chemistry, 285(53), 41501-41508. doi: 10.1074/jbc.M110.177600

Sierksma, A. S., van den Hove, D. L., Pfau, F., Philippens, M., Bruno, O., Fedele, E., . . . Prickaerts, J. (2014). Improvement of spatial memory function in APPswe/PS1dE9 mice after chronic inhibition of phosphodiesterase type 4D. Neuropharmacology, 77, 120-130. doi: 10.1016/j.neuropharm.2013.09.015

Stierl, M., Stumpf, P., Udwari, D., Gueta, R., Hagedorn, R., Losi, A., . . . Hegemann, P. (2011). Light Modulation of Cellular cAMP by a Small Bacterial Photoactivated Adenylyl Cyclase, bPAC, of the Soil Bacterium Beggiatoa. Journal of Biological Chemistry, 286(2), 1181-1188. doi: 10.1074/jbc.M110.185496

Sutherland, E. W. (1972). Studies on the mechanism of hormone action. Science, 177(4047), 401-408.

Taslimi, A., Vrana, J. D., Chen, D., Borinskaya, S., Mayer, B. J., Kennedy, M. J., & Tucker, C. L. (2014). An optimized optogenetic clustering tool for probing protein interaction and function. Nat Commun, 5, 4925. doi: 10.1038/ncomms5925

Toettcher, J. E., Voigt, C. A., Weiner, O. D., & Lim, W. A. (2011). The promise of optogenetics in cell biology: interrogating molecular circuits in space and time. Nat Methods, 8(1), 35-38. doi: 10.1038/nmeth.f.326

Page 80: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

69

Tye, K. M., Prakash, R., Kim, S. Y., Fenno, L. E., Grosenick, L., Zarabi, H., . . . Deisseroth, K. (2011). Amygdala circuitry mediating reversible and bidirectional control of anxiety. Nature, 471(7338), 358-362. doi: 10.1038/nature09820

Ueki, N., Ide, T., Mochiji, S., Kobayashi, Y., Tokutsu, R., Ohnishi, N., . . . Wakabayashi, K.-i. (2016). Eyespot-dependent determination of the phototactic sign in Chlamydomonas reinhardtii. Proceedings of the National Academy of Sciences of the United States of America, 113(19), 5299-5304. doi: 10.1073/pnas.1525538113

Ugarte, A., Gil-Bea, F., Garcia-Barroso, C., Cedazo-Minguez, A., Ramirez, M. J., Franco, R., . . . Cuadrado-Tejedor, M. (2015). Decreased levels of guanosine 3', 5'-monophosphate (cGMP) in cerebrospinal fluid (CSF) are associated with cognitive decline and amyloid pathology in Alzheimer's disease. Neuropathol Appl Neurobiol, 41(4), 471-482. doi: 10.1111/nan.12203

van Donkelaar, E. L., Rutten, K., Blokland, A., Akkerman, S., Steinbusch, H. W., & Prickaerts, J. (2008). Phosphodiesterase 2 and 5 inhibition attenuates the object memory deficit induced by acute tryptophan depletion. Eur J Pharmacol, 600(1-3), 98-104. doi: 10.1016/j.ejphar.2008.10.027

Wang, X., Zhang, C., Szábo, G., & Sun, Q.-Q. (2013). Distribution of CaMKIIα expression in the brain in vivo, studied by CaMKIIα-GFP mice. Brain research, 1518, 9-25. doi: 10.1016/j.brainres.2013.04.042

Weber, T., Renzland, I., Baur, M., Monks, S., Herrmann, E., Huppert, V., . . . Bartsch, D. (2012). Tetracycline inducible gene manipulation in serotonergic neurons. PLoS One, 7(5), e38193. doi: 10.1371/journal.pone.0038193

Wincott, C. M., Kim, S., Titcombe, R. F., Tukey, D. S., Girma, H. K., Pick, J. E., . . . Ziff, E. B. (2013). Spatial memory deficits and motor coordination facilitation in cGMP-dependent protein kinase type II-deficient mice. Neurobiology of Learning and Memory, 99, 32-37. doi: http://dx.doi.org/10.1016/j.nlm.2012.10.003

Woods, G., & Zito, K. (2008). Preparation of gene gun bullets and biolistic transfection of neurons in slice culture. J Vis Exp(12). doi: 10.3791/675

Wu, Y. I., Frey, D., Lungu, O. I., Jaehrig, A., Schlichting, I., Kuhlman, B., & Hahn, K. M. (2009). A genetically encoded photoactivatable Rac controls the motility of living cells. Nature, 461(7260), 104-108. doi: 10.1038/nature08241

Xia, Z., Choi, E. J., Wang, F., Blazynski, C., & Storm, D. R. (1993). Type I calmodulin-sensitive adenylyl cyclase is neural specific. J Neurochem, 60(1), 305-311.

Zeng, H., & Madisen, L. (2012). Mouse transgenic approaches in optogenetics. Prog Brain Res, 196, 193-213. doi: 10.1016/b978-0-444-59426-6.00010-0

Zhang, F., Gradinaru, V., Adamantidis, A. R., Durand, R., Airan, R. D., Lecea, L. d., & Deisseroth, K. (2010). Optogenetic interrogation of neural circuits: technology for

Page 81: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

70

probing mammalian brain structures. Nature Protocols, 5(3), 439-456. doi: 10.1038/nprot.2009.226

Zhang, F., Wang, L. P., Boyden, E. S., & Deisseroth, K. (2006). Channelrhodopsin-2 and optical control of excitable cells. Nat Methods, 3(10), 785-792. doi: 10.1038/nmeth936

Zhang, F., Wang, L. P., Brauner, M., Liewald, J. F., Kay, K., Watzke, N., . . . Deisseroth, K. (2007). Multimodal fast optical interrogation of neural circuitry. Nature, 446(7136), 633-639. doi: 10.1038/nature05744

Zhao, C. Y., Greenstein, J. L., & Winslow, R. L. (2016). Roles of phosphodiesterases in the regulation of the cardiac cyclic nucleotide cross-talk signaling network. Journal of molecular and cellular cardiology, 91, 215. doi: 10.1016/j.yjmcc.2016.01.004

Zhu, J., Yang, Q., Dai, D., & Huang, Q. (2013). X-ray crystal structure of phosphodiesterase 2 in complex with a highly selective, nanomolar inhibitor reveals a binding-induced pocket important for selectivity. J Am Chem Soc, 135(32), 11708-11711. doi: 10.1021/ja404449g

Zhuo, M., Hu, Y., Schultz, C., Kandel, E. R., & Hawkins, R. D. (1994). Role of guanylyl cyclase and cGMP-dependent protein kinase in long-term potentiation. Nature, 368(6472), 635-639. doi: 10.1038/368635a0

Ziegler, T., & Moglich, A. (2015). Photoreceptor engineering. Front Mol Biosci, 2, 30. doi: 10.3389/fmolb.2015.00030

Page 82: Two Novel Photoactivatable Phosphodiesterases to ...€¦ · Two Novel Photoactivatable Phosphodiesterases to Independently Hydrolyze cAMP and cGMP . Fiona Bergin . Master of Science

71