51
APPROVED: Douglas Root, Major Professor Robert Benjamin, Committee Member Gerard O’Donovan, Committee Member Sam Atkinson, Chair of the Department of Biological Sciences Mark Wardell, Dean of the Toulouse Graduate School SYNTHETIC PEPTIDES MODEL INSTABILITY OF CARDIAC MYOSIN SUBFRAGMENT-2 Nasrin Taei Thesis Prepared for the Degree of MASTER OF SCIENCE UNIVERSITY OF NORTH TEXAS August 2013

Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

APPROVED: Douglas Root, Major Professor Robert Benjamin, Committee Member Gerard O’Donovan, Committee Member Sam Atkinson, Chair of the Department of

Biological Sciences Mark Wardell, Dean of the Toulouse Graduate

School

SYNTHETIC PEPTIDES MODEL INSTABILITY OF CARDIAC MYOSIN SUBFRAGMENT-2

Nasrin Taei

Thesis Prepared for the Degree of

MASTER OF SCIENCE

UNIVERSITY OF NORTH TEXAS

August 2013

Page 2: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2.

Master of Science (Biology-Biochemistry and Molecular Biology). August 2013, 44 pp., 1 table,

18 figures, reference list, 37 titles.

Hypertrophic cardiomyopathy (HCM), a heart-related abnormality, is the most prevalent

cause of sudden death in young athletes at sporting events. A cluster of cardiomyopathy

mutations are localized in β-cardiac myosin at the N-terminal region of subfragment-2. Using

resonance energy transfer probes, a synthetic peptide model system was developed to study

stability of the coiled coil (S2 fragment) structure by determining monomer-dimer equilibrium

of the peptide. Fluorescence resonance energy transfer and MacroModel software suite were

used to obtain distance measurements along with measurement of coiled coil formation. The

model peptide was used to characterize the effects of disease-causing-mutations and examine

potential candidate drugs (polyamines) to counteract effects of mutations causing HCM.

Distance measurements between donor and acceptor probes obtained by

computational simulation and fluorescence resonance energy transfer (FRET) were consistent.

Measurements also agreed with simulations of unlabeled wildtype, indicating coiled coil

structural stability of the peptide. Interaction of the site-specific antibody with the peptide

strongly inhibited dimerization and destabilized coiled coil structure of the peptide. Presence of

negatively charged glutamate residues in the region of subfragment-2 strongly suggested a

potential interaction site for positively charged polyamines. Binding of certain polyamines, such

as poly-L-Lysine 11 residues and poly-D-Lysine 17 residues, demonstrated the ability to enhance

dimerization and improve stability of the coiled coil structure, while some other polyamines

were shown to have insignificant impact on the structure. In an attempt to characterize the

Page 3: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

effect of HCM-causing-mutations, peptides containing E924K mutation and lethal mutation

E930 deletion were synthesized. Fluorescence resonance probes were conjugated to the

mutant peptides to determine coiled coil formation. Results obtained from both dynamic

simulations and resonance energy transfer experiments indicated that these mutations strongly

inhibit dimerization, and thus, destabilize coiled coil structure of the peptide. Further

experiments were conducted using heterodimers containing a chain of wildtype and a chain of

mutant peptide. Both E924K & Edel930 mutations destabilized coiled coil formation and

prevented dimerization. This peptide model system would provide a promising tool for drug

development targeting HCM-causing-mutations along the S2 region of myosin.

Page 4: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Copyright 2013

by

Nasrin Taei

ii

Page 5: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

ACKNOWLEDGEMENTS

I would like to acknowledge my appreciation for the endless encouragement and

immeasurable support of my mentor and major professor, Dr. Douglas Root. I would also like to

thank the support of my family, friends and colleagues.

This research is based upon National Science Foundation grant 842736 and the

American Recovery and Reinvestment Act for support.

iii

Page 6: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

TABLE OF CONTENTS

Page

ACKNOWLEDGEMENTS ................................................................................................................... iii

LIST OF TABLES AND FIGURES .......................................................................................................... v

CHAPTER 1. INTRODUCTION ........................................................................................................... 1

1.1. Muscle Contraction ................................................................................................. 1

1.2. Structure and Function of Subfragment-2 (S2) ....................................................... 3

1.3. Clinical Significance ................................................................................................. 5

1.4. The Purpose of This Study ...................................................................................... 5

CHAPTER 2. MATERIALS AND METHODS ...................................................................................... 10

2.1. Fast Protein Liquid Chromatography (FPLC) ......................................................... 10

2.2. Reverse Phase Chromatography (RPC) ................................................................. 10

2.3. Ion-exchange Chromatography (IEX) .................................................................... 11

2.4. Fluorescence Resonance Energy Transfer (FRET) Overview ................................. 12

2.5. Structure of the Chelate Complex and Fluorescein Probe ................................... 14

2.6. Molecular Dynamic Simulation ............................................................................. 16

2.7. Experimental Approach: Synthesis Reactions ...................................................... 19

2.8. Purification Using Chromatography ..................................................................... 20

2.9. Spectroscopy ......................................................................................................... 21

CHAPTER 3. RESULTS ..................................................................................................................... 25

3.1. Molecular Dynamic Simulation ............................................................................. 25

3.2. FRET Measurements ............................................................................................. 29

CHAPTER 4. DISCUSSION ............................................................................................................... 36

4.1. Efficiency of Energy Transfer ................................................................................ 36

4.2. Comparison of FRET with Dynamic Simulation..................................................... 36

4.3. Interaction of Conjugated Peptide with Site-Specific Antibody and Polyamines 37

4.4. The Effects of HCM Mutations on Labeled Peptide .............................................. 39

4.5. Conclusions ........................................................................................................... 40

LIST OF REFERENCES ..................................................................................................................... 42

iv

Page 7: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

LIST OF TABLES AND FIGURES

Page

Tables

3.1. Comparison of dissociation constant of HCM mutant peptides with wildtype ............... 35

Figures

1.1. Schematic representation of the structure of myosin II..................................................... 2

1.2. Distribution of HCM mutations across beta-cardiac myosin .............................................. 4

1.3. Representation of computational approach ...................................................................... 6

1.4. Illustration of experimental steps. ...................................................................................... 7

2.1. The structure of FITC conjugated to the N-terminus of peptide ...................................... 15

2.2. Molecular structure of the chelate complex .................................................................... 15

2.3. Molecular structure of human myosin S2 fragment ........................................................ 18

2.4. Schematic representation of peptide-FITC conjugation reaction .................................... 20

2.5. Conjugation reaction of DTPA-cytosine to peptide .......................................................... 20

2.6. Absorbance spectrometer readings of reverse phase chromatography run ................... 23

2.7. Absorbance spectrometer readings of anion exchange chromatography run ................ 24

3.1. Distance measurements of computational simulation of conjugated peptide ................ 26

3.2. Results of molecular dynamic simulations of the conjugated peptide .......................... 28

3.3. Lifetime decay of the labeled peptide .............................................................................. 31

3.4. The effect of binding of site-specific antibody to the conjugated peptide ...................... 32

3.5. Interaction of polyamines with labeled peptide .............................................................. 33

3.6. Dissociation constants (Kd) comparison of wildtype with mutant conjugated peptide. .. 34

4.1. Molecular representation of antibody binding to the labeled peptide .......................... 38

v

Page 8: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

CHAPTER 1

INTRODUCTION

1.1. Muscle Contraction

According to the sliding filament model, muscle contraction is principally the result of

sliding thick and thin filaments along each other (Huxley and Simmons, 1971). The thin filament

is primarily composed of F-actin, which is a right-handed helical structure. F-actin serves as a

site to which troponin complex and tropomyosin bind. Troponin complex is composed of three

subcomponents: troponin T (Tn T), troponin I (Tn I), and troponin C (Tn C). Sequential

conformational changes in troponin complex and tropomyosin play a crucial role in actomyosin

interaction during contractility (Garrett and Grisham, 2007; Root et al., 2006).

Thick filament is primarily composed of myosin, which is a large protein composed of

different fragments. Myosin consists of six peptides including two pairs of light chains and two

heavy chains. The heavy chains are composed of globular heads at N-terminus that are linked to

a long coiled-coil tail. Muscle myosin (Figure 1.1) is composed of heavy meromyosin (HMM)

and light meromyosin (LMM). HMM consists of subfragment 1 (S1), which exhibits ATPase

activity and contains actin-binding domains, and subfragment-2 (S2), which exists in the dimeric

structure through coiled-coil formation.

Release of Ca+2 from sarcoplasmic reticulum triggers a contractile signal that results in a

series of conformational rearrangements and leads to the movement of tropomyosin along the

actin filament. When cytosolic Ca+2 concentration increases highly enough, Ca+2 molecules bind

to the Tn C Ca+2 binding site which induces a conformational change in Tn C, causing the

troponin complex and tropomyosin to undergo a conformational change across actin.

1

Page 9: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 1.1. Schematic representation of the structure of myosin II. Subfragment 1 (S1) is attached to a coiled coil tail at S1/S2 hinge. The rod region contains Subfragment-2 (S2) linked to LMM at S2/LMM hinge region. (Root et al., 2006).

This results in a shift in the position of tropomyosin along the actin filament which allows the

myosin heads to bind to actin.

The sliding pattern of thin and thick filaments primarily involves stepwise movement of

S1 heads along the actin filament. This mechanism is driven by hydrolysis of ATP. When bound

to ADP and Pi, myosin heads are in a relaxed state and dissociated from the actin filament.

Release of the contraction signal causes the myosin heads to extend from the thick filaments

and bind to actin. Interaction of myosin heads with actin leads to the release of Pi from the

myosin heads, causing a conformational change in the myosin heads referred to as power

stroke. At this stage, ADP dissociates and the myosin heads travel along the thin filament,

resulting in the myosin heads to undergo a lower energy conformation. Upon hydrolysis of ATP,

myosin heads dissociate from the actin filament and move back to their high energy

conformation with the heads being oriented closer and nearly perpendicular to the thick

filament and prepared to start another cycle within a few seconds (Garrett and Grisham, 2007).

S1 (Head)

S2

LMM

2

Page 10: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

It is known that contractility is principally the result of actomyosin interaction. However, the

details of serial conformational rearrangements of the cycle are yet to be investigated.

1.2. Structure and Function of Subfragment-2 (S2)

Subfragment-2 coiled-coil structure contains two alpha-helical strands that are twisted

around to form a substring heptad repeat (a-b-c-d-e-f-g). In this seven-residue pattern, the first

(a) and fourth (d) positions contain hydrophobic residues oriented internally, enabling alpha-

helices to coil around one another through hydrophobic interactions. The positions (b, c, e, f, g)

are oriented toward the surface, providing intra-interchain interaction which contributes to

further stabilization of the S2 fragment. The myosin rod is further stabilized through interaction

between light meromyosin (LMM) regions contributing to a thick filament assembly (Root et al.,

2006).

The functional importance of subfragment-2 has been proven by the presence of HCM

causing mutations at this region (Figure 1.2) (Erdmann et al., 2003; Cammarato et al.,

2011). Some earlier studies proposed interaction of the myosin binding protein C (MyBP-C)

with S2 fragment, along with actin, conducted a regulatory role in actomyosin interaction.

Furthermore, binding of protein C to the rod domain of myosin has been reported to enhance

stability and assembly of the thick filament structure (Flashman et al., 2004; Ackermann and

Kontrogianni-Konstantopoutos, 2011). According to the earlier model, S2 fragment of myosin

acts as a spring to pull on myosin during actomyosin interaction (Huxley and Simmons, 1971). A

recent study showed that MyBP-C played a regulatory role in muscle contraction through

interaction with actin filament much more than with myosin. It has been proposed that, in the

3

Page 11: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

presence of calcium, MyBP-C interacts with actin to facilitate movement of tropomyosin and

hence, provides interaction sites for S1 heads along actin filament (White et al., 2013).

During muscle contraction, actomyosin interaction is facilitated by myosin heads

reaching to thin filament in proper orientation. Several studies have proposed that extension of

S1 is made possible through elastic property of S2 fragment. (Huxley and Simmons, 1971;

Holmes and Geeves, 2000; Root, 2002; Levitsky, 2004). Experimental data obtained in our lab

strongly suggests that S2 fragment could undergo reversible uncoiling under physiological force

(Dunn, 2010). This study aims to characterize the coiled coil formation of N-terminal region of

subfragment-2.

Figure 1.2. Distribution of HCM mutations across beta-cardiac myosin (Richard et al., 2003).Mutations in amino acid residues of myosin heavy chain lead to hypertrophic cardiomyopathy (HCM). Most of the disease-causing mutations are localized in regions with critical functional properties (S1 region). A number of HCM mutations are found in N-terminal region of subfragment-2, indicating the significant role of S2 fragment during muscle contraction.

4

Page 12: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

1.3. Clinical Significance

Hypertrophic cardiomyopathy (HCM), a heart related abnormality, is caused by

mutations in one of the encoding genes for sarcomeric proteins (Crilley et al., 2003; Redwood

et al., 1999). HCM is characterized by hypertrophy (thickening) of the left ventricle of the heart

(Wigle et al., 1995). Clinical symptoms of HCM vary from being asymptomatic, to heart failure

and syncope (Cirino et al., 2011). HCM causes a wide range of symptoms in individuals from

children to the elderly (Barsheshet et al., 2011). Such symptoms can vary from being mild such

as chest pain, dc to more critical symptoms such as cardiac arrest. HCM is characterized as the

leading cause of cardiac arrest in young athletes. Studies show that about 80% of HCM-causing

mutations are associated with MYH7 and MYBPC3 genes, which encode for myosin heavy chain

beta isoform and cardiac myosin binding protein C respectively (Cirino et al., 2011; Erdmann et

al., 2003). Most HCM-causing mutations target regions with significant functional properties.

Mutations in S1 domain affect actomyosin interaction and or ATPase activity of subfragment-1.

A number of disease-causing mutations are found in the region of subfragment-2, indicating

important function of this region during muscle contraction (Root et al., 2006).

1.4. The Purpose of This Study

Mutations across the S2 region of subfragment-2 are highly likely to cause HCM. A

cluster of HCM-causing-mutations is localized in the region of residue 921 to 939 (Figure 1.2).

This study focuses on the region of myosin subfragment-2 susceptible to HCM mutations. The

project uses dynamic simulation (Figure 1.3) along with fluorescence resonance energy transfer

(FRET) (Figure 1.4) to characterize stability of the region of S2 fragment residues 921 to 939.

5

Page 13: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 1.3. Representation of computational approach. The peptide model was conjugated to FITC on one chain and chelate complex at the other. Using energy minimization and conformational search, the lowest energy conformation of the conjugated peptide mode was obtained and utilized for molecular dynamic simulation.

Peptide Model

chelate conjugation

Energy minimization

conformational searching

FITC conjugation

Dynamic simulation at biological condition

6

Page 14: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 1.4. Illustration of experimental steps. The synthetic peptide was labeled with resonance energy transfer probes and purified using column chromatography techniques. Dimerization and coiled coil formation of the conjugated peptide was determined with FRET.

Synthetic peptide

,Labeled with

chelate

purification Anion

exchange

Peptide-Lys-chelate complex

Dimerization FRET Lifetime measurements

FiTC-NH-peptide

Purification RPC

Labeld with FITC

7

Page 15: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

In this study, the peptide of subfragment-2 residues 921 to 939 was synthesized and

covalently attached to fluorescent probes to determine dimer-monomer equilibrium. The

peptide was conjugated to fluorescein isothiocyanate (FITC), the acceptor probe, and the

chelate complex donor probe. The conjugated peptide was purified using fast protein liquid

chromatography (FPLC).

Reverse phase chromatography was utilized to purify FITC-conjugated peptide and ion

exchange chromatography to separate chelate-labeled peptide from unreacted components

(Figure 1.4). Molecular dynamic simulations (Schrodinger Press, 2009) and fluorescent

resonance energy transfer (FRET) (Förster, 1948) were performed to analyze the stability of the

conjugated peptide.

With the use of dynamic simulations, the compatibility of the probes was determined

with the coiled coil structure of the peptide and obtained distance measurements that were

comparable with FRET experiments. Efficiency of energy transfer was used to estimate

separation distance between the probes. Lifetime decay of donor-acceptor pairs was measured

to determine monomer-dimer equilibrium. In our experiments, high dimer concentration

indicated the presence of a stable coiled coil structure. A decrease in dimer concentration

implied a reduced dimerization and hence, an instable coiled coil structure. In a separate set of

experiments, a site specific antibody was added to the conjugated peptide to determine the

effect of antibody binding to the labeled peptide.

A presence of a cluster of negatively charged glutamate residues in the region of

subfragment-2 suggested potential interaction site for polyamines. The stability or instability

caused by the binding of polyamines to the labeled peptide was determined using resonance

8

Page 16: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

energy transfer. It is anticipated that interaction with positively charged polyamines improves

dimerization and enhances the coiled coil structure of peptide.

Two of HCM causing mutations at the N-terminal region of subfragment-2 include

substitution of glutamate 924 to lysine (E924K) and lethal mutation of glutamate deletion at

residue 930 (E930del) (Richard et al., 2003). To analyze the effect of HCM-mutations on the

stability of subfragment-2, peptides containing E924K mutant and E930del were synthesized.

The synthetic mutant peptides were conjugated to resonance energy transfer probes, FITC and

chelate complex. Dynamic simulation and FRET experiments were used to determine the extent

of coiled coil formation. It was hypothesized that HCM mutations cause structural instability in

the N-terminal region of S2 fragment. HCM mutations at S2 region could alter elastic property

of this region, resulting in malfunction of myosin during actomyosin interaction.

9

Page 17: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

CHAPTER 2

MATERIALS AND METHODS

2.1. Fast Protein Liquid Chromatography (FPLC)

Fast protein liquid chromatography (FPLC) is a liquid type chromatography that is

commonly used to separate and purify protein mixtures. FPLC consists of a stationary phase

made of beads or pours, and a mobile phase which contains liquid or buffer that flows over

immobilized phase. In FPLC, separation is achieved as compounds in a mixture interact with

different affinity with the stationary and the mobile phase. An FPLC system is made of several

compartments including; one or two pumps, a computer control system, detection units, and a

fraction collector. FPLC pumps draw the aqueous solution from buffer reservoirs to the

immobilized phase. The column is packed with a type of resin that binds the compound of

interest. The end of the column is connected to UV and conductivity detectors, which enable

UV absorbance and conductivity measurements of the compounds as they are eluted from the

column. Sample fractions are collected using a fraction collector. FPLC is connected to a

computer system with a user interface that enables the user to control the experimental

settings. Additionally, the user is able to create and modify methods, monitor the run while in

process, as well as store and analyze chromatograms (Robyt and White, 1987).

2.2. Reverse Phase Chromatography (RPC)

Reverse phase chromatography (RPC) consists of a nonpolar stationary phase and a

polar mobile phase. Using RPC, compounds are eluted in the order of increasing

hydrophobicity. Consequently, the polar compounds are eluted first and the most nonpolar

10

Page 18: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

compounds are eluted last. This is achieved by setting the gradient from 0% to 100% nonpolar

solvent. Ion-pairing is often used to facilitate binding of compounds to the column. Chemically

synthesized compounds such as trifluoroacetic acid (TFA) interact with the charges on the

biomolecules, increasing their hydrophobicity and thus, improving their binding to the column.

Partitioning of biomolecules could also be improved using long narrow columns. In this way, the

compound mixture interacts with a greater number of theoretical plates as it travels down the

column. This results in a highly resolved partitioning of molecular compounds. High resolution

is also achieved under acidic conditions. Many biomolecules such as nucleic acids and proteins

however, undergo denaturation in acidic environments and consequently, only small

compounds, such as short peptides, can be purified under such conditions. One issue

associated with using RPC involves back pressure caused by the interaction of the sample with

the column. Conducting the run at a low flow rate will minimize pressure. One should consider

however, that too slow flow rate could also be problematic as it leads to diffusion along the

column and hence, a decrease in resolution (Amersham Pharmacia Biotech AB., 2001).

2.3. Ion-exchange Chromatography (IEX)

Ion exchange chromatography is a technique that is often used to separate molecules

based on their molecular charge. Like other chromatography techniques, IEX consists of a

stationary phase, containing charged compounds that bind oppositely charged molecules, and a

mobile phase, which has charged ions (salt) that alter the binding of the compound to the

column. Media is selected based on the net surface charges of the target compound.

Separation is achieved through reversible interaction of a charged biomolecule with the

11

Page 19: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

oppositely charged column matrix. Biomolecules bind to the matrix as they are loaded onto the

column. Compounds are then eluted from the column by altering the ionic strength of the

mobile phase. Elution is performed with stepwise or gradient separation in which an increase in

salt concentration causes salt ions to compete with bound compounds over binding to the

matrix, resulting in dissociation and elution of compounds from the column (Amersham

Pharmacia Biotech AB., 2001).

2.4. Fluorescence Resonance Energy Transfer (FRET) Overview

FRET is a technique used to measure distances between two points within biological

compounds. Measurements are often performed under physiological conditions. The technique

is capable of detecting measurements near Angstrom resolution. In FRET, a donor fluorophore

is excited and transfers energy to the near-by acceptor fluorescent probe in a distance-

dependent manner. Efficiency of energy transfer relates to distance measurements through the

following equation:

E = Ro6 / (Ro

6 + R6)

where Ro represents critical distance and R represents the actual distance between donor and

acceptor probe (Förster, 1948). The efficiency of energy transfer (E), defines the probability of

energy transfer from donor to acceptor probe while excited donor probe decays to ground

state. Energy transfer depends on several factors. Donor-to-acceptor probe energy transfer

relies on the level of spectral overlap of emission wavelength of donor and excitation

wavelength of acceptor probe. Energy transfer also depends on the orientation of donor and

acceptor probe relative to each other as the electric field of the donor could be polarized. It is

12

Page 20: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

often assumed that donor and acceptor probe rotate randomly relative to one another (Selvin

2002; Selvin and Hearst, 1994).

The efficiency of energy transfer could be measured using several methods, some of

which include: the decline in the lifetime of the donor in the presence of the acceptor probe,

decrease in the intensity of the donor’s emission in the presence of the acceptor, apparent

increase in acceptor fluorescence resulted by energy transfer from the donor probe, and

decline in the quantum yield of the donor in the presence of the acceptor probe (Clegg, 1992;

Schiller, 1975; Root et al., 1999; Selvin, 1995; Selvin, 2002). In cases where the lanthanide-

chelate complex serves as the energy donor probe, efficiency of energy transfer is most

effectively measured using apparent lifetime increase of fluorophore acceptor probe. This is

referred to as sensitized emission lifetime (Clegg, 1992).

Acceptor fluorophore emission decays in nanoseconds and hence, any fluorescence

observed after the initial decay is the result of energy transfer from long-lived (millisecond)

lanthanide donor probe. Furthermore, the lanthanide ion provides sharp emission spectral

peaks containing dark areas, with no donor fluorescence, where acceptor fluorophore emits.

For instance, terbium has no fluorescence at or around 520 nm while fluorescein fluoresces at

this wavelength (Bunzli, 1989). Considering the fact that fluorophore has nanosecond lifetime

decay, the appearance of any emission at or around 520 nm represents only donor acceptor

pairs that are transferring energy. Consequently, the apparent increase in the lifetime of

acceptor probe is proportional to the quenched terbium, which is measured as change in

lifetime. The efficiency of energy transfer is measured by comparison of the sensitized emission

13

Page 21: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

lifetime (Тda) and terbium-only lifetime decay (Тd). Using the following equation was used to

measure the efficiency of energy transfer:

E= 1 – (Тda /Тd)

Hence, E is measured mainly based on donor-acceptor probes that are transferring energy

(Root, 1999; Selvin, 2002).

2.5. Structure of the Chelate Complex and Fluorescein Probe

In this study, a chelate-lanthanide complex was utilized as the donor probe and a

fluorescein as the acceptor probe. Chelate complex is composed of an antenna, a chelate and a

lanthanide ion (Figure 2.2). The antenna, which is an organic chromophore (cytosine), absorbs

photons of light, transferring the energy to lanthanide ion (terbium III). The chelate (DTPA)

component serves several structural features. Chelates contain two reactive groups that enable

attachment of biomolecule to one side and the antenna to the other. In addition, chelates

contain negatively charged carboxylic groups which facilitate tight binding of chelate to

lanthanide ion and, hence, prevent quenching effects caused by water. Lanthanides, such as

terbium, are metallic compounds with long (milliseconds) lifetime that are located on the F-

block of the periodic table. Lanthanides are week energy acceptors and thus, energy is first

absorbed by antenna and transferred to lanthanide ion (Hemmila and Harju, 1994; Selvin,

2002). In FRET experiments, as the lanthanide decays, energy is transmitted to the near-by

fluorescein. The acceptor FITC (Figure 2.1) contains a xanthene ring that provides fluorescent

properties and a reactive thiol group (R-C=S), which serves as an attachment site for

biomolecules.

14

Page 22: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 2.1. The structure of FITC conjugated to the N-terminus of peptide. The lowest energy conformation was obtained using energy minimization and conformational search (Schrodinger MacroModel 8).

Figure 2.2. Molecular structure of the chelate complex. The complex contains an antenna (cytosine), chelate (DTPA carboxylic groups) and lanthanide ion (terbium b III). The chelate complex donor probe was attached to the amine group of Lys (K) residue (939) of the peptide. The lowest energy conformation of the chelate complex was obtained using energy minimization and conformation search.

FITC

N-terminus →

15

Page 23: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Luminescence spectrometer is the instrumentation used to detect resonance energy

transfer. The instrument is equipped with UV excitation source, which is often a nitrogen laser

and sensitive detectors. The instrument is connected to a computer system which enables data

storage and provides interface to control settings and determine experimental parameters. The

instrument also contains gates and filters that enable the user to control and monitor the

intensity of light pulsing through and emitting from the sample. To measure the lifetime decay,

pulses of light pass through the sample causing donor excitation. A photomultiplier tube (PMT)

with proper color filters then detects and amplifies any emission signal caused by decay of

sample molecules (Selvin, 2002; Xiao and Selvin, 1999; Xiao, Selvin, 2001).

2.6. Molecular Dynamic Simulation

The atomic structure of human beta-myosin subfragment-2 (Blankenfeldt et al., 1999)

was obtained from Protein Databank. The N-terminal region of S2 fragment was cut to the

region of interest, residues 921 to 939. Using MacroModel 9.7, minimization, conformational

search and multiple minimization were performed to obtain optimal structure of the peptide.

The lowest energy conformation was used for dynamic simulation of the unlabeled peptide. In

another section, the peptide was computationally labeled with fluorescence resonance energy

probes (Figure 2.3).

Fluorescein isothiocyanate (FITC) was used as the fluorophore accepter probe and

diethylene triamine pentaacetic acid-cytosine terbium (lanthanide DTPA-cytosine) as the

chelate complex donor probe. Probes were selected based on R0, critical value of the acceptor

16

Page 24: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

probe and the overlap of excitation wavelength of the acceptor FITC with emission wavelength

of the terbium donor probe.

Energy minimization followed by conformational search and multiple minimizations

obtained optimal conformation of the peptide. The lowest energy conformation of the labeled

peptide was used to run dynamic simulation. The simulation was performed in biological

condition at 310 K temperature and water solvent.

The force field used was OPLS-AA and all bonds were allowed to “shake” freely.

Simulation was run for 60 nanoseconds. Dynamic simulation was also conducted on peptides

containing HCM mutations. E924K mutant and E930del were the two HCM mutations that were

studied. The E924K and E930del conjugated peptides were modeled separately and obtained a

low energy conformation using minimization and conformational search. Using the same

parameters as in other simulations, a dynamic simulation on mutant peptides was performed.

17

Page 25: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

7A

7B

Figure 2.3. Molecular structure of human myosin S2 fragment. (A) Molecular model of 258 residues of N-terminal region of S2 fragment was adapted from protein data bank (B) The structure was cut to the region susceptible to HCM mutations, residues 921-939. After energy minimization and conformational search the lowest energy conformation was labeled with fluorescent resonance energy probes (FITC and chelate lanthanide complex).

18

Page 26: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

2.7. Experimental Approach: Synthesis Reactions

A synthetic peptide of subfragment-2 residues 921 to 939 (wild-type) was synthesized

and covalently attached to resonance energy probes (selected through dynamic simulation

experiments). Additionally, mutant peptides (E924K and E930del) of subfragment-2 residues

921-939 was synthesized and labeled with resonance energy probes.

Wild-type: NH2-EMNERLEDEEEMNAELTAK-COOH

E924K: NH2-EMNKRLEDEEEMNAELTAK-COOH

E930del: NH2-EMNERLEDEEMNAELTAK-COOH

Peptide was labeled with FITC at N-terminus (Glu 921) at pH 8. Synthetic peptide was

dissolved in 20mM HEPES buffer pH 8 (Figure 2.4). FITC was dissolved in N, N dimethyl

formamide. The reaction was performed in 1:1 molar ratio. In a separate reaction, the peptide

was labeled with chelate complex (Root, 1997). DTPA anhydride (chelate) was attached to

cytosine (antenna molecule) at one of its reactive sites and conjugated to a peptide on the

other (Figure 2.5). Labeling was prepared in dimethylsulfoxide (DMSO) solvent at room

temperature. In the synthesis reaction, DTPA reacted with cytosine in a 1:1 ratio. In a separate

reaction, DTPA-cytosine was then allowed to react with the peptide in a 1:1 molar ratio. The

DTPA-cytosine complex was attached to the primary amine group of Lysine, residue

939. Terbium ion was used as the long-lived lanthanide which was added to the chelate-

labeled peptide in a 1:1 ratio prior to FRET experiment. Fluorescence resonance energy transfer

(FRET) was used to measure the distance between the probes.

19

Page 27: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 2.4. Schematic representation of peptide-FITC conjugation reaction. The N-terminus of the peptide reacts with reactive thiol group of FITC, resulting in formation of a covalent bond between the nitrogen of the amino group of the peptide and carbonyl carbon of FITC.

Figure 2.5. Conjugation reaction of DTPA-cytosine to peptide. DTAP anhydride reacts with cytosine at one reactive group and attaches to peptide at the other reactive site. DTPA serves as a scaffold that connects cytosine (antenna) to the peptide. The three carboxylic groups on the reacted DTPA serve an a chelate that tightly holds the lanthanide ion.

2.8. Purification Using Chromatography

FPLC was used to purify the conjugated peptide from other unreacted compounds. The

Fluorecein-conjugated peptide was separated from the unreacted FITC using reverse phase

chromatography on pepRPC column. Trifluoroacetic acid (TFA) was used as ion paring to

NO

O

O

N H +N

O

O

O

O

O -

NN

O

O -

N

O

O -

O

O -

Cytosine Peptide1. Cytosine

2.Peptide

20

Page 28: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

enhance binding of conjugated peptide to the column. Buffers used for RPC run included 0.05%

TFA in water (buffer A) and 0.05% TFA in acetonitrile (buffer B). The gradient was set to 0-100%

buffer B. Flow rate was set to 0.7 mL per minute. To remove TFA, I further purified the fractions

from the second peak of TFA-RPC run on divilobenzene column with 0-100% gradient buffer B.

In this run, water was used as buffer A and methanol as buffer B. The chelate-labeled peptide

was purified using anion exchange chromatography on Q-sephrose column. Separation was

performed in salt gradient with 10mM imidazole pH 7 in buffer A and B plus 1 M KCl in buffer

B. Flow rate was set to 1 mL per minute using step gradient.

2.9. Spectroscopy

Absorbance spectrometer (Hewlett-Packard) was used to determine concentration of

the conjugated peptide. The peak absorbance at 495 nm indicated presence of FITC in

unreacted form or conjugated to the peptide. Using absorbance readings, two key peaks were

obtained, with the first peak representing fractions that contain unreacted FITC and the second

larger peak corresponding to fluorescein-conjugated peptide. Beer-Lambert law (A=α C l) was

used to calculate the concentration of conjugated peptide (Figure 2.6). In this equation, A,

represents absorbance reading at the excitation wavelength, α is the extension coefficient and l

refers to the distance that light travels through sample which is often 1 cm. The fluorescein

absorbance peak was observed at 495 nm in non-acidic condition with α= 59000 M-1cm-1. The

same equation was used to calculate the concentration of DTPA-cytosine labeled peptide with

absorbance readings at 290 nm (Figure 2.7), α=9300 M-1cm-1, and l =1 cm.

21

Page 29: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Furthermore, a luminescent spectrometer was utilized (Aminco-Bowman Series 2) to

performed FRET experiments and obtain lifetime measurements. In our experiments, the

parameters were set as follows: excitation wavelength 247nm, emission wavelength 545 nm

(emission wavelength of terbium) and band pass of 16 nm. Additionally, the PMT collected

phosphorescence released after 200 μs following flashlamp excitation with the gate width set

to 4.5 ms and flash period of 5 ms. Emission wavelength of 517 nm was used to measure

sensitized emission lifetime (donor-acceptor probe energy transfer). Fluorecein-conjugated

peptide and chelate-labeled peptide were added in 1:1 molar ratio. Lifetime measurements

were performed in 10 mM imidazole, 150 m M KCl concentration, and pH 7 at room

temperature.

Quantum yield was determined using ratios method with fluorescein in 0.1 M NaOH as

the standard (Scott, et al., 1970; Root, 1999). To calculate dissociation constant, lifetime

measurements were obtained at different concentrations of labeled peptide. The sample was

prepared in 1.4 μM total protein concentration and then diluted to 0.2 μM concentration.

Measurements were obtained at total protein concentration of 1.4 μM, 1 μM, 0.6 μM, 0.4 μM,

and 0.2 μM. Kd was calculated using:

Kd = [Dimer]2/[Monomer]x[Monomer]

Using the same experimental assay, site specific antibody (polyclonal antibody) was

added to the peptide. Additionally, several sizes of polyamines were purified and tested on the

labeled peptide. Some tested polyamines include, spermine (27 μM, 85 μM), poly-D-Lysine 17

residues (50 μM, 60 μM), poly-D-Lys 23 residue (27 μM), poly-L-Lysine 9 (2 μM) and 11 (50 μM)

residues (purified by Alysha Joseph and Diana Wang, unpublished data). Lifetime

22

Page 30: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

measurements of the conjugated peptide were obtained in the presence of antibody and, in

separate experiments, different types of polyamines. Lifetime values were used to calculate

dissociation constant (Kd) and monomer-dimer equilibrium.

Figure 2.6. Absorbance spectrometer readings of reverse phase chromatography run. Fractions containing FITC showed an absorbance peak at 440 nm. Fluorecein has excitation peak at 495 nm. The buffer solution (0.05% TFA) caused a shift in absorbance peak. After removal of TFA from the fractions the peak absorbance shifted back to 490 nm. Two well-resolved peaks were obtained, indicating unreacted FITC and fluoresein-conjugated peptide were eluted in well-separated peak fractions.

-1.00E-03

4.90E-02

9.90E-02

1.49E-01

1.99E-01

0 20 40 60

Abso

rban

ce

Fraction Number

Conjugated peptide

Unreacted FITC

23

Page 31: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 2.7. Absorbance spectrometer readings of anion exchange chromatography run. Absorbance readings at 290 nm are plotted versus fraction number. Appearance of peak absorbance at 290 indicated DTPA-cytosin complex formation. Comparison of DTPA-cytosin control run (red) with DTPA-cytosine conjugated peptide run (blue) indicated that chelate-labeled peptide was eluted in the last peak fraction. In IEX chromatography, fractions are eluted from the column based on increasing in negativity. Conjugated peptide is large and the most negatively charged compared to other unreacted components and so interacts strongly with the column.

0

0.1

0.2

0.3

0.4

0.5

0.6

0 10 20 30 40 50

Abso

rban

ce

Fraction Number

Unreacted DTPA unreacted DTPA -cytosine

Unreacted Cytosine

conjugated peptide

24

Page 32: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

CHAPTER 3

RESULTS

3.1. Molecular Dynamic Simulation

Our results obtained from dynamic simulation revealed that resonance energy probes

were compatible with α-helical structure of the peptide. Also, the coiled coil structure of the

conjugated peptide was intact during dynamic simulation (Figure 3.2 B). Distance

measurements between the xanthene ring of fluorescein probe and the lanthanide terbium ion

were about 4 nm.

Additionally, distance measurements between N-terminus and primary amine of

Lysine residue (the attachment site of the probes), were obtained and compared to unlabeled

peptide (Figure 3.1 B). The distance separation was also measured at approximately 4 nm

between resonance energy transfer probes for mutant peptides. Computational

results demonstrated that HCM-mutant labeled peptides, E924K and E930del, were

substantially less stable than wildtype (Figure 3.2 D and F).

25

Page 33: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

12A

12B

Figure 3.1. Distance measurements of computational simulation of conjugated peptide. (A) Separation distance between xanthene group of FiTC acceptor probe and terbium donor probe were about 4 nm. (B) Distance measurements between N-terminus on Glu 921 and amine group of Lys residue were compared in unlabeled peptide (blue) and labeled peptide (red).

0

20

40

60

80

100

120

140

160

28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Mor

e

Freq

uenc

y

Distance Å

0

50

100

150

200

250

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40 42M

ore

Freq

uenc

y

Distance Å

26

Page 34: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

13A 13B

13C 13D

27

Page 35: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

13E 13F

Figure 3.2. Results of molecular dynamic simulations of the conjugated peptide. With the use of energy minimization and conformational search, we obtained the lowest energy conformation for wildtype and mutant conjugated peptides (E924K and E930del). The dynamic simulation was run for 60 ns at 310 k, and 1200 conformations were recorded. The initial conformation of wildtype (A) shows the coiled coil structure of the conjugated peptide. The final conformation of the labeled peptide (B) indicated the resonance energy transfer probes are compatible with the α-helical structure of the peptide. The initial structure of the labeled peptide containing HCM mutations E924K (C) and E930del (E) are presented. Final conformations of dynamic simulation run for E924K (D) and E930del (F) show that HCM-causing mutations disrupt the α-helical structure and destabilize coiled coil formation of the peptide.

28

Page 36: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

3.2. FRET Measurements

Peptide chains were labeled with resonance energy transfer probes. In this case,

dimerization was determined as a measurement of the quenching effect caused by donor-to-

acceptor energy transfer. The terbium chelate probe served as the energy donor to fluorescein

acceptor probe. The extent of dimerization was measured as the change was observed in

lifetime of terbium donor at 545 nm. The lifetime of the terbium chelate donor probe

decreased in the presence of FITC acceptor probe. In the absence of fluorescein acceptor

probe, the lifetime of terbium was measured 1.8 ms. After addition of FITC, the lifetime

decreased to 0.97 ms. Different lifetime measurements were obtained for various isoforms of

the peptide. In some measurements, lifetime decreased from 1.8 ms lanthanide chelate-

conjugated peptide to 0.68 ms upon addition of fluorescein-conjugated peptide. In all

measurements, the quenching effect caused by the acceptor probe was observed indicating

coiled coil formation. The sensitized emission lifetime at 517 nm was simultaneously measured.

At 1.4 μM total conjugated-peptide concentration, lifetime measurements at 545 nm (terbium)

and 517 nm (sensitized emission) closely matched.

Lifetime measurements at 545 nm (Figure 3.3 A) represent the extent of quenching

caused by nearby acceptor probes, and sensitized emission lifetime at 517 nm (Figure 3.3 B)

determines only donor-acceptor pairs that are transferring energy. Close agreement in values

obtained for lifetime measurements at 545 nm and 517 nm indicates that quenching effect

observed at 545 nm is mostly caused by donor-acceptor probe coupling energy transfer. Our

results strongly support that dimerization is favorable at or around 1.4 μM conjugated peptide

concentration.

29

Page 37: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Efficiency of energy transfer (E), was used to determine actual separation distance (R),

between donor-acceptor probes. E was calculated as the ratio of lifetime at 517 nm and 545 nm

using the equation: E= 1 – (Тda /Тd). Based on experimental results E was calculated to be 44%.

Using Föster equation, the R values were calculated for wildtype and mutant (E924K and

E930del) as follows: wildtype 4.2 nm, E924K 4.3 nm, E930del 4.3. Our results demonstrated

that distance measurements from FRET experiments closely matched those obtained from

dynamic simulation (Figure 3.3 C).

14A

14B Lifetime at 517 nm

0

1

2

3

4

5

6

7

8

0 0.001 0.002 0.003 0.004 0.005

Lum

ines

cenc

e

Time (msec)

30

Page 38: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

14C

Figure 3.3. Lifetime decay of the labeled peptide. The conjugated peptide was excited by a flash lamp at 247 nm. (A) The emission lifetime of the donor-labeled peptide was measured at 545 nm (blue curve). The emission lifetime was also measured for donor and acceptor labeled peptide (red curve). )B) The sensitized emission lifetime was measured with labeled peptide with donor and acceptor probe at 517 nm. The lifetimes were obtained using nor-linear single exponential curve fitting. (C) The graph demonstrates total protein versus dimer concentration. Measurements were obtained using lifetimes at 545 nm and 517 nm. Data indicates dimerization is favorable 9≈80%) around 1.4 μM protein concentration.

With the same experimental assay, the binding-effect of site-specific antibody and

polyamines to the labeled peptide were analyzed. The addition of antibody caused an increase

in lifetime at 545 nm (Figure 3.4). Experimental data demonstrated that Kd increased from 0.03

μM (no antibody) to 1.5 μM (antibody added). Furthermore, addition of certain polyamines

caused changes in lifetime decay at 545 nm. Interaction of spermine, poly-L-Lys 9 residues, and

poly-D-Lys 23 residues caused an increase in lifetime at 545 nm, whereas binding of poly-D-Lys

17 residues and poly-L-Lys 11 residues demonstrated a decrease in lifetime relative to the

0

0.2

0.4

0.6

0.8

1

1.2

1.4

0 0.5 1 1.5 2

Dim

er C

once

ntra

tion

(μM

)

Total Protein Concentration (μM)

31

Page 39: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

control run (Figure 3.5). Lifetime measurements on the graph reflect percent increase in

lifetime in presence of polyamines compare to control, where no polyamine was added.

15A

15B

Figure 3.4. The effect of binding of site-specific antibody to the conjugated peptide. (A) Dimerization was determined in the absence and the presence of the antibody. Dissociation constant (Kd) increased from 0.3 μM (without antibody) to 1.5 μM (antibody added). (B) Titration of conjugated peptide with the antibody showed that dimerization decreased as the concentration of the antibody increased.

0

0.2

0.4

0.6

0.8

1

1.2

1.4

0 0.5 1 1.5 2

Dim

er C

once

ntra

tion

(µM

)

Total Peptide Concentration (µM)

conjugated peptide

conjugated peptide + Ab

0

2

4

6

8

0.3 0.4 0.5 0.6 0.7 0.8% A

b co

ncen

trat

ion

Dimer concentration (µM)

32

Page 40: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Compound % Increase p value spermine (27uM) 13.8 ± 0.11 0.037500353 spermine (85uM) 7 ±0.072 0.300707627

poly-D-K 23res (27uM) 6.6±0.06 0.049252153 poly-L-K 9 res (2uM) 6.1±0.058 0.055049735

poly-L-K 11res (50uM) -4.4±0.023 0.003263966 poly-D-K 17res (50uM) -17±0.12 0.022078885 poly-D-K 17res (60uM) −20 ±0.068 0.000869982

Figure 3.5. Interaction of polyamines with labeled peptide. Several polyamines with different concentrations were tested on conjugated peptide. Lifetimes of conjugated peptide with polyamines were compared to lifetime of labeled peptide. The graph indicates the percent increase in lifetime of the labeled peptide in the presence of polyamines. Spermine (27 μM, 85 μM), poly-D-Lys 23 residues (27 μM) poly-L-Lys 9 residues (2 μM), showed a positive % increase in lifetime. Poly-D-Lys 17 residues (50 μM, 60 μM) and poly-L-Lys 11 residues (50 μM) had a negative % increase in lifetime. The Table represents calculated values that are shown on the graph. Results indicate that poly-D-Lys 17 residues and poly-L-Lys 11 residues improved dimerization of labeled peptide.

sper

min

e (2

7uM

)

sper

min

e (8

5uM

)

poly

-D-K

23r

es (2

7uM

)

poly

-L-K

9 re

s (2u

M)

poly

-L-K

11r

es (5

0uM

)

poly

-D-K

17r

es (5

0uM

)

-pol

y-D-

K 17

res

(60u

M)

1 2 3 4 5 6 7

% In

crea

se in

Life

time

33

Page 41: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Finally, the peptide model system was used to experimentally characterize the effect of

two of the HCM-causing mutations (E924K and E930del) on the structural stability of labeled

peptide. Using same experimental parameters, high Kd values for mutant peptides were

obtained (Figure 3.6). Our data showed the extent of dimerization was significantly affected in

HCM mutant peptide. The binding of poly-D-Lys 17 residues to E930del conjugated peptide

caused a decrease in Kd, indicating an increase in the extent dimerization. Using a t-test, The Kd

values of mutant peptide with wild-type were compared (Table 3.1). In another analysis with

the use of t-test, a significant difference was obtained between the Kd values of Edel930 with

poly-D-Lys (1.211 μM) and without poly-D-Lys (6.95 μM) with p-value equal to 0.00522.

Figure 3.6. Dissociation constants (Kd) comparison of wildtype with mutant conjugated peptide. Kd was determined based on lifetime measurements at different concentrations of conjugated peptide. Larger Kd values indicate higher monomer concentration and less dime formation. HCM mutations (E924K and E930del) contributed to a substantial increase in Kd (=6 μM) compared to wildtype (0.006 μM). Binding of poly-D-Lys 17 residues to mutant conjugated peptide E930del caused a decrease in Kd, indicating improvement in dimerization and coiled coil formation.

-2-10123456789

10111213

1 2 3 4 5 6

Kd μM

WT

WTE

del9

30

Edel

930+

poly

-D-K

17r

es

E924

K

WT+

E924

K

Edel

930

34

Page 42: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Table 3.1. Comparison of dissociation constant of HCM mutant peptides with wildtype

Compound Kd Values(μM) p-value WT LT (ms) 0.006 ±0.08

W+Edel930 LT(ms) 1.202081584±0.711 0.034 Edel930+poly-D-K 17res LT (ms) 1.211120228 ±0.8 0.02

E924K LT (ms) 5.322059815 ±1.83 0.0045 WT+E924K LT(ms) 5.379387457 ±2.76 0.058 Edel930 LT(ms) 6.94891463 ±2.14 0.0014

35

Page 43: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

CHAPTER 4

DISCUSSION

4.1. Efficiency of Energy Transfer

In this study, a model peptide system was developed to determine the extent of

dimerization under physiological condition in vitro. With the use of FRET and dynamics

simulations, the distance measurements and analyzed coiled coil formation of the labeled

peptide were calculated. Dimerization was indicated as the donor’s lifetime decreased as the

result of nearby acceptor fluorescein. Efficiency of energy transfer was measured only for

donor-acceptor resonance energy probes that were in close proximity. A decrease in donor's

lifetime at 545 nm and the apparent increase in fluorescein acceptor probe (sensitized emission

lifetime) at 517 nm strongly supported coiled coil formation of conjugate-peptide. Furthermore,

dimerization was observed to be favorable at or around 1.4 μM total peptide concentrations.

The close agreement of lifetime measurements at 545 nm and 517 nm indicated that

dimerization is favorable at this concentration. As the peptide sample was diluted, the lifetime

at 545 nm gradually approached the lifetime of terbium donor. Additionally, nanomolar Kd

values were obtained which highly implicates favorability of coiled coil formation of labeled

peptide. Our FRET experimental data strongly supported the hypothesis that the labeled

wildtype peptide forms a stable coiled coil structure.

4.2. Comparison of FRET with Dynamic Simulation

The fact that selected probes were compatible with the coiled coil structure of the

peptide was demonstrated through strong correlation of dynamic simulation and FRET

36

Page 44: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

experimental results. Consistency in distance measurements between probes obtained from

dynamic simulation and FRET experiments supported the contention that OPLS force field

provides a precise atomic representation of peptide coiled coil formation in biological

condition. Comparison between computational and experimental data presents accurate

information about point of attachment of FITC acceptor probe and terbium DTPA-Cytosine

donor probe (Figure 3.2 A and B). Both Dynamic simulations and FRET experiments indicated

that fluorescence resonance energy probes were highly compatible with the structure of the

peptide. Close agreement of computational and experimental data implies that synthetic

conjugate-peptide provides a precise model system that enables us to analyze stability of the

coiled coil structure of N-terminal region of subfragment-2.

4.3. Interaction of Conjugated Peptide with Site-Specific Antibody and Polyamines

To further our understanding of coiled coil stability of the N-terminal region of

subfragment-2, the monomer-dimer equilibrium upon binding of site-specific antibody and

polyamines to the labeled peptide was determined. FRET experimental results showed that

interaction of site specific antibody with labeled peptide strongly inhibited dimerization,

indicated by an increase in Kd from 0.03 μM to 1.5 μM in the presence of the antibody. Titration

of conjugated peptide with antibody showed that lifetime increased as more antibody was

added. Site-specific antibody is anticipated to inhibit dimerization by binding one antibody

molecule to each chain of the peptide and hence, prevent redimerization (Figure 4.1). Thus,

interaction of the site-specific antibody with the peptide strongly destabilizes dimer formation

and disrupts the coiled coil structure of the region of S2 fragment.

37

Page 45: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Figure 4.1. Molecular representation of antibody binding to the labeled peptide. The site-specific antibody molecules bind to each chain of the conjugated peptide, inhibiting redimerization.

Presence of negatively charged glutamate residues along the N-terminal region of

subfragment-2 strongly suggested potential site of interaction for positively charged

polyamines. Consequently, the extent of dimerization as the consequence of polyamines

interaction with labeled peptide was determined. FRET experimental results demonstrated that

binding of certain polyamines enhanced the stability of the region of subfragment-2. Increase

or decrease in lifetime at 545 nm upon addition of polyamines strongly supported interaction of

these positively charged molecules with the region of S2 fragment.

Antibodybinding

Antibodybinding

38

Page 46: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Polyamines are anticipated to bind along each chain or in-between chains. It is highly

plausible that binding of polyamines along the chains disrupts dimer formation (increase in

lifetime) while binding in between two chains promote dimerization and stabilizes coiled coil

structure of peptide. Polyamines such as poly-D-Lys 17 residues and poly-L-Lys 11 residues are

highly likely to bind in such a unique orientation in which polyamine chains bind in-between or

wrap around the peptide chains, holding them together. Details about how and to what extent

polyamines interact with subfragment-2 will require intense and meticulous analysis. It is

crucial to understand and characterize at the molecular level the structural changes that are

caused by binding of polyamines to N-terminal region of subfragment-2.

4.4. The Effects of HCM Mutations on Labeled Peptide

In the last part of our study, the peptide model system was used to analyze the effects

of HCM-causing mutations on the stability of N-terminal region of S2 fragment. Our

observations revealed that HCM mutations are incompatible with the coiled coil structure of

the peptide. Experimental and computational data demonstrated that HCM-causing mutations

destabilize coiled coil structure of the region of subfragment-2. Increase in lifetime at 545 nm

for two HCM mutations, E924K mutant and E930 deletion, indicated donor-to-acceptor energy

transfer was reduced as a result of decrease in peptide dimer concentration. Increase in Kd

values indicated that dimer formation substantially reduced in mutant peptides as compared to

wildtype. Experimental data strongly supported that HCM mutations affect dimer formation

and, hence, destabilize coiled coil structure of subfragment-2 (Figure 3.6). Furthermore,

dynamic simulations of mutant peptide models demonstrated that HCM mutations strongly

39

Page 47: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

disrupt α-helices of coiled coli structure and cause instability in N-terminal region of S2

fragment (Figure 3.2 D and F). In attempt to counteract the destabilizing effects of HCM

mutations, the effect of poly-D-Lys 17 residue on the E930del peptide was tested. Our data

illustrated that addition of poly-D-Lys 17 residues to E930del peptide, improved coiled coil

formation. Higher Kd values obtained for labeled E930del peptide indicated that binding of poly-

D-Lys 17 residues to mutant peptide lead to increase in dimer formation. Our experimental

results provide new hope that certain molecules such as polyamines could potentially reverse

the adverse effects of disease-causing mutations, and in the near future, save many lives.

4.5. Conclusions

In this study, a model peptide system was developed that enabled us to understand, at

the molecular level, stability/instability of N-terminal region of subfragment-2 susceptible to

HCM-causing mutations. The synthetic peptide of residues 921 to 939 was conjugated to

fluorescent resonance energy probes and used FRET and dynamic simulation to determine

stability of coiled coil structure of the region of S2 fragment. Our experimental assay allowed us

to measure the extent of dimerization as the result of donor-acceptor probe energy transfer.

Strong dimer formation was observed for wildtype conjugated peptide. Furthermore, the

model peptide system was used to study the binding effect of the site specific antibody and

polyamines to the labeled peptide. The antibody binding strongly prevented redimerization and

destabilized the coiled coil structure of the labeled peptide. However, our data demonstrated

that binding of polyamines such as poly-D-Lys 17 residues and poly-L-Lys 11 residues enhanced

dimer formation and improved the stability of the coiled coil structure.

40

Page 48: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

To analyze the effect of HCM mutations on S2 fragment, two of the disease-causing

mutations were studied that are localized in this region. Experimental and computational

observations illustrated that E924K and E930del significantly affect dimerization and destabilize

the coiled coil structure of the S2 fragment. I propose that instability in S2 fragment is likely to

induce structural and functional changes in subfragment-2, which could lead to abnormities in

actomyosin interaction. This study provides a model system that could not only determine

coiled coil formation but also test potential candidate drugs on the stability of the S2 region.

In summary:

1. Model peptide system enables us to analyze the region of subfragment-2 susceptible

to HCM mutations.

2. FRET experiments demonstrated that while binding of antibody to peptide strongly

inhibits dimerization, interaction of certain polyamines (poly-D-Lys 17 residues &

poly-L-Lys 11 resides: are likely to enhance dimer formation.

3. Distance measurements obtained using FRET & dynamic simulations were

consistent.

4. FRET experiments & dynamic simulations highly suggest that E924K & E930del

mutations strongly disrupt dimerization and destabilize coiled coil formation of the

S2 fragment.

5. Model peptide system provides a powerful tool for drug development targeting

disease-causing-mutations along N-terminal region of subfragment-2.

41

Page 49: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

LIST OF REFERENCES

Ackermann, M.A., and Kontrogianni-Konstantopoulos, A. (2011). Myosin binding protein-C:A regulator of actomyosin interaction in striated muscle. Journal of Biomedicine and Biotechnology. doi: 10.1155/2011/636403.

Amersham Pharmacia Biotech AB. (2001). Protein purification handbook. (AB ed.) Uppsala, Sweden: Amersham Pharmacia Biotech AB.

Barsheshet, A., Brenyo, A., Moss, A.J., and Goldenberg, I. (2011). Genetics of sudden cardiac death. Current Cardiology Reports, 13(5), 364-76. doi: 10.1007/s11886-011-0209-y.

Blair, E., Redwood, C., de Jesus Oliveira, M., Moolman-Smook, J. C., Brink, P., Corfield, V. A., Ostman-Smith, I., and Watkins, H. (2002). Mutations of the light meromyosin domain of the b-myosin heavy chain rod in hypertrophic cardiomyopathy. Circulation Research, 90, 263-260. doi: 10.1161/hh0302.104532.

Blankenfeldt, W., Thoma, N.H., Wray, J.S., Gautel, M., and Schlichting, I. (2006). Crystal structures of human cardiac beta-myosin II S2-Delta provide insight into the functional role of the S2 fragment. Proceedings of the National Academy of Sciences of the United States of America. 103 (47) 17713-17717; published ahead of print November 9, 2006, doi:10.1073/pnas.0606741103

Bunzli, J. C., and Choppin, G. R. (1989). Lanthanide probes in life, chemical and earth sciences: Theory and practice. (qsr ed., pp. 219-293). Amsterdam: Elsevier Science Ltd.

Cammarato, A., Li, X., reedy, M. C., Lee, C. F., Lehman, W., and Bernstein, S. I., (2011). Structural basis for myopathic defects engendered by alterations in the myosin rod. Journal of Molecular Biology, 414(4), 477-484.

Cirino, A. L., and Ho, C. (2008) [Updated 2011 May 17]. Familial hypertrophic cardiomyopathy overview. (2008). GeneReviews™, Retrieved from http://www.ncbi.nlm.nih.gov/books/NBK1768Clegg, R.M. (1992) Fluorescence resonance energy transfer and nucleic acids, Methods Enzymol. 211, 353—388.

Crilley, J. G., Boehm, E. A., Rajagopalan, B., Blamire, A. M., Styles, P., McKenna, W. J., Ostman-Smith, I., and Clarke, K. (2003). Hypertrophic cardiomyopathy due to sarcomeric gene mutations is characterized by impaired energy metabolism irrespective of the degree of hypertrophy. Journal of the American College of Cardiology, 41(10), 1776-82.

Dunn, J.W. (2010). Stretching the flexible myosin II subfragment using the novel gravitational force spectrometer. Master’s thesis, University of North Texas, Denton.

Erdmann, J., S. Daehmlow, S. Wischke, M. Senyuva, U. Werner, J. Raible, N. Tanis, S. Dyachenko, M. Hummel, R. Hetzer, and V. Regitz. (2003). Mutation spectrum in a large cohort of

42

Page 50: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

unrelated consecutive patients with hypertrophic cardiomyopathy. Clinical Genetics Journal, 64(4), 339–349.

Flashman, E., Redwood, C., Moolman-Smook, J., and Watkins, H. (2004). Cardiac myosin binding protein C. Circulation Research, 94, 1279-1289. doi:10.1161/01.RES.0000127175.21818.C.

Förster, T. (1948) Zwischenmolekulare energiwanderung und fluoreszence [Intermolecular energy migration and fluorescence], Annals of Physics, 2, 55–75.

Garrett, R.H. and Grisham, C.M. (2007). Biochemistry (3rd edition). Boston: Brooks/Cole.

Hemmila, I., and Harju, R. (1994). Bioanalytical applications of labeling technologies. (pp. 83-119). Turku, Finland : Wallac.

Holmes, K. C., and M. A. Geeves. (2000). The structural basis of muscle contraction. Philosophical Transactions of the Royal Society of London. Series B: Biological Sciences, 355(1396), 419–431.

Huxley, A. F., and Simmons, R. M. (1971). Proposed mechanism of force generation in striated muscle. Nature, 233, 533-538.

Levitsky, D. I. 2004. Actomyosin systems of biological motility. Biochemistry (Moscow.). 69,1177–1189.

Lu, Q., Ye, F., Wei, Z., Wen, Z., and Zhang, M. (2012). Antiparallel coiled-coil-mediated dimerization of myosin x. Proceedings of the National Academy of Sciences, 109(43), 17313-17314.

Redwood, C.S., Moolman-Smook, J.C., and Watkins. H. (1999). Properties of mutant contractile proteins that cause hypertrophic cardiomyopathy. Cardiovascular Research, 44, 20-36.

Richard, P., Charron, P., Carrier, L., Ledeuil, C., Cheav, T., Pichereau, C., Benaiche, A., and Isnard, R. (2004). Hypertrophic cardiomyopathy: Distribution of disease genes, spectrum of mutations, and implications for a molecular diagnosis strategy. Circulation, 109(25), 3258.

Robyt, J. F., and White, B. J. (1987). Protein protocols handbook. Belmont, CA: Brooks/Cole.

Robyt, J. F., and White, B. J. (1987). Biochemical techniques: Theory and practice (pp. 99-103). Monterey: Brooks/Cole.

Root, D. D. (1997) In situ molecular association of dystrophin with actin revealed by sensitized emission immuno-resonance energy transfer. Proceedings of the National Academy of Sciences. 94, 5685–5690.

43

Page 51: Synthetic peptides model instability of cardiac myosin ... › ark: › 67531 › metadc... · Taei, Nasrin. Synthetic peptides model instability of cardiac myosin subfragment-2

Root, D. D. (2002). The dance of actin and myosin: a structural and spectroscopic perspective. Cell Biochemistry and Biophysics. 37, 111–139.

Root , D. D., Shangguan, X., Xu, J., and McAllister, M. A. (1999). Determination of fluorescent probe orientations on biomolecules by conformational searching: Algorithm testing and applications to the atomic model of myosin. Journal of Structural Biology, 127, 22-34.

Root, D. D., Yadavalli, V. K., Forbes, J. G., and Wang, K. (2006). Coiled-coil nanomechanics and uncoiling and unfolding of the superhelix and a-helices of myosin. Biophysical Journal. 90(8), 2852-2866.

Schiller, P. W. (1975) The measurement of intramolecular distances by energy transfer, in Chen, R. F., and Edelhoch, H. (Eds.), BiochemicalFluorescence: Concepts I, pp. 285–303, Dekker, New York.

Schrodinger Press. (2009). MacroModel 9.7 reference manual. New York: Schrodinger Press.

Scott, T. A., Spencer, R. D., Leonard, N. J., and Weber, G. (1970). Emission properties of NADH:Studies of fluorescence lifetimes and quantum efficiencies of NADH, AcPyADH, and simplified synthetic models, Journal of American Chemical Society, 92, 687–695.

Selvin, P. R. (1995) Fluorescence resonance energy transfer, Methods Enzymol. 246, 300–334.

Selvin, P. R. (2002). Principles and biophysical applications of lanthanide- based probes. Annual Review of Biophysics and Biomolecular Structure,31, 275-302. doi: 10.1146/annurev.biophys.31.101101.140927.

Selvin, P. R., and Hearst, J. E. (1994). Luminescence energy transfer using a terbium chelate: Improvements on fluorescence energy transfer. Proceedings of the National Academy of Sciences, 91(21), 10024-10028.

White, H. D., Belknap, B., Harris, S. P. (2013). Activation and inhibition of f-actin and cardiac thin filaments by the n-terminal domains of cardiac myosin binding protein C. Biophysical Journal, 104(s, Supplement 1), 158a–159a.

Wigle, E. D., Rakowski, H., Kimball, B. P., and Williams, W. G. (1995). Hypertrophic cardiomyopathy: Clinical spectrum and treatment. American Heart Association Journals, 92(7), 1680-1692. doi: 10.1161/01.CIR.92.7.1680.

Xiao, M., and Selvin, P. R. (1999). An improved instrument for measuring time-resolved lanthanide emission and resonance energy transfer. Review of Scientific Instruments, 70(10), 3877-3881. doi: 10.1063/1.1150005.

Xiao, M., and Selvin, P. R. (2001). Quantum yields of luminescent lanthanide chelates and far-red dyes measured by resonance energy transfer. Journal of the American Chemical Society, 123(29), 7067-7073. doi: 10.1021/ja0031669.

44