68
Quantum Chemical Studies of Epoxide- Transforming Enzymes Kathrin H. Hopmann Department of Theoretical Chemistry Royal Institute of Technology Stockholm, Sweden, 2007

Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

Quantum Chemical Studies of Epoxide-

Transforming Enzymes

Kathrin H. Hopmann

Department of Theoretical Chemistry

Royal Institute of Technology

Stockholm, Sweden, 2007

Page 2: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

ii

© Kathrin H. Hopmann, 2007 ISBN 978-91-7178-640-1 ISSN 1654-2312 TRITA-BIO-Report 2007:3 Printed by Universitetsservice US-AB, Stockholm, Sweden.

Page 3: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

iii

Abstract

Density functional theory is employed to study the reaction mechanisms of different

epoxide-transforming enzymes. Calculations are based on quantum chemical active

site models, which are build from X-ray crystal structures. The models are used to

study conversion of various epoxides into their corresponding diols or substituted

alcohols. Epoxide-transforming enzymes from three different families are studied. The

human soluble epoxide hydrolase (sEH) belongs to the α/β-hydrolase fold family. sEH

employs a covalent mechanism to hydrolyze various epoxides into vicinal diols. The

Rhodococcus erythrobacter limonene epoxide hydrolase (LEH) constitutes a novel

epoxide hydrolase, which is considered the founding member of a new family of

enzymes. LEH mediates transformation of limone-1,2-epoxide into the corresponding

vicinal diol by employing a general acid/general base-mediated mechanism. The

Agrobacterium radiobacter AD1 haloalcohol dehalogenase HheC is related to the

short-chain dehydrogenase/reductases. HheC is able to convert epoxides using various

nucleophiles such as azide, cyanide, and nitrite. Reaction mechanisms of these three

enzymes are analyzed in depth and the role of different active site residues is studied

through in silico mutations. Steric and electronic factors influencing the

regioselectivity of epoxide opening are identified. The computed energetics help to

explain preferred reaction pathways and experimentally observed regioselectivities.

Our results confirm the usefulness of the employed computational methodology for

investigating enzymatic reactions.

Page 4: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

iv

Acknowledgements

I would like to express my sincere gratitude to my supervisor Dr. Fahmi Himo. Thank

you for giving me the opportunity to work in this field. This work would not have

been possible without your enthusiasm and inspiration. Thanks to Professor Hans

Ågren for accepting me in his group and for providing a stimulating working

environment. I am also grateful that I was given many opportunities to travel and

participate in conferences, winter schools, and collaborations.

A big thanks to all my colleagues at the Department of Theoretical Chemistry in

Stockholm. Special thanks to Emil, Peter, Robin, Polina, and Elias for all the good

times together. Thanks to my dear roommates over the years, Viviane, Luca, Freddy,

Oscar, Katia and Qiong. Thanks to Pawel for always being extremely helpful and for

guiding a biochemist through the jungle of Linux. Also thanks to Laban, Emanuel,

Prakash, Chen, Johanna, Cornel, Bin Gao, Yong Zeng, Liao, Mathias, YanHua,

Yassen, Kai Liu, Na Lin, TT, JJ, Phoenix, Wenhua, Ke Zhao, Pekka, Haakan, and

Luo. All of you contributed to making the department more than a place of work.

Thanks for movie evenings, opera nights, poker games, Chinese New Year parties,

and Friday pubs. Also thanks to our administrative staff, Pia and Lotta, for helping

with many practical issues, and to Elizabeth, Cecilia, and Maria from the library for

providing me with every literature reference I wished for.

Most of this thesis was written during an extended stay in Calabria, Italy, and I would

like to thank Professor Nino Russo for inviting me to his group. Thanks to Monica for

a fruitful collaboration. And thanks also to all the wonderful friends I met in Calabria,

especially Francesca, Fatima, Maria Fernanda, Tanja, and Rosa. Baci to all of you for

making my stay in Italy such a wonderful experience.

Danke auch an meinen lieben Zwilling David. Ich bin froh, dass es Dich gibt.

Finally, a special thanks to Anders. I will never forget you.

Page 5: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

v

Papers included in this thesis

I. Catalytic Mechanism of Limonene Epoxide Hydrolase, a Theoretical Study. Kathrin H. Hopmann, B. Martin Hallberg, Fahmi Himo, J. Am. Chem. Soc. 2005, 127, 14339-347.

II. Theoretical Study of the Full Reaction Mechanism of Human Soluble Epoxide Hydrolase. Kathrin H. Hopmann, Fahmi Himo, Chem. Eur. J. 2006, 12, 6898-909.

III. Insights into the Reaction Mechanism of Soluble Epoxide Hydrolase from Theoretical Active Site Mutants. Kathrin H. Hopmann, Fahmi Himo, J. Phys. Chem. B. 2006, 110, 21299-310.

IV. A Theoretical Study of the Azidolysis and Cyanolysis of Epoxides by Haloalcohol Dehalogenase. Kathrin H. Hopmann, Fahmi Himo, Manuscript

Comments on my contributions to the included papers: I was responsible for all presented calculations, as well as for preparation of all

manuscripts.

Page 6: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

vi

Abbreviations

CIU Cyclohexyl-N´-(Iodophenyl) Urea CPCM Conductor-like Polarizable Continuum Model DFT Density Functional Theory E enantiomeric ratio ee enantiomeric excess EchA Epoxide hydrolase from A.radiobacter AD1 EH Epoxide hydrolase GGA Generalized Gradient Approximation HF Hartree-Fock HheC Haloalcohol dehalogenase C from A.radiobacter AD1 LDA Local Density Approximation LD Limonene-1,2-diol LE Limonene-1,2-epoxide LEH Limonene Epoxide Hydrolase from R.erythropolis DCL14 MAD Mean Absolute Deviation MP2 Second order Møller Plesset perturbation theory MSO (1S,2S)-β-methylstyrene oxide PDB Protein Data Bank QM Quantum Mechanical RSO (R)-styrene oxide SDR Short-chain Dehydrogenase/Reductase SDS Sodium Dodecyl Sulfate sEH soluble Epoxide Hydrolase SSO (S)-styrene oxide StEH Epoxide hydrolase from potato (Solanum tuberosum) TMSCN Trimethylsilyl cyanide TS Transition State ZPV Zero Point Vibrational

Page 7: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

vii

Amino acid symbols

A/Ala C/Cys D/Asp E/Glu F/Phe G/Gly H/His I/Ile K/Lys L/Leu M/Met N/Asn P/Pro Q/Gln R/Arg S/Ser T/Thr V/Val W/Trp X/Xaa Y/Tyr

Alanine Cysteine Aspartate Glutamate Phenylalanine Glycine Histidine Isoleucine Lysine Leucine Methionine Asparagine Proline Glutamine Arginine Serine Threonine Valine Tryptophan Any residue Tyrosine

Page 8: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

viii

Contents

Acknowledgements .......................................................................................List of papers ...............................................................................................Abbreviations ...............................................................................................Symbols for Amino Acids .............................................................................. 1 Introduction ........................................................................................... 2 Computational chemistry ........................................................................ 2.1 Density functional theory .................................................................... 2.2 B3LYP ............................................................................................. 2.2.1 Accuracy of B3LYP .................................................................. 2.2.2 Sources of error: self-interaction and near-degeneracy ................... 3 Modelling enzyme active sites ................................................................... 3.1 Modelling of active site residues .......................................................... 3.2 Modelling of substrates ....................................................................... 3.3 Computational strategy ....................................................................... 3.3.1 Geometry optimizations ............................................................. 3.3.2 Modelling of the surroundings .................................................... 3.3.3 Final energies .......................................................................... 3.4 Comparison with experiment ............................................................... 4 Epoxide chemistry ................................................................................. 4.1 Regioselectivity of epoxide opening ...................................................... 4.2 Enantioselectivity of epoxide conversion ............................................... 4.3 Non-enzymatic transformation of epoxides ............................................ 4.3.1 Hydrolysis ............................................................................... 4.3.2 Azidolysis ............................................................................... 4.3.3 Cyanolysis ............................................................................... 4.3.4 Haloalcohol formation ............................................................... 4.3.5 Conclusions on non-enzymatic epoxide transformation ................... 4.4 Enzymatic transformation of epoxides ................................................... 4.4.1 The soluble epoxide hydrolases ................................................... 4.4.2 Limonene epoxide hydrolase ....................................................... 4.4.3 The haloalcohol dehalogenases .................................................... 4.4.4 Conclusions on enzymatic epoxide transformation ..........................

iv v vi vii 1 3 3 6 6 8 10 10 11 12 12 12 13 14 15 15 16 17 17 17 18 19 19 19 20 21 21 22

Page 9: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

ix

5 Quantum chemical studies of epoxide-transforming enzymes ...................... 5.1 Limonene epoxide hydrolase (Paper I) .................................................. 5.1.1 Elucidating the mechanism of LEH ............................................. 5.1.2 Analysis of the regioselectivity of LEH ........................................ 5.1.2.1 Regioselectivity of 1-methylcyclohexene oxide hydrolysis .. 5.1.2.2 Regioselectivity of limonene-1,2-epoxide hydrolysis .......... 5.2 The soluble epoxide hydrolase (Papers II and III) .................................... 5.2.1 The reaction mechanism of sEH .................................................. 5.2.2 Tyrosine mutants of sEH ........................................................... 5.2.3 Studies on the regioselectivity of sEH .......................................... 5.3 The haloalcohol dehalogenase HheC (Paper IV) ..................................... 5.3.1 The epoxide-opening mechanism of HheC .................................... 5.3.2 Regioselectivity of wild type HheC ............................................. 5.3.3 Regioselectivity of HheC mutants ............................................... 6 Conclusions ............................................................................................ 6.1 Mechanistic strategies of epoxide-transforming enzymes .......................... 6.2 Regioselectivities of epoxide-transforming enzymes ................................ 6.3 Final Conclusions .............................................................................. References .............................................................................................. Papers I – IV ...........................................................................................

23 24 24 25 29 30 31 31 36 38 40 40 43 45 51 51 52 53 55 59

Page 10: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

x

Page 11: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

1

Chapter 1

Introduction

Computational chemistry has grown into a versatile tool for exploring enzymatic

catalysis. Mechanistic proposals can be analyzed in depth, and optimization of

intermediate structures and transition states enables detailed characterization of the

reaction pathways employed by enzymes. Earlier limitations of quantum chemistry,

only allowing accurate calculations of small systems, have been surpassed. Evolution

of quantum mechanical methods and improvement of computational performance now

allow treatment of increasingly larger models and complex systems at a relatively high

level of theory. Especially the Density Functional Theory (DFT) method has proven to

be superior in treatment of enzymatic systems. DFT, and in particular the B3LYP

functional, exhibits high accuracy in regard to structure optimizations and energy

related properties, in many cases comparable to highest-quality wave-function-based

methods. At the same time, relatively large systems can be treated at a reasonable

speed. With DFT, convenient treatment of quantum chemical models of up to 150

atoms is now possible. Although this is quite large for a quantum chemical model, at

first sight it still appears to be a rather crude approximation to an enzyme containing

thousands of atoms. However, enzymatic reactions usually take place within a small

area of the enzyme only, the so-called active site. Here, the chemical reaction will be

mediated by a limited number of functional groups. With models of the above size, it

is usually possible to include the catalytic residues as well as other groups important

for stabilization and binding of intermediate structures, thus arriving at a reasonable

approximation to the enzyme active site.

In this thesis, B3LYP has been used to study the reaction mechanism of several

epoxide-transforming enzymes. Conversion of epoxides is a useful route in organic

chemistry to arrive at various chiral compounds. However, epoxide transformation in

Page 12: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

2

solution often suffers from drawbacks in regard to selectivity and reaction conditions.

Epoxide-converting enzymes are of great aid here, as they can act as biocatalysts for

selective formation of desired products under ambient reaction conditions. Detailed

understanding of the reaction mechanism and selectivities of these enzymes is not

only of general scientific interest, but can also help to improve reaction conditions,

indicate beneficial mutational modifications and aid in design of specific inhibitors.

In the following, a brief outline will be given of the theoretical approach employed in

this thesis (Chapter 2). The protocol for quantum chemical modelling of enzyme

active sites is discussed in Chapter 3. A presentation of enzymatic and non-enzymatic

epoxide conversion is given in Chapter 4. The results of our theoretical investigations

of three different epoxide-transforming enzymes are presented in Chapter 5. For each

enzyme, the mechanism of action is studied and the regioselectivity of epoxide

opening is analyzed. Finally, in Chapter 6, the results are summarized. The papers, in

which our work was published, are attached at the end of this thesis.

Page 13: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

3

Chapter 2

Computational chemistry

All calculations presented in this thesis have been performed using the density

functional theory (DFT) approach, in particular the hybrid functional B3LYP. A short

introduction of DFT and B3LYP is given below.a

2.1 Density functional theory Conventional quantum chemical methods are based on obtaining the wave function

for a given molecule. Once the wave function is known, all properties of the molecule

can be obtained. However, since the wave function depends on 3 spatial and 1 spin

variables for each electron, a total of 4N variables enter the system. This becomes an

unmanageable approach for models with many atoms, such as those studied in this

thesis. A different approach to wave-function based methods is to use the electron

density as variable. The density only depends on three spatial variables, independently

of the number of electrons. However, it is not straightforward to see that a density-

based approach can give us a similar amount of information as a wave-function-based

approach. To begin with, it is not even clear that there is a unique relationship

between the density of a system and its properties. In 1964, Hohenberg and Kohn

published a paper, in which they present a functional for the ground state energy of an

electron gas.1,2 The variable of the functional is the density, ρ(r). In their paper,

Hohenberg and Kohn prove that the ground state density of a system uniquely defines

all molecular properties. It is also shown that the density-dependent functional obeys

the variational principle, which states that any calculated energy is always higher than

or equal to the true ground state energy.2

aFor a more comprehensive discussion of quantum chemical methods and DFT in particular, please refer to Koch and Holthausen1 or Jensen4.

Page 14: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

4

In the density functional theory approach, the electronic energy of the ground state of

a molecule can be divided into different terms, each with the density ρ as the input

variable1:

(2.1) Etot[ρ] = T[ρ] + Eee[ρ] + ENe[ρ]

where T is the kinetic energy, Eee is the electron-electron repulsion, and ENe is the

nuclear-electron attraction. The later term is often referred to as the external potential

and is system dependent. However, the first two parts are universally valid and can be

grouped together into the density functional F[ρ]:

(2.2) Etot[ρ] = F[ρ] + ENe[ρ]

If F[ρ] was known, one would operate with exact DFT. However, exact determination

of F[ρ] is not straightforward. As Hohenberg and Kohn stated:

“If F[ρ] were a known and sufficiently simple function of n, the problem of determining the ground-state energy and density in a given external potential would be rather easy since it requires merely the minimization of a functional of the three-dimensional density function. The major part of the complexities of the many-electron problems are associated with the determination of the universal functional F[ρ].” 2, b

Early attempts to approximate F[ρ] showed rather bad results. Kohn and Sham

realized that the failure of certain density functionals in large part is due to the way the

kinetic energy of the electrons is calculated.1 They developed instead an orbital-based

scheme in which the total kinetic energy (T) is divided into two parts, the kinetic

energy (TS) of a non-interacting system of N electrons (with the same density as the

real interacting system) and the residual part, TC3:

(2.3) T[ρ] = TS[ρ] + TC[ρ] With this approach, the F[ρ] functional can be written as: bIn the original notation by Hohenberg and Kohn, the density was referred to as n. This has been replaced with ρ here, for sake of clarity.

Page 15: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

5

(2.4) F[ρ] = TS[ρ] + J[ρ] + EXC[ρ]

where J is the classical Coulomb interaction and EXC is the exchange-correlation

energy defined as :

(2.5) EXC [ρ] = (T[ρ] - TS[ρ]) + (Eee[ρ]- J[ρ]) = TC[ρ] + Encl[ρ]

where T, Ts, and J are as defined above and Encl is the non-classical part containing

correlation and exchange. The total energy can now be written as:

(2.6) Etot = ET + EJ + EV + EXC

where ET is the kinetic energy of the non-interacting system, EJ is the classical

Coulomb repulsion, EV the nuclear-electron energy, and EXC the exchange-correlation

energy as defined above. It should be noted that all contributions to (2.6) can be

calculated explicitly, except the exchange-correlation energy. The exchange-

correlation functional EXC[ρ] is unknown and can currently only be approximated.

One approach to calculate the exchange-correlation energy involves the Local Density

Approximation (LDA), where it is assumed that the density only varies slowly and

locally can be treated as a uniform gas.4 A popular correlation functional developed

using the uniform electron gas model is the correlation functional VWN introduced by

Vosko, Wilk, and Nusair.5 However, the error in exchange energy using the LDA

scheme is in general about 10%, making this approach less promising.4 The assumed

uniform electron distribution works well for certain systems, but is not useful for most

molecules, where the electron distribution is far from uniform. An improvement to

LDA was the introduction of gradient corrected methods (GGA), which consider not

only the density in a given point, but also the derivative of the density. Becke

introduced a gradient-corrected functional for the exchange energy referred to as

B88.6 A popular gradient-corrected functional for the correlation energy has been

introduced by Lee,Yang, and Parr, which is referred to as LYP.7 The combination of

B88 and LYP results in the widely used BLYP functional.4

Page 16: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

6

2.2 B3LYP

Becke argued that further improvement to the GGA scheme could be achieved if a

certain amount of Hartree-Fock exchange also is included in the functional.8 This

leads to the so-called hybrid functionals. One hybrid functional that incorporates B88,

LYP, VWN and exact exchange is the hybrid functional B3LYP, which is defined as:

(2.7) VWNC

LYPC

B88X

HFX

LDAX

B3LYPXC c)E(1cEbEaEa)E(1E −++++−=

The coefficients a = 0.20, b = 0.72 and c = 0.81 were taken from the B3PW91 hybrid

functional (having PW91 instead of LYP).1,8 The values of the coefficients were

determined empirically for B3PW91 by a linear least-squares fit to 116 experimentally

determined energies.8

2.2.1 Accuracy of B3LYP

Evaluation of functionals is usually done by comparing to experimentally determined

geometries and energies. The accuracy of B3LYP in regard to structural parameters

has been evaluated on 53 molecules from the G2 test set.9 The set includes 71 bond

lengths, 26 bond angles and 2 dihedral angles. Calculations at the B3LYP/6-31G(d)

level of theory yielded average absolute errors of 0.013 Å for bond lengths, 0.62º for

angles, and 0.35º for dihedral angles.9 The error in bond lengths and angles was only

slightly reduced if a larger basis set was used, i.e. for calculations at the B3LYP/6-

311+G(3df,2p) level of theory, the average absolute errors were 0.008 Å for bond

lengths and 0.61º for angles. For dihedral angles, the error was increased to 3.66º with

the large basis set, but this result should be taken with caution, as only two dihedral

angles are present in the test set.9 Overall, it can be concluded that the accuracy of

B3LYP in regard to geometrical parameters is highly satisfactory.

The accuracy of B3LYP in determining different types of energies has also been

tested. For example, atomization energies were calculated for 41 molecules, 26

diatomics from the G2 test set plus additional 15 other molecules, yielding an average

error of 2.2 kcal/mol at the B3LYP/6-311+G(3df,2p) level (with the 6-31G(d) basis

set, an error of 5.18 kcal/mol is observed).9 Atomization energies calculated with

Page 17: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

7

Table 2.1 Mean Absolute Deviation of B3LYP-energies calculated on the G3/05 set.11 Energies (# of energies) MAD (kcal/mol) Enthalpies of formation (270) 4.63 Non-hydrogens (79) 5.77 Hydrocarbons (38) 5.40 Substituted hydrocarbons (100) 4.52 Inorganic hydrides (19) 2.16 Radicals (34) 2.87 Ionization energies (105) 3.83 Electron affinities (63) 2.99 Proton affinities (10) 1.39 Hydrogen-bond strength (6) 1.19 All (454) 4.11

B3LYP/6-311+G(3df,2p) for the entire G2 set (55 molecules) resulted in the same

average error, 2.2 kcal/mol regardless if geometries were optimized with 6-31G(d) or

with the 6-311+G(3df,2p) basis set.10 These results indicate that B3LYP is able to

approach so-called chemical accuracy, which is considered to lie within 1-2 kcal/mol

of error. However, the above test set of molecules is relatively small. A much bigger

evaluation has recently been done by Curtis et al., who tested a number of density

functionals on the G3/05 test set.11 This set includes 454 energies, all of which have

experimental uncertainties less than ±1 kcal/mol. The computed results are based on

single-point B3LYP/6-311+G(3df,2p) energies at MP2/6-31G(d) geometries with

scaled (0.89) HF/6-31G(d) zero-point energies. Table 2.1 shows that B3LYP achieves

chemical accuracy for certain energies in the G3/05 set such as proton affinities, while

it performs less well for enthalpies of formation of for example hydrocarbons.

For our approach it is of particular interest to establish what error B3LYP usually

displays in calculations of reaction barriers. In general, DFT is often quoted for

underestimating barriers. For example for the SN2 attack of chlorine or bromide at a

saturated carbon atom, the barriers are underestimated with B3LYP by at least 3-5

kcal/mol.12 An investigation of 12 small organic reactions with various DFT

Page 18: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

8

functionals also showed a tendency to underestimate reaction barriers.13 For 9

pericyclic reactions, the enthalpies of activation calculated at the B3LYP/6-31G level

had a mean absolute deviation from predictedc ΔH‡ values of 1.7 kcal/mol.14 For

enzymatic reactions, Siegbahn concludes that using B3LYP, the error in relative

energies in general is about 3 kcal/mol for molecules containing first- and second-row

atoms.15 For systems involving transition metals, the error is up to 5 kcal/mol.15 Some

sources of errors are discussed below.

2.2.2 Sources of error: self-interaction and near-degeneracy

In Hartree-Fock theory, an artificial term is observed, which corresponds to the

Coulomb repulsion of an electron with itself. This is obviously non-physical and is

referred to as the self-interaction error. Fortunately, the magnitude of this term

corresponds exactly to the size of the exchange term, and as these enter the equation

with opposite signs, the self-interaction is cancelled.1 In exact DFT, the exact EXC

corrects for the error introduced by EJ. However, in approximate DFT functionals, a

certain amount of self-interaction error remains.1 One consequence of the self-

interaction error can be illustrated with the dissociation of H2+. If the H-H bond is

stretched, the system converges to a delocalized H0.5+ + H0.5+ state, instead of the

correct localized H + H+ situation. The artificial stabilization of the former state

occurs, because the delocalized electron exhibits less self-interaction. The error in

dissociation becomes as large as 55 kcal/mol.16 This effect can also be assumed to be

observed in transition states of certain reactions. In the reactants, the separate

fragments have an integer number of electrons, but as the reaction proceeds to the

transition state, some electron transfer occurs, resulting in the formation of systems

with a fractional number of electrons.17 The self-interaction error artificially stabilizes

the delocalized transition state, leading to an underestimation of the barrier. For

example, with B3LYP the barrier height for the hydrogen abstraction reaction H2 + H

→ H + H2 is calculated to be 4.1 kcal/mol, which is 5.6 kcal/mol lower than the

experimental value.18 If the functional is corrected for the self-interaction, a barrier of

11.1 kcal/mol is obtained, which is only 1.4 kcal/mol in error.18 Also for a set of nine

cThe predicted ΔH‡ values at 0 Kelvin were derived from experimental ΔH‡ by subtracting theoretically derived thermal corrections.

Page 19: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

9

reactions including proton transfer, hydrogen abstraction and radical dissociation

reactions, the average absolute deviation from experimental and accurate ab initio

energies was found to be as much as 12-14 kcal/mol for different density

functionals.19 Including self-interaction corrections reduced the average absolute

deviation to 3-5 kcal/mol.19 It can be noted that reaction energies in general are

unaffected by the self-interaction error and no improvement is observed upon

inclusion of self-interaction corrections.19

Siegbahn has pointed out that an underestimation of the barrier due to the self-

interaction error usually only is observed for reactions involving proton or hydrogen

transfer.15 Other reactions often exhibit an overestimation of barriers, because the

dominating error here is the near-degeneracy effect, also referred to as non-dynamical

correlation error.15 Non-dynamical correlation is a long-range effect that for example

becomes of importance if a single bond such as in H2 is stretched to give two non-

interacting atoms, each with one electron.1 Two determinants are required to describe

the dissociation of H2 correctly, each with equal weight. If only one determinant is

used, a wrong potential energy curve emerges, at least if a restricted approach is used.1

The energetic problem can often be solved by using an unrestricted approach.1,15

Page 20: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

10

Chapter 3

Modelling enzyme active sites

Building of the model is a critical step in quantum chemical calculations. For

modelling of enzymatic mechanisms, careful considerations about what to include in

the model are necessary. Often a number of models are built to asses the effect and

importance of various groups or residues on the investigated reaction mechanism.

3.1 Modelling of active site residues

Usually, the protein X-ray crystal structure serves as a basis for the model. The

structure is analyzed and the active site residues that are assumed to be involved in

substrate conversion and stabilization are extracted from the PDB file. The

identification of important residues is usually facilitated if mutational studies have

been performed on the enzyme in question. In certain cases, there is no hint about the

identity of the catalytic residues and different possibilities have to be tested. The

residues that were extracted from the PDB file are usually truncated to reduce the

model size. Often only parts of the side-chains are used in the quantum model (Figure

3.1). The truncated residues are normally fixed at the atom of truncation to preserve

the spatial arrangement of residues. In general, only the functional groups of the side

chains are included, extended by 1-2 carbons. For example aspartate and glutamate are

often modelled as acetic acid, tyrosine and phenylalanine as phenol and phenyl,

respectively, arginine as N-methyl-guanidine, serine as ethanol, and histidine as

imidazole. In certain cases, the whole side chain and sometimes even the backbone

part is included, if it is evaluated that increased flexibility of the residue in question is

crucial for the mechanism. In some cases the backbone part is involved in explicit

interactions with the catalytic residues or the substrate, and it is thus included in the

model.

Page 21: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

11

Figure 3.1 Building a quantum chemical active site model. Important active site residues are extracted from the X-ray crystal structure and truncated. Points of truncation (denoted with asterisks) are fixed in calculations. An appropriate substrate is manually modelled in place of an inhibitor (or substrate analogue/product).

*

**

**

Tyr53

Arg99

Substrate

Asp101

Asp132Asn55

Tyr53

Arg99

Asp101

Asp132

Asn55

Inhibitor

Active site

WatWat

X-ray crystal structure Active site residues Final QM model

Current protein crystal structures mainly exhibit resolutions of 2-3 Å, which does not

allow assignment of hydrogen atoms. These are therefore added manually to the

extracted residues. The low resolution also does not allow identification of the

protonation state of the residue side chains. In some cases, the pKa value of the

residue in question has been established experimentally and can be used as a guideline

for its protonation state. Otherwise, models with different protonation states might be

build to asses the effect on the reaction mechanism.

3.2 Modelling of substrates Some enzymatic structures are determined in presence of products, substrate

analogues or inhibitors, which serve as a good starting point for manual modelling of

a substrate. If this is not the case, a suitable substrate is modelled from scratch and

added to the model. The substrate is chosen based on the known substrate specificity

of the enzyme. The substrate might be truncated if it is concluded that certain parts are

not of importance for the reaction. In some cases, different substrate orientations are

tested to establish if the position is critical for the mechanism or energetics of the

reaction.

Page 22: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

12

3.3 Computational strategy The chemical model is now used to investigate the reaction mechanism with the

chosen substrate. The computational strategy comprises calculations done in separate

steps. First, geometries are optimized. For minima, this is usually straight forward. For

transition state structures, a linear transit scan along a chosen reaction coordinate

might be employed to locate the transition state structures. The optimized geometries

corresponding to the reaction pathway of interest are then subjected to different single

point calculations to calculate solvation effects and big basis set corrections to the

energy. Finally, frequency calculations are performed, which are used to obtain zero-

point vibrational (ZPV) effects. Details of the computational protocol employed in this

thesis are given below.

3.3.1 Geometry optimizations

Calculations are performed using B3LYP as implemented in the Gaussian03 program

package.20 Geometry optimizations are done using the split-valence double-ζ basis set

6-31G(d,p). Frequency calculations are performed on the optimized geometries at the

same level of theory to confirm the nature of the stationary points. Minima should

only exhibit positive eigenfrequencies, while transition states exhibit one negative

(imaginary) eigenfrequency. Freezing some atoms to their crystallographically

observed positions introduces a few small negative eigenvalues for the optimized

structures, but this are usually only on the order of -10 to -20 cm-1, and do not affect

the results significantly.

3.3.2 Modelling of the surroundings

The conductor-like polarizable continuum model21 (CPCM) is used to calculate the

solvation effect that the surrounding protein would have on the energetics. Single

point calculations are performed on the optimized structures at the same level as

optimizations. In the CPCM model, the solvent is represented by a homogenous

dielectric medium surrounding a cavity containing the solute. The dielectric constant

of the medium is set to ε = 4, which is the standard value used in modelling protein

surroundings.22 This value corresponds roughly to a mixture of a protein medium

(dielectric constant of 2-3) and water (dielectric constant of 80). Estimating the effects

Page 23: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

13

of the surrounding as single point calculations includes the implicit assumption that

the gas-phase-optimized geometries will not differ substantially from the geometries

that would be optimized in presence of solvent. This assumption is usually valid,

except in certain cases, for example for geometries with a high degree of charge

separation, where the solution structure might differ substantially from the gas-phase-

optimized geometry.

Another implicit approximation in the CPCM approach is the assumption that the

heterogeneous protein environment can be modelled with a homogenous medium.

This approach might seem crude but has nonetheless shown to be a reasonably

accurate method in determining enzymatic reaction mechanisms.22 The explicit effect

of various surrounding groups can of course not be assessed with this method.

However, care is usually taken to ensure that explicit interactions between the

catalytic groups and the surrounding protein are included in the quantum chemical

model.

The solvation corrections calculated for the systems considered in this thesis are

around 1-4 kcal/mol. In some cases, if a much bigger effect is observed, this can

indicate that the model size is insufficient. In general, as the size of quantum chemical

models tends to increase, the calculated solvation corrections on the reaction

energetics become less, since most of the solvation effects are already present in the

quantum model itself.

3.3.3 Final energies

Normally, single-point big basis set calculations are performed on the optimized

geometries at the B3LYP/6-311+G(2d,2p) level to obtain more accurate energies. For

the systems considered here, the difference in small basis set (6-31G(d,p)) energies

and the big basis set calculations are typically less than 2 kcal/mol. Zero-point

vibrational (ZPV) effects, which are obtained from frequency calculations, are added

to the final energies. These effects are often less than 1 kcal/mol, but can in some

cases be 2-3 kcal/mol. If very large models are employed, the computation of ZPV

effects can become too demanding and the effect is instead estimated from a smaller

model describing the same reaction. The final energies are composed of the big basis

set energies, corrected for solvation effects and for ZPV effects.

Page 24: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

14

3.4 Comparison with experiment In order to compare theoretically determined energies with experimentally determined

reaction rates, classical transition state theory can be used. The relationship between

the rate constant k of a reaction and the free energy of activation (∆G≠) can be

expressed as:4

(3.1) ⎟⎟

⎜⎜

⎛ ≠−=

TΔGexp Tk B

Rhk

where k is the rate constant (s-1), kB is Boltzmann’s constant (1.38 × 10−23 J/K), T is

the temperature (in Kelvin, 298.15 at room temperature), h is Planck’s constant (6.626

× 10-34 Js), ΔG≠ is Gibbs free energy difference between the reactant and the transition

state, and R is the gas constant (8.314 J/K).

With the above equation, experimentally derived rate constants can be converted into

activation energies and compared to the computationally determined reaction barriers.

It is important to note the exponential relationship between the rate and the barrier in

(3.1). For every 1.4 kcal/mol increase in barrier, the rate changes with a factor of 10.

Since the error in computed barriers often is 2-3 kcal/mol, it is not possible to predict

reaction rates from computed barriers.

It should also be realized that the barriers calculated in this work only correspond to

the enthalpy part (∆H≠) of the free energy of activation (∆G≠ = ∆H≠ - T∆S≠). The

entropic part could not be calculated, due to the freezing scheme employed in

geometry optimizations. The small negative eigenvalues that are generated in this

approach would generate an artificial effect on the entropic contribution to the energy.

It is therefore assumed here that ∆G≠ ≈ ∆H≠. This is in many cases a valid assumption,

since the change in entropy (∆S≠) in going from the reactant to the transition state

often is small.

Page 25: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

15

Chapter 4

Epoxide chemistry

Epoxides are versatile intermediates in organic chemistry. They generally exhibit high

reactivity due to the strain of the three-membered ring and can relatively easily be

converted into various substituted alcohols.23 Regioselectivity is an important issue in

epoxide chemistry, since epoxides are ambident substrates, i.e. they can react at

different carbon centers (Scheme 4.1). Opening of asymmetric epoxides will thus in

many cases lead to multiple products.23 Another issue concerns the enantioselectivity

of epoxide conversion (Scheme 4.1). Epoxides are often obtained in racemic mixtures,

and methods for kinetic resolution and enantioselective reaction with one of the two

epoxide isomers are thus of high interest.

O

CCR

Nu

O

CCR

Nu

O

CCR

α β

Regioselectivity Enantioselectivity

Scheme 4.1 Regioselectivity and enantioselectivity of epoxide conversion.

4.1 Regioselectivity of epoxide opening The regioselectivity of epoxide opening with a given nucleophile depends on various

factors such as the acidity of the medium, the size of the nucleophile and the

substitution pattern on the epoxide. These factors can in general be divided into steric

and electronic effects. Under conditions with neutral or basic pH values, steric factors

usually dominate and the reaction occurs through an SN2 attack of the nucleophile on

the less substituted epoxide carbon23 (attack at the less substituted carbon is

Page 26: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

16

sometimes referred to as anti-Markovnikov-type epoxide opening24). Under acidic

conditions, the reaction occurs either through an SN1 mechanism (with protonation of

the epoxide and ring opening prior to nucleophilic attack) or through an SN2

mechanism (opening and attack occur concertedly). During epoxide opening, a

positive charge evolves at the epoxide carbon (Scheme 4.2), which is better stabilized

at the more substituted carbon, due to electron donating effects. Polarization of the

oxygen-carbon bond through protonation or hydrogen bonding enhances the

preference for the more substituted carbon. Under acidic conditions, electronic effects

can thus reverse the regioselectivity of epoxide opening and lead to attack at the more

substituted carbon.23

O

CCR

AH

Nu

δ+δ-

Scheme 4.2 At the transition state for epoxide-opening, a build-up of positive charge occurs at the oxirane carbon.

4.2 Enantioselectivity of epoxide conversion Epoxides are often obtained as racemates, i.e. equal mixtures of two enantiomers. The

two isomers can in principle be viewed as two different compounds that will exhibit

different properties and functionalities. It is of high interest to develop methods that

can lead to cheap separation of the racemates. The principle of kinetic resolution is

based on finding conditions that will mediate conversion of only one of the

enantiomers, while the other remains untouched and can be isolated in pure form.

Since the reactivities of the epoxide enantiomers with a free nucleophile often are

identical, this is a difficult task. However, the use of chiral catalysts can result in

enantioselective conversion of epoxides. Enzymes are especially useful for this

approach, since they provide a chiral environment and often exhibit large affinity

differences for epoxide enantiomers.

Page 27: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

17

4.3 Non-enzymatic transformation of epoxides In solution, epoxide ring opening can occur with a large variety of nucleophiles, such

as water, azide, cyanide, halides, alcohols, or amines.25 Regioselectivities for

reactions involving these nucleophiles depend on the reaction conditions as well as on

the epoxide. Enantioselectivity is in general poor or absent. One epoxide of interest in

our studies of enzymatic reactions is the phenyl-substituted epoxide styrene oxide

(Scheme 4.3). Different protocols for conversion of epoxides, in particular styrene

oxide, are discussed below.

O

CC*α β

Scheme 4.3 The terminal epoxide styrene oxide. One chiral center is present (denoted with *). The benzylic and terminal carbons are referred to as α and β, respectively.

4.3.1 Hydrolysis

Hydrolysis of epoxides results in the formation of vicinal diols. The reaction can be

catalysed by acid or by base.23 Regioselectivities of attack depend on the conditions,

as described above. Basic and neutral conditions usually lead to attack at the more

accessible carbon, acidic conditions to attack at the more substituted carbon.23

However, for aryl-substituted epoxides, substantial attack at the benzylic position

might occur even under basic conditions. For example, in a 3M KOH solution, styrene

oxide exhibits 51% attack at Cα and 49% attack at Cβ.26 Under acidic conditions,

styrene oxide is exclusively opened at the benzylic carbon (α).26,27

4.3.2 Azidolysis

If nucleophiles other than water/hydroxy are used, the opening of epoxides will lead to

the formation of substituted alcohols. Azidolysis of epoxides under different

conditions has been reported. Epoxides react with NaN3, for example in presence of

acid or metal salts.28 Asymmetric 1,2-epoxides will mainly react at the terminal

Page 28: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

18

carbon (β), but regioselectivity is not complete.28 Aryl substituted epoxides are

different and exhibit preferred (but not exclusive) attack at the benzylic position under

almost all conditions. Regioselectivities of 81-83% for the benzylic position are

reported for the reaction of styrene oxide with NaN3 in presence of metal salts.28 With

diiodosamarium as catalyst, trimethylsilylazide can be used to convert styrene oxide

into the protected azidoalcohol, with 90% attack at the benzylic carbon.29 In aqueous

medium containing NaN3, a preference of 97% for the benzylic carbon of styrene

oxide is observed under both acidic and basic conditions (respective pH values of 4.2

and 9.5).30 Interestingly, high enantiomer-selective azidolysis of various epoxides can

be achieved with a (salen)Cr(III)N3 complex.31 However, using this catalyst, very

poor regioselectivity is obtained for styrene oxide.31

4.3.3 Cyanolysis

Introduction of a cyanide group into a compound is often useful, because the cyanide

can be transformed into various other functional groups such as amines and carboxylic

acids. Epoxides can be converted into cyanoalcohols with for example trimethylsilyl

cyanide, TMSCN. In presence of solid base (CaO or MgO), TMSCN will donate a

cyanide group to the β-carbon of styrene oxide with 93-96% regioselectivity.32 With

TMSCN and lithiumperchlorate, styrene oxide exhibits 92% attack at the terminal (β)

carbon.33 Also diiodosamarium can be used as a catalyst for the TMSCN mediated

reaction, however, regioselectivities for styrene oxide are poor (58-77% attack at

Cα).29 The use of TMSCN is generally associated with two major drawbacks. CN is

an ambident nucleophile, and using TMSCN, both isonitriles (covalent bond to N) and

nitriles (covalent bond to C) are often formed.34,35 Another drawback is that the TMS

group will attach to the alcohol group, generating trimethylsilyl O-protected alcohols,

which need to be cleaved to release the desired product.32,35 Opening of epoxides can

instead be performed with NaCN or KCN. If KCN is used in presence of metals salts,

no isonitriles are formed and preferred attack at the terminal position is observed for

most epoxides.35 However, for aryl substituted epoxides a mixture of products is

observed with 77% to 84% attack at the β-carbon.35 With NH4Cl as acid and KCN,

92% attack at the β-carbon of styrene oxide occurs.35 In presence of β-cyclodextrin,

conversion of various 1,2-epoxides with NaCN leads to complete regioselectivity for

Page 29: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

19

the β-carbon, also for styrene oxide.36 With Ce(OTf)4 as catalyst in water, styrene

oxide will react with NaCN, but the reaction exhibits very poor regioselectivity.37

Addition of sodium dodecyl sulfate (SDS) to the mixture increases the regioselectivity

for the benzylic carbon to 92%.37 Under mildly basic conditions using triethylamine

in THF, acetone cyanohydrin can be used to convert terminal epoxides to the

corresponding cyano alcohols, with exclusive attack at the terminal (β) carbon, also

for styrene oxide.38

4.3.4 Haloalcohol formation

Haloalcohols (also known as halohydrins) can be formed from epoxides with for

example lithium halides such as LiI, LiBr, or LiCl.39 Asymmetric 1,2-epoxides mainly

react at the terminal carbon, while reaction with styrene oxide results in a mixture of

products with the major haloalcohol regioisomer exhibiting addition to the benzylic

carbon.39 In aqueous media containing SDS and Ce(OTf)4 as catalyst, the reaction of

styrene oxide with NaCl or NaBr will lead to halide attack at the benzylic position.37

4.3.5 Conclusions on non-enzymatic epoxide transformation

The above section describes some of the many different ways epoxides can be

transformed in solution. Multiple protocols exist, but they often exhibit important

drawbacks such as low regioselectivity, formation of protected alcohols that need to

be cleaved subsequently, or involvement of harmful compounds. If one was to list the

desired criteria for an epoxide-converting process, this would include mild reaction

conditions, non-toxic catalysts, formation of pure products, high regioselectivity, high

chemoselectivity (i.e. reaction occurs only at the desired site and not with other

functional groups), and preferably also high enantioselectivity. Interestingly, epoxide

conversion using enzymes as biocatalysts seem to fulfil many of the wishes on the

above list.

4.4 Enzymatic transformation of epoxides Enzymes usually react in aqueous solutions around physiological pH values at

ambient temperatures, thus providing mild reaction conditions. However, what makes

them truly attractive as biocatalysts is the high selectivity displayed in many reactions.

Page 30: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

20

This regards regioselectivity, chemoselectivity, and in particular enantioselectivity.

Since enzymes are chiral catalysts, different binding modes and different reaction

rates are often observed for enantiomers of the same compound. This makes enzymes

extremely useful for the kinetic resolution of racemic mixtures of epoxides.40

We have studied epoxide-converting enzymes from three different classes in this

thesis. All three classes exhibit enzymes that display biocatalytic potential. The two

classes of epoxide hydrolases are useful for transforming epoxides into vicinal diols,

while the haloalcohol dehalogenases can be used for formation of substituted alcohols,

including azido- and cyanoalcohols. An overview of the usefulness of different

members of these three enzyme classes in biocatalytic applications will be given here.

4.4.1 The soluble epoxide hydrolases

The soluble epoxide hydrolases (sEHs) catalyze conversion of epoxides into vicinal

diols. Homologues of sEH are found in most organisms, including plants, fungi,

mammals, and insects. The different sEH display different substrate specificities,

regio- and eantioselectivities, i.e. a multitude of epoxide hydrolysis reactions can be

performed. One sEH with biocatalytic potential is the soluble epoxide hydrolase

EchA, which was isolated from the bacterium Agrobacterium radiobacter AD1.41,42

EchA has relatively broad substrate specificity and reacts with for example styrene

oxide, which is converted into the corresponding diol. Attack occurs exclusively at the

terminal carbon (β) of styrene oxide.43 The enantioselectivity of the wild-type EchA

with styrene oxide is low, displaying an enantiomeric ratiod of 17.43 However, protein engineering of EchA resulted in an I219F mutant with an enantiomeric ratio of 91 for

styrene oxide transformation.44

It has been shown that conversion of epoxides in presence of two sEHs with different

regio- and enantioselectivities can result in formation of very pure products in high

yields. In conventional kinetic resolutions, the yield of optically pure product cannot

exceed 50%, however, in this approach a product yield of up to 100% can be dThe enantiomeric ratio is defined as the ratio of the reaction rates for the two enantiomers, E = (kcat/KM)R / (kcat/KM)S (where kcat is the rate constant and KM is the Michaelis–Menten constant, and R/S refers to the enantiomers).

Page 31: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

21

achieved. For example, styrene oxide conversion in simultaneous presence of both the

EchA-I219F mutant and the StEH from potato (Solanum tuberosum) resulted in

enantioconvergent production of (R)-1-phenyl-1,2-ethanediol from a racemic mixture

of styrene oxide, resulting in almost 100% yield with 98% enantiomeric excess

(ee)e.45 This is possible since EchA prefers to convert (R)-styrene oxide, attacking

exclusively the β-carbon, while StEH prefers (S)-styrene oxide, attacking almost only

the α-carbon. In both cases, the product is (R)-1-phenyl-1,2-ethanediol.45 Also the

sEHs from Aspergillus niger and Beauveria sulfurescens has been utilized with the

same purpose, yielding (R)-1-phenyl-1,2-ethanediol with 92% yield and 89% ee.46

4.4.2 Limonene epoxide hydrolase

The limonene epoxide hydrolase (LEH) originates from the bacterium Radiobacter

erythropolis DCL14.47,48 Structural and biochemical studies revealed that it belongs to

a different class than the sEHs introduced above. LEH is therefore considered to be

the founding member of a new protein class, with only few putative members so far.47

The biocatalytic potential of LEH is in principle low, since LEH has narrow substrate

specificity and accepts only few substrates, these include limonene-1,2-epoxide, 1-

methyl cyclohexene oxide and indene oxide.48 However, LEH displays high

enantioselectivity and regioselectivity in conversion of the four stereoisomers of the

natural substrate, limonene-1,2-epoxide (LE). For example, conversion of the

diastereomeric mixture of (1S,2R,4R)-LE and (1R,2S,4R)-LE lead to enatioconvergent

formation of (1S,2S,4R)-limonene-1,2-diol.48 Conversion of (1S,2R,4S)-LE and

(1R,2S,4S)-LE lead to enatioconvergent formation of (1R,2R,4S)-limonene-1,2-diol.

The carbon skeleton of limonene is similar to several important flavour and medicinal

compounds, making formation of optically pure products of interest.

4.4.3 The haloalcohol dehalogenases

Haloalcohol dehalogenases catalyze the reversible dehalogenation of vicinal halo-

alcohols to form epoxides. They have exclusively been identified in microbes,

including Corynebacterium sp.,49 Arthrobacter sp.,50,51 Agrobacterium sp.,51,52 and

eEnantiomeric excess, ee, is defined as: ee (%) = ([R] - [S])/([R] +[S]) * 100

Page 32: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

22

Mycobacterium sp.51

. In addition to dehalogenation, haloalcohol dehalogenases also

catalyze the irreversible ring opening of various epoxides with the nucleophiles CN¯,

N3¯, and NO2¯.53,54,55,56,57 One haloalcohol dehalogenase that has been studied

intensively is HheC, which originates from the bacterium A. radiobacter AD1.51,51

HheC displays high enantioselectivity for both aromatic and non-aromatic halo-

hydrins. The native enzyme displays an E value of 150 for conversion of p-nitro-2-

bromo-1-phenylethanol.58 For a W249F-mutant of HheC, the E value of this reaction

is 900.58 W249F-HheC has been used successfully in kinetic resolution of racemic 4-

chloro-3-hydroxybutyric acid methylester, yielding the (S)-enantiomers of 4-cyano-3-

hydroxybutanoate methylester with 97% ee and 4-chloro-3-hydroxybutyric acid

methylester with 95% ee.59 Kinetic resolution on preparative scale has been reported

with various 3-Alkenyl and heteroaryl chloroalcohols with ee values of >99%.60

In the epoxide opening reaction, HheC mediates high β-regioselectivity for different

1,2-epoxides.54,55,57 This regioselectivity is opposite to that observed for azidolysis of

aryl-substituted epoxides in water, where for example styrene oxide and p-chloro-

styrene oxide display a regioselectivity of 98(α):2(β) and 97(α):3(β), respectively.57

Using HheC, this regioselectivity is changed to 21(α):79(β) for styrene oxide and

11(α):89(β) for p-chloro-styrene oxide.57 HheC has broad substrate specificity and can

be used to prepare a large variety of β–substituted aliphatic and aromatic azido- and

cyano-alcohols.53 With nitrite as nucleophile, the final product is a diol, since the

formed nitrite ester is unstable in water.55 However, nitrite can be used in for example

kinetic resolution of racemic p-nitro-styrene oxide, since HheC displays high

enantioselectivity for the (R)-enantiomer of this substrate.55

4.4.4 Conclusions on enzymatic epoxide transformation

The above section illustrates how enzymes can be used for epoxide-transforming

reactions. High enantioselectivities and regioselectivities can be achieved, making

formation of optically pure products possible. However, for several of these enzymes,

the detailed reaction mechanism is not known and the causes for the displayed

selectivities are not identified. A theoretical study of epoxide-transforming enzymes

helps to understand their selectivity and reaction mechanisms, providing valuable

information that can be used for design of new epoxide-transforming reactions.

Page 33: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

23

Chapter 5

Quantum chemical studies of epoxide-transforming enzymes

In the present thesis, one member from each of the three classes of epoxide-converting

enzymes introduced above was studied computationally. The first enzyme studied is

the Limonene Epoxide Hydrolase (LEH), which is found in the bacterium

Rhodococcus erythropolis DCL14.47 The second enzyme studied here is the human

soluble Epoxide Hydrolase (sEH), which belongs to the superfamily of α/β-fold

hydrolases.61 The third enzyme is the haloalcohol dehalogenase HheC from

Agrobacterium radiobacter AD1.51

We analyzed different aspects of enzymatic epoxide conversion. The reaction

mechanisms employed by the above enzymes were studied in detail, and the role of

different active site residues was analyzed. The importance of certain residues for a

given reaction step was investigated through in silico mutations. Another major topic

of our study was the regioselectivity of epoxide opening. Factors that influence the

regioselectivity were identified and quantified.

O OHOH

21 1 2

4 4

Limonene-1,2-epoxide Limonene-1,2-diol

H2O

Scheme 5.1 Hydrolysis reaction catalyzed by LEH.

Page 34: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

24

5.1 Limonene epoxide hydrolase (Paper I) The limonene epoxide hydrolase (LEH) catalyzes the conversion of limonene-1,2-

epoxide (LE) to limonene-1,2-diol (LD, Scheme 5.1).47,62 LEH is part of a degradation

pathway for limonene in Rhodococcus erythropolis DCL14.63 The crystal structure of

LEH revealed a novel fold and a novel putative active site.64 Due to the fundamental

differences between LEH and other known epoxide hydrolases, LEH was suggested to

be the founding member of a new protein family.47

HO

H

OO

NH2

OOH

OHOH

R

O

ONH2

NN

OHO

NH2

O

NH2

NHNH2

O

OH

OH

R

O

Arg99

Asp101

-

+

Tyr53

Asp132

Asn55

Asp101

-

Tyr53

Asp132

Asn55

Hydrolysis

Arg99

+2H

Scheme 5.2 Epoxide hydrolysis mechanism of LEH.

5.1.1 Elucidating the mechanism of LEH

LEH constitutes a novel enzyme and clues about its mechanism were thus limited.

Mutational studies identified several potentially important residues, including Asp132,

Asp101, Tyr53, and Asn55.64 Also Arg99 was suggested to be of importance, but it

was not clear if its function is only structural or also catalytical. Based on the above

results and the X-ray crystal structure of LEH, a possible Asp132-Arg99-Asp101

catalytic triad was suggested (Scheme 5.2).64 Asp132 was proposed to activate the

nucleophilic water molecule, which attacks the oxirane ring. Asp101 was suggested to

donate a proton to the emerging oxyanion of the substrate. Tyr53 and Asn55 are

involved in binding of the nucleophilic water molecue.64 We studied the putative

mechanism of LEH to establish if the proposal is energetically feasible. The quantum

chemical model was based on the crystal structure of LEH in complex with the

Page 35: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

25

inhibitor valpromide (PDB 1NU3)64. The model was composed of the putative

catalytic triad Asp132-Arg99-Asp101, Tyr53 and Asn55, as well as a crystallo-

graphically observed water molecule (Figure 5.1). Conversion of different substrates

was studied with this model. The LEH-mediated hydrolysis of one stereoisomer of

LE, (1R,2S,4R)-LE, is shown in Figure 5.1. For formation of the diol, (1S,2S,4R)-LD,

a barrier of 14.9 kcal/mol was obtained, and a reaction energy of -9.7 kcal/mol. The

experimental activation energy for LEH-mediated conversion of (1R,2S,4R)-LE is

12.4 kcal/molf, which is close to the computed value.62

Our results show that the suggested catalytic triad indeed is able to catalyze the

conversion of limonene-1,2-epoxide to limonene-1,2-diol. As proposed, Asp132

abstracts a proton from the nucleophilic water molecule, which attacks the epoxide.

Asp101 donates a proton to the epoxide oxygen, in concert with epoxide opening. A

step-wise mechanism was also investigated, but was found to be unlikely based on our

results (Paper I). Arg99 stabilizes and orients the two aspartate residues. It could also

play a pivotal role in transferring a proton from Asp132 to Asp101 in a subsequent

step to restore the active site for the next catalytic cycle. Tyr53 and Asn55 bind the

catalytic water molecule and position it for attack on the substrate. The same non-

covalent general acid/general base mechanism was observed for all studied substrates

(Paper I).

5.1.2 Analysis of the regioselectivity of LEH

LEH accepts only few substrates, among these are the four stereoisomers of limonene-

1,2-epoxide and both enantiomers of 1-methylcyclohexene oxide. Experimental

studies with the two enantiomers of 1-methylcyclohexene oxide (1 and 2, Scheme 5.3)

showed preferred attack at the more substituted oxirane carbon, C1.65 The

regioselectivity for 1-methylcyclohexene oxide was reported to be 85(C1):15(C2).65

However, conversion of limonene-1,2-epoxide exhibited conflicting results. Exclusive

attack at the less substituted carbon (C2) is observed for stereoisomers 3 and 6, while

f The activation energy was determined with a diastereomeric mixture of (1R,2S,4R)- and (1S,2R,4R)-LE. However, as their hydrolysis occurs sequentially, it is assumed that the determined value only is based on the substrate that is converted first, that is, (1R,2S,4R)-LE.

Page 36: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

26

Figure 5.1 LEH-mediated conversion of (1R,2S,4R)-limonene-1,2-epoxide (helicity 3,4 P) to (1S,2S,4R)-limonene-1,2-diol. A) Reactant, B) Transition state, C) Product. Insets show the substrate conformation. Asterisks indicate atoms, which in calculations were fixed to their crystallographically observed position.

Arg99 Asp101

Asp132

Tyr53

Asn55

Substrate (1R,2S,4R)-LE

*

*

*

*

* Water

OH

CH3chair

*

*

*

*

*

O12

half-chair, P-helicityCH3

2.31

1.981.08

1.37

1.021.56

Asp101Arg99

Asp132

Tyr53

Asn55

C1

1.01

1.59

1.70 1.48

1.471.68

1.65

1.99

1.99

1.821.99

1.62

1.79

1.83

1.721.78

1.711.99

1.75

*

*

*

**

*

Asn55

Tyr53

Arg99

Asp132

Asp101

1.001.72

1.64

0.99

1.47

1.62

1.77

1.95 2.01

1.721.96

1.91

A

B

C

OH

OH

CH3chair

Product(1S,2S,4R)-LD

Page 37: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

27

O1 2

4

(1R,2S,4S)

O1 2

4

(1R,2S,4R)

O1 2

4

(1S,2R,4S)

O1 2

4

(1S,2R,4R)

OH1 2

4

(1R,2R,4S)

OHOH

1

4

(1S,2S,4R)

2OH

limonene-1,2-epoxide

(1R,2R)

1-methylcyclohexene oxide

(1S,2S)

O1 2

(1S,2R)

OH1 2

OH

O1 2

(1R,2S)

OH

1 2OH

1-methylcyclohexane-1,2-diol limonene-1,2-diol

1 2 3 4 5 6

7 8 9 10

Scheme 5.3 Conversion of the two enantiomers of 1-methylcyclohexene oxide and the four stereoisomers of limonene-1,2-epoxide by LEH.

exclusive attack at the more substituted carbon (C1) is seen for the stereoisomers 4

and 5 (Scheme 5.3).48,63 The differences in regioselectivity are intriguing, since in

each case the two limonene-1,2-epoxide stereoisomers with the same stereochemistry

at the oxirane ring, (1R,2S) for 3 and 5 and (1S,2R) for 4 and 6, exhibit attack at

opposite carbons (Scheme 5.3). A suggested explanation for the differences was

differential binding of the substrates in the active site, which would lead to attack at

different carbons.64

We investigated the hydrolysis of all stereosiomers of limonene-1,2-epoxide and 1-

methylcyclohexene oxide with our QM model. Interestingly, this relatively small

model is able to reproduce the experimentally observed regioselectivity for all studied

substrates (Paper I). In our model, the orientations of the different stereosiomers are

very similar, as the substrate only interacts with Asp101 (Figure 5.1). These results

indicated that the differences in epoxide opening is not due to differential binding of

the substrate in the active site but has other causes.

Page 38: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

28

OO

O=

Cyclohexane

chair half-chair

Cyclohexene oxide

=

3,4 M helicity 3,4 P helicity half-chair

Scheme 5.4 Preferred conformations of cyclohexane and cyclohexene oxide.

The regioselectivity for conversion of the different cyclohexene oxides is instead

caused by the conformation of the substrate. A cyclohexane ring typically prefers to

be in a chair conformation, but presence of the oxirane ring in cyclohexene oxides

forces the epoxide into a half-chair conformation (Scheme 5.4). The half-chair

conformation can exist in two different forms, which are best described by the helicity

about the 3,4 bond. These will here be referred to as the P and the M helicity

respectively (Scheme 5.4). The two helicities are in equilibrium and are present in

ratios depending on the energy difference between the two conformers. For example,

if a large substituent is present on the ring, one helicity might be significantly

preferred over the other (see below).

The presence of the two different helicity forms is crucial for understanding the

regioselectivity of the above substrates. Equally important is it to realize that in a

given helicity, the two carbons will display very different reactivities. Depending on

which epoxide carbon is attacked, a chair or a twist-boat is formed in the transition

state and in the subsequent product (Scheme 5.5). The chair-like conformation is

energetically preferred and it can be assumed that attack exclusively occurs at the

carbon leading to a chair-like transition state (Paper I). This is independent of the

substitution at the oxirane carbons, i.e. the preferred carbon can be the more or the less

substituted carbon. The above observations can be summarized as follows:

Cyclohexene oxides are half-chairs that can exist in two different helicities. Each

helicity can be attacked at the two oxirane carbons, but the one that will lead to a

chair-like transition state will be preferred. The different regioselectivities reported for

limonene-1,2-epoxide and 1-methyl-cyclohexene oxide can be fully explained

considering these observations.

Page 39: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

29

OOH

OH

OOH

OHH2OH2Ochair twist-boat

3,4 M helicity 3,4 M helicity

1 2 21

Scheme 5.5 Attack on an epoxide half-chair in a given helicity leads to products (and transition states) that have either a chair or a twist-boat conformation.

5.1.2.1 Regioselectivity of 1-methylcyclohexene oxide hydrolysis

Of the two stereoisomers of 1-methylcyclohexene, the (1R,2S) isomer (1, Scheme

5.3), is the preferred substrate of LEH. The two helicities of 1 have almost the same

energy and are assumed to exist in equal amounts (Paper I). For each helicity, attack

occurs at the carbon that will lead to a chair transition state, for 1 in the M-helicity this

is C2 and for the P-helicity this is C1 (Paper I). The two helicity forms of 1 are

competing substrates of LEH, but since attack at the more substituted carbon C1 of the

P-helicity has a lower barrier (14.9 kcal/mol, Table 5.1) than attack at C2 of the M-

helicity (15.9 kcal/mol, Table 5.1), attack occurs preferentially, but not exclusively, at

C1 of the P-helicity (Paper I). A mixture of products is thus expected, with preferred

attack at C1 (of the P-helicity) and minor attack at C2 (of the M-helicity). The

difference in barrier of 1 kcal/mol correspond very well to the experimentally

observed regioselectivity of 85C1:15C2 for 1-methylcyclohexene oxide.65

Table 5.1 Calculated barriers and reaction energies (kcal/mol) for LEH–mediated conversion of 1-methylcyclohexene oxide to 1-methylcyclohexane-1,2-diol.

Substrate a Attack at b

TS b Product c Barrier Reaction (1R,2S), 3,4 M C1 twist-boat (1S,2S) 17.5 -3.4 (1R,2S), 3,4 M C2 chair-like (1R,2R) 15.9 -9.9 (1R,2S), 3,4 P C1 chair-like (1S,2S) 14.9 -9.5 (1R,2S), 3,4 P C2 twist-boat (1R,2R) 19.2 -4.0 (1S,2R), 3,4 M C1 chair-like (1R,2R) 16.0 -9.0 (1S,2R), 3,4 M C2 twist-boat (1S,2S) 19.1 -3.2 (1S,2R), 3,4 P C1 twist-boat (1R,2R) 19.0 -2.8 (1S,2R), 3,4 P C2 chair-like (1S,2S) 15.7 -9.5

a Epoxide stereochemistry and helicity around the 3,4 bond. b Conformation of the substrate in the transition state. c Stereochemistry of resulting diol.

Page 40: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

30

OH

CH3chair

OH

CH3

twist-boat

*

**

*

*

*

*

*

*

*

2.31

1.981.08

1.37

1.021.56

2.141.96

1.511.06

1.42

1.03

Arg99

Asp132

Tyr53

Asn55

Asp101Asp101Arg99

Asp132

Tyr53

Asn55

C2C1

1.79

1.83

1.721.78

1.711.99

1.75

1.76

1.78

1.78

1.81

1.711.96

1.91*

Figure 5.2 Hydrolysis of (1R,2S,4R)-limonene-1,2-epoxide (helicity 3,4 P) with the LEH active site model. The TS for attack at C1 is shown to the left and for attack at C2 to the right. Insets show the substrate conformation. Asterisks indicate atoms, which in calculation were fixed to their crystallographically observed position.

5.1.2.2 Regioselectivity of limonene-1,2-epoxide hydrolysis

For the natural substrate of LEH, limonene-1,2-epoxide, the situation is less complex.

The isopropyl substituent on C4 of limonene-1,2-epoxide determines the preferred

helicity of the half-chair, and for each stereoisomer, only the helicity with the

substituent in an equatorial position will be observed (Paper I). For this helicity, attack

occurs at the carbon that leads to a chair transition state. The transition states for

attack at either C1 or C2 of the stereoisomer 5 are shown in Figure 5.2. Attack at C1

of 5 leads to a chair-like transition state and exhibits a barrier of 14.9 kcal/mol. Attack

at C2 of 5 results in a twist-boat transition state and exhibits a barrier of 19.5 kcal/mol.

The difference in barriers indicates that exclusive attack at C1 is expected, in perfect

agreement with experimental results.48,62 Calculated regioselectivities for the other

stereoisomers are also in agreement with experimental results, with 4 exhibiting

preferred attack at C1, and 3 and 6 exhibiting preferred attack at C2 (Table 5.2).48,63

The regioselectivity of limonene-1,2-epoxide opening is thus not determined by

binding of the substrate in the LEH active site, but by the conformation of the

potential transition states.

Page 41: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

31

5.2 The soluble epoxide hydrolase (Papers II and III)

Table 5.2 Calculated barriers and reaction energies (kcal/mol) for LEH–mediated conversion of limonene-1,2-epoxide to limonene-1,2-diol.

Substrate a Attack at TS b Product c Barrier Reaction (1R,2S,4S), 3,4 M C1 twist-boat (1S,2S,4S) 17.6 -3.5 (1R,2S,4S), 3,4 M C2 chair-like (1R,2R,4S) 16.5 -9.7 (1S,2R,4S), 3,4 M C1 chair-like (1R,2R,4S) 16.1 -9.5 (1S,2R,4S), 3,4 M C2 twist-boat (1S,2S,4S) 19.0 -3.6 (1R,2S,4R), 3,4 P C1 chair-like (1S,2S,4R) 14.9 -9.7 (1R,2S,4R), 3,4 P C2 twist-boat (1R,2R,4R) 19.5 -4.1 (1S,2R,4R), 3,4 P C1 twist-boat (1R,2R,4R) 19.0 -2.8 (1S,2R,4R), 3,4 P C2 chair-like (1S,2S,4R) 16.3 -9.4

a Epoxide stereochemistry and helicity around the 3,4 bond. b Conformation of the substrate in the transition state. c Stereochemistry of resulting diol.

Most known EHs are members of the superfamily of α/β-fold hydrolases. Only a

handful of EHs do not belong to this family. These include the LEH enzyme discussed

above and the mammalian Leukotriene A4 epoxide hydrolase.66 The α/β-fold

hydrolase EHs comprise a large family of enzymes identified in almost all species,

including bacteria, fungi, plants, insects, and mammals. One EH from the α/β-fold

hydrolase family is the soluble epoxide hydrolase (sEH, also referred to as cytosolic

EH, CEH). In humans, sEH is mainly found in the liver, where it converts various

xenobiotic epoxides into their corresponding vicinal diols.66,67

5.2.1 The reaction mechanism of sEH

The mechanism of α/β-fold hydrolase EHs differs substantially from the mechanism

of LEH. In sEH (and the closely related microsomal EHs), a covalent mechanism is

observed, where an active site aspartate becomes covalently bonded to the substrate

(Scheme 5.6).68,69,70,71 The ester bond is subsequently hydrolyzed by water, which is

activated by a His-Asp charge relay, similar to the charge relay in serine

hydrolases.72,73,74,75,76,77 Additionally, the sEHs contain two active site tyrosines that

seem to be of importance in stabilizing and protonating the emerging epoxide

oxyanion.42,61,78,79,80,81,82 A conserved HGXP motif (X = any residue), which is

involved in formation of the oxyanion hole, is also found in sEH.83 The oxyanion hole

stabilizes the tetrahedral intermediate formed in the second part of the reaction.

Page 42: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

32

Scheme 5.6 Hydrolysis mechanism of soluble epoxide hydrolase (residue numbering as in human sEH). Step A is in general referred to as the alkylation half-reaction, while step B and C comprise the hydrolytic half-reaction.

Although the mechanism of sEH has been studied quite well experimentally, a

detailed theoretical analysis has been lacking. We studied the mechanism of sEH

using a quantum chemical model based on the X-ray crystal structure of the human

sEH in complex with the inhibitor cyclohexyl-N´-(iodophenyl) urea (CIU), PDB

1VJ561 (Paper II). The model is composed of parts of the residues Asp495, His523,

Asp333, Tyr381, Tyr465, Trp334, Phe265, Gly264, and Val497. Also included are a

crystallographically observed water molecule and parts of CIU, which was remodelled

into the substrate (1S,2S)-β-methylstyrene oxide (MSO) (Paper II). The model was

used to study the full reaction pathway for attack at C1 and C2 of MSO. The

computed energetics are shown in Figure 5.3.

Page 43: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

33

In general, our results confirm the suggested sEH mechanism (Scheme 5.6). The

alkylation half-reaction seems to occur in two steps. In the first step, the Asp333

carboxylate attacks the epoxide carbon, resulting in formation of a covalent enzyme-

substrate intermediate. At the TS for attack at C1 of MSO, the Asp333 oxygen is

positioned at a distance of 2.29 Å to C1 (Figure 5.4A). The epoxide ring is partially

opened with an O-C1 distance of 1.89 Å. The barrier for this step was calculated to

7.8 kcal/mol (Figure 5.3). The emerging epoxide oxyanion is stabilized by the two

tyrosine residues, but a proton transfer from tyrosine to the formed intermediate

occurs in a separate subsequent step. It was discussed that the stepwise epoxide

opening and protonation might be an artefact of our model (Paper II). However, recent

experimental results for a homologue of human sEH, the potato StEH, give support to

a scenario in which alkylation and protonation do not occur concertedly.84 For StEH,

the amount of tyrosinate formed during the alkylation step was measured to 0.4,

indicating only partly ionization of one or both tyrosines.84 We compute a small

energy difference between the protonated and the unprotonated intermediate,

indicating that the two species will be in equilibrium. The high barrier for the

subsequent hydrolytic step allows formation of such an equilibrium. The mixture of

unprotonated and protonated intermediates would explain the experimentally observed

partial ionization.

In the hydrolytic half-reaction, Asp495 and His523 act in concert to activate a water

molecule, which attacks the Asp333-substrate bond (Figure 5.4B). At the TS for water

attack, the water oxygen is positioned 1.74 Å from the Asp333 carbonyl carbon. One

water proton is transferred to His523 and is located between Nε of His523 and the

water oxygen with distances of 1.23 and 1.27 Å, respectively. The barrier for water

attack was computed to 18.1 kcal/mol (for the C1 pathway). We also observe a proton

transfer from Nδ of His523 to Asp495 in this step (Figure 5.4B). It is possible that this

proton transfer is an artefact of the model that could be avoided if additional residues stabilizing the anionic Asp495 would be included in the model (Paper II).

The formed tetrahedral intermediate is stabilized by the oxyanion hole composed of

hydrogen bonds from the backbone nitrogens of Trp334 and Phe265. In our

calculations, dissociation of the tetrahedral intermediate occurs in concert with proton

Page 44: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

34

transfer from Nε of His523 to the forming diol (Figure 5.4C). The barrier for this step

is computed to 5.5 kcal/mol (Figure 5.3). At the transition state, the scissile O-C bond

has a length of 2.02 Å (Figure 5.4C). The His523 proton is positioned at 1.14 Å from

His523-Nε and 1.42 Å from the emerging anion of the diol. In the overall mechanism,

His523 does thus first act as a general base and then as a general acid. The energetics

computed for conversion of MSO show that the hydrolytic half-reaction is rate-

limiting (Figure 5.3). This is in line with experimental results for the sEH homologues

EchA and StEH.43,85 For attack at C2 of MSO, we observe the same mechanism, but

with slightly different energies (Figure 5.3).

We also tested another mechanistic proposal, in which His523 is protonated at Nε

during the first step of the reaction (Paper III). It has been suggested that such a

protonation would be essential to orient and activate Asp333.85,86 Protonation of

His523 during the alkylation half-reaction was found to be unlikely based on the

calculated energetics (Paper III).

-20

-15

-10

-5

0

5

10

15

20

Attack C2 of MSOAttack C1 of MSO

Covalent intermediate

(protonated)

Covalent intermediate (unprotonated)

7.8 9.5 8.5

14.0

-17.7

-6.4

-8.6

0.0

Reactant

TSwater attack

TSdissociation

Tetrahedral intermediate

11.0 11.1 11.3

14.2

-9.2-8.0

-8.9

Rel

ativ

e en

ergy

(kca

l/mol

)

TS alkylation

Product

This difference is due to differences in hydro-gen bonding patterns.

Figure 5.3 Computed energies for MSO hydrolysis in the wild type human sEH active site model (Paper II).

Page 45: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

35

Figure 5.4 Optimized transition states for hydrolysis of MSO in the wild type sEH model, attack at C1. A) Alkylation TS, B) TS for water attack, C) TS for dissociation.

O

CH3

Ph

O O

H

OO

Tyr465

Asp495His523

Tyr381

HH

OO

O

NCH3

HAsp333

H OH

O

HNH

NN

O

H

Phe265 Gly264

Trp334*

* *

* *

**

*

εδWater1.65

1.94

2.29

1.611.66

1.95

1.78

1.89

2.29

CH3

CH3

Val497*

1.00 1.01

H1.06

1.02

1.03

Tyr465Tyr381

Val497

Asp495His523

Asp333

Trp334

Phe265

Gly264

*

*

*

*

*

*** Water

*A

B

C

Tyr465Tyr381

Val497

Asp495 His523

Trp334

Phe265

Gly264

*

*

*

*

**

*

*

*

Asp333

O

CH3

Ph

O O

H

OO

Tyr465

Asp495 His523

Tyr381

HH

O

O

O

NCH3

HH O

H

O

HNH

NN

O

H

Phe265 Gly264

Trp334*

* *

* *

**

*εδ

1.41

1.27

2.04

1.361.69

1.72

1.69

CH3

CH3

Val497*

1.00 1.52

H1.14 1.23 1.74

1.41 1.271.46

1.03

1.03

Tyr465

Tyr381

Val497

Asp495His523

Trp334

Phe265

Gly264

*

*

*

*

**

*

*

*

Asp333

O

CH3

Ph

O O

H

OO

Tyr465

Asp495His523

Tyr381

HH

OO

O

NCH3

HH

OH

O

HNH

NN

O

H

Phe265 Gly264

Trp334*

* *

* *

**

*εδ

1.09

1.14

2.32

1.021.64

1.75

1.75

1.431.41

CH3

CH3

Val497*

1.01 1.57

1.432.02

H

1.42 1.25

1.03

1.03

Page 46: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

36

5.2.2 Tyrosine mutants of sEH

Table 5.3 Calculated barriers (kcal/mol) for MSO hydrolysis with the wild type and mutant models of human sEH.

Model TS alkylation

TS Dealkylationa

TS water attack

TS dissociation

Overall barrier

WT 7.8 16.4 18.1 5.5 22.6Y381F 15.7 15.7 16.1 6.2 20.9 Y465F 13.2 16.2 16.6 4.7 19.8 Y381F/Y465F 24.8 11.7 - - - a The reverse reaction of the reversible alkylation step

The role of the two active site tyrosine residues in sEH was investigated by in silico

mutations to phenylalanine (Paper III). Full reaction pathways were calculated for the

single mutants Y381F and Y465F (Y = tyrosine, F = phenylalanine), while only the

alkylation step was investigated for the double mutant Y381F/Y465F. The optimized

alkylation transition states for the tyrosine mutants are shown in Figure 5.5. The

barrier of 7.8 kcal/mol observed for attack at C1 of MSO in the wild type sEH model

increased to 13.2 and 15.7 kcal/mol in the Y465F and Y381F models, respectively

(Table 5.3). If both tyrosines are mutated, a barrier of 24.8 kcal/mol is obtained. The

results indicate that the single mutants would remain catalytically active, while the

double mutant would not. This is in agreement with experimental results for the

mouse sEH and the EHs from bacteria and potato, EchA and StEH.80,81,85 It should be

noted that the effect of the single mutations might be overestimated in our model. For

EchA, the rate for alkylation of (R)-styrene oxide changed from 1100 s-1 in the wild

type to 60 s-1 in the Y215F mutant (Y215 in EchA equals Y465 in human sEH).80 This

corresponds to a barrier increase of about 1.8 kcal/mol, while in our calculations, we

observe a barrier increase of 5.4 kcal/mol for mutation of Y465. It is possible that in

the enzyme active site, other polarization effects might compensate for loss of the

Tyr465 hydrogen bond and thus reduce the effect of the Y465F mutation.

The barriers for the subsequent steps of the reaction mechanism, i.e. for attack by

water on the Asp333-ester and for dissociation of the ester bond, changed only 1-2

kcal/mol from the energies computed for the wild type model (Table 5.3). The major

effect of the tyrosine residues is thus in the first step of the reaction, the alkylation.

Page 47: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

37

Figure 5.5 Optimized geometries for the alkylation TS in the Y381F, Y465F, and Y381F/Y465F models. Optimized distances (Å) are shown to the left.

*

O

CH3

Ph

O

H

OO

Tyr465Phe

Asp495His523

Tyr381

H

OO

O

NCH3

HAsp333

H OH

O

HNH

NN

O

H

Phe265 Gly264

Trp334*

* *

* *

**

*

εδWater1.64

1.93

2.29

3.231.55

1.89

1.81

1.97

2.16

CH3

CH3

Val497*

1.02

H1.07

1.01

1.03

Y381F

Tyr465Phe

Tyr381

Val497

Asp495His523

Asp333

Trp334

Phe265

Gly264

*

*

*

*

*

*** Water

*

O

CH3

Ph

H

OO

Tyr465

Asp495His523

Tyr381Phe

OO

O

NCH3

HAsp333

H OH

O

HNH

NN

O

H

Phe265 Gly264

Trp334*

* *

* *

**

*

εδWater1.65

1.93

2.23

3.45

1.49

1.89

1.79

1.97

2.15

CH3

CH3

Val497*

1.04

H1.06

OH

1.03

1.02

Y465F

Tyr465Tyr381Phe

Val497

Asp495

His523

Asp333

Trp334

Phe265

Gly264

*

*

*

**

*

*

Water

*

Tyr465Phe

Tyr381Phe

Val497

Asp495 His523

Asp333

Trp334

Phe265

Gly264

*

*

*

*

*** Water

*

O

CH3Ph

H

OO

Tyr465Phe

Asp495His523

Tyr381Phe

OO

O

NCH3

HAsp333

H OH

O

HNH

NN

O

H

Phe265 Gly264

Trp334*

* *

* *

**

*

εδWater1.58

2.01

2.55

3.154.18

1.79

1.86

1.97

CH3

CH3

Val497*

H1.08

2.01

1.02

1.03

Y381F/Y465F

*

Page 48: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

38

5.2.3 Studies on the regioselectivity of sEH

We used the sEH model to investigate the regioselectivity of epoxide opening for two

substrates, (1S,2S)-β-methylstyrene oxide (MSO) and (S)-styrene oxide (SSO).

Regioselectivities were explored with both the wild type and the tyrosine mutant

models (Paper II and III). The results are summarized in Table 5.4 and 5.5.

The substrate MSO is singly substituted at each oxirane carbon, with a methyl group

on C2 and a phenyl group on C1. As discussed in section 4.1, opening of the epoxide

ring leads to formation of a positive charge at the attacked carbon. Better stabilization

of this charge is expected at the benzylic position of MSO. Indeed, in our calculations,

attack is favoured at the C1 of MSO in all models, even for the double mutant, in

which no general acid is present (Table 5.4). The differences in barriers for attack at

C1 and C2 are 2.9 – 4.4 kcal/mol for the different models, indicating that attack

exclusively occurs at the benzylic position of MSO, also in absence of the two

tyrosine residues.

For SSO, the results are somewhat different (Table 5.5). Removal of the methyl group

from C2 reduces the steric hindrance for attack at this carbon and the barrier for

opening of SSO at C2 is thus only 7.0 kcal/mol, compared to 11.0 kcal/mol for

opening at C2 of MSO. However, the steric advantage for attack at C2 is still lower

than the electronic advantage for attack at C1 of SSO, and attack at C1 remains

preferred, albeit only with 0.7 kcal/mol. The small difference in barriers suggests that

a mixture of products would be formed. The single tyrosine mutants also exhibit

preferred attack at C1 of SSO, while in complete absence of a general acid, steric

factors dominate, resulting in preferred attack at C2 for the double mutant (Table 5.5).

Table 5.4 Regioselectivity of MSO attack with different sEH models. Barriers (in kcal/mol) for the alkylation TS for attack at C1 or C2.

Model Barrier C1 (benzylic) Barrier C2 (homo-benzylic) Regioselectivitya

WT 7.8 11.0 +3.2 Y465F 13.2 16.9 +3.7 Y381F 15.7 20.1 +4.4 Y381F/Y465F 24.8 27.7 +2.9 aDefined as difference in barrier, (barrier C2 – barrier C1).

Page 49: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

39

Table 5.5 Regioselectivity of SSO attack. Barrier (in kcal/mol) for the alkylation TS for attack at C1 or C2.

Model Barrier C1 (benzylic) Barrier C2 (terminal) Regioselectivitya

WT 6.3 7.0 +0.7 Y465F 14.0 14.8 +0.8 Y381F 13.7 15.5 +1.8 Y381F/Y465F 24.2 23.6 -0.6 aDefined as difference in barrier, (barrier C2 – barrier C1).

To our knowledge, the regioselectivities of MSO and SSO opening have not been

studied experimentally with human sEH. However, studies have been performed with

a number of homologues. Conversion of (1S,2S)-β-methylstyrene oxide by the

Aspergillus terreus EH leads to 95% attack at the benzylic position.87 Conversion of

the same substrate by the EH from Rhodotorula glutinis, the rabbit sEH or the mouse

sEH yield virtually exclusive attack at the benzylic position.88,89,90 These

regioselectivities are very much in line with the energetics calculated for MSO.

The regioselectivity of styrene oxide opening with various EHs has given a broad

range of results. Several enzymes exhibit significant attack at both C1 and C2 of SSO,

as suggested from the calculated energetics. For example conversion of SSO leads to

regioselectivities of 45%(α):55%(β) for the rabbit sEH,87 15%(α):85%(β) for the

Aspergillus niger EH91 and 64%(α):36%(β) for Rhodococcus NCIMB 11216 EH

(although the latter seems to be slightly biased by non-enzymatic hydrolysis).92 Other

enzymes exhibit almost exclusive attack at one carbon center, for example with EchA

from Agrobacterium radiobacter AD1, attack occurs exclusively at the terminal

carbon (β) of styrene oxide.43 For the EH from Beauveria sulfurescens and the potato

(Solanum tuberosum) StEH, attack occurs respectively 99% and 98% at the benzylic

position (α) of SSO.91,93 Thus, regioselectivities of styrene oxide opening vary

significantly for different epoxide hydrolases. The experimental results seem

understandable in light of the calculated energetics. For styrene oxide, there is a

considerable steric advantage for attack at the terminal carbon. At the same time there

is an electronic advantage for attack at the benzylic carbon. In our calculations, the

Page 50: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

40

difference in barriers for attack at either carbon of SSO is only 0.7 kcal/mol. The

results indicate that the regioselectivity of SSO opening can be influenced by

differential binding in the active site. Binding that reduces the steric advantage for

attack at C2 would increase the amount of benzylic attack, while binding that reduces

the stabilization through the active site tyrosines would enhance attack at the terminal

position.

5.3 The haloalcohol dehalogenase HheC (Paper IV) The third class of epoxide-transforming enzymes studied in this thesis comprises the

bacterial haloalcohol dehalogenases, which are related to the superfamily of

NAD(P)H-dependent short-chain dehydrogenase/reductases (SDRs).51 The

haloalcohol dehalogenase HheC from Agrobacterium radiobacter AD1 was

introduced above. It catalyzes the conversion of halohydrins into epoxides, but is also

able to catalyze the reverse reaction, the transformation of epoxides into various

substituted alcohols. In the epoxide opening reaction, a number of interesting

nucleophiles are accepted, such CN¯, N3¯, and NO2¯.53,54,55,59

5.3.1 The epoxide-opening mechanism of HheC

The SDRs exhibit a Lys-Tyr-Ser catalytic triad, and based on the similarities of HheC

to SDRs, an Arg149-Tyr145-Ser132 catalytic triad was proposed for HheC (Scheme

5.7). Mutational studies confirmed the importance of these residues for the activity of

HheC.51,52 In the dehalogenation mechanism, Tyr145 is suggested to act as a catalytic

base, abstracting a proton from the halohydrin substrate. Arg149 activates Tyr145 by

abstracting a proton from Tyr145 in concert with epoxide formation. The role of

Ser132 seems to be primarily substrate binding.51,52 In the epoxide opening

mechanism, the proposed catalytic triad can be expected to operate in the reverse

fashion, with Arg149 donating a proton to Tyr145, which donates a proton to the

emerging oxyanion of the alcohol. We studied the epoxide opening mechanism of

HheC with the substrate (R)-styrene oxide (RSO, Paper IV). Two different

nucleophiles were tested, azide and cyanide. The model was based on the crystal

structure of HheC in complex with (R)-1-para-nitro-phenyl-2-azido-ethanol, (PDB

code 1PXO).52 The final QM model with azide and RSO has a size of 130 atoms and

Page 51: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

41

OH

Cl

OH

NN

N

H

H

H

H

OH

R

O

HO

HNN

N

H

H

H

H

OH

R

Cl

Ser132

Tyr145

Arg149

halide binding site

Ser132

Tyr145

Arg149

halide binding site

-

+

Scheme 5.7 Proposed dehalogenation mechanism of HheC.

will in the following be referred to as the wild type model. With this model, we find a

one-step mechanism for the epoxide opening, where nucleophilic attack by azide and

proton transfer from Tyr145 occur concertedly. The transition state for azidolysis at

the β-carbon of RSO is shown in Figure 5.6A. Attack of azide at the terminal carbon

occurs at a distance of 2.16 Å. The Cβ-O bond is elongated to 1.79 Å. A proton

transfer from Arg149 to Tyr145 was not observed, and in the formed product

geometry, a tyrosinate has been formed (Paper IV). Arg149 stabilizes the tyrosinate

through hydrogen-bonding. The barrier for the concerted proton transfer from Tyr145

and epoxide opening at Cβ has a computed barrier of 8.1 kcal/mol. The overall

reaction energy is calculated to -25.2 kcal/mol, which agrees well with the observed

irreversibility of HheC-mediated azidolysis.57

The computed energies show that the proposed epoxide opening mechanism with

azide is energetically feasible. For cyanolysis of RSO, the same mechanism is found.

The TS for attack at the β-carbon of RSO by cyanide is shown in Figure 5.6C. The

optimized distances are similar to the TS for azidolysis, with the main difference

being the nucleophile-Cβ distance, which is somewhat longer (2.32 Å). The computed

energies for cyanolysis are somewhat different than for azidolysis. The barrier for

nucleophilic attack of cyanide at Cβ is calculated to 7.0 kcal/mol, while the overall

reaction energy is -46.0 kcal/mol.

Page 52: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

42

C D

1.91

*

*

*

*

*

*

*

*

*

*

*

Phe12

Arg149

Tyr145

Pro175

Leu178

Tyr177

Tyr187

Asn176

Ser1321.79

1.02

2.121.96 1.93

1.07

2.13

1.74

2.32

1.82 1.39Tyr1451.13

*

*

* *

*

*

*

*

*

*

*

Phe12

Arg149

Pro175

Leu178

Tyr177

Tyr187

Phe186Asn176

Ser1321.92

1.281.85

1.73

1.972.13

2.68

1.03

1.87

1.92

1.82

Phe186

1.77

1.83 1.36

1.95

2.16

1.79

1.08

1.03

1.031.91

2.181.98

2.08

*

*

*

*

**

*

*

*

*

*

**

Phe12

Arg149Tyr145

Pro175

Leu178

Tyr177

Tyr187

Phe186Asn 176

Ser132

A

*

1.08

1.821.36

2.09

2.45

1.021.99

1.741.03

1.80

1.96

2.182.10

* *

*

*

*

**

*

*

*

*

*

**

Phe12

Arg149

Tyr145

Pro175

Leu178

Tyr177 Tyr187

Phe186

Asn176

Ser132

B

Figure 5.6 Transition states for cyanolysis and azidolysis of RSO in the HheC active site model. A) Attack of azide at Cβ, B) Attack of azide at Cα, C) Attack of cyanide at Cβ, D) Attack of cyanide at Cα.

Page 53: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

43

From the computed epoxide opening reactions it can be concluded that the role of

Tyr145 is binding and protonation of the epoxide, with aid of Arg149. It is less clear

what function Ser132 is fulfilling. An experimental Ser132Ala mutant of HheC

exhibited complete loss of activity in the dehalogenation reaction, indicating that

Ser132 is essential for catalysis.51 The interaction between Ser132 and the substrate

might be important for substrate positioning. Additionally, Ser132 could have a role in

stabilizing the emerging epoxide oxyanion, analogous to the role of the second

tyrosine residue in sEH (Paper III). We prepared a Ser132Ala mutant and calculated

the barriers for cyanolysis in this model (Paper IV). Mutation of Ser132 raises the

barrier for attack at Cβ by 2.2 kcal/mol compared to the wild type model (Table 5.7).

This shows that Ser132 plays a role in lowering the barrier for epoxide opening.

However, the barrier increase in absence of Ser132 does not explain the loss of

activity of the experimental Ser132Ala mutant.51 It is possible that Ser132 also is

essential for substrate binding in the active site, which could explain the observed

inactivity of the Ser132Ala mutant.

5.3.2 Regioselectivity of wild type HheC

Experimental studies show that HheC mediates attack at the less substituted carbon of

various epoxides with the nucleophiles azide, cyanide, and nitrite.53,54,55,57,59 For

azidolysis of styrene oxide, the observed regioselectivity is different from that in

solution. While chemical conversion of styrene oxide with NaCN results in 3% attack

at the terminal carbon (Cβ), HheC-mediated azidolysis increases this value to 79%.57

We studied the regioselectivity for azidolysis of styrene oxide with two different

models. The first model comprises only RSO, azide, and a phenol residue as acid

(Figure 5.7). In this model, the calculated barriers for attack of azide at Cα and Cβ of

RSO are 10.7 and 13.5. kcal/mol, respectively (Table 5.6). The observed

regioselectivity is in line with the strong preference for the Cα in the chemical

azidolysis of styrene oxide.57 With the HheC wild type active site model of 130 atoms,

the barriers for azidolysis were computed to 8.6 and 8.1 kcal/mol, respectively, for

attack at Cα and Cβ (Figure 5.6, Table 5.6). The regioselectivityg has thus changed

gThe regioselectivity is here defined as the difference in barriers, (Barrier Cβ) – (Barrier Cα).

Page 54: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

44

Figure 5.7 Phenol-catalyzed azidolysis and cyanolysis of RSO. A) Attack of azide at Cα, B) Attack of azide at Cβ, C) Attack of cyanide at Cα, D) Attack of cyanide at Cβ.

2.40

1.561.02

1.81

2.23

1.77

1.611.01

A B

α

β

2.49

1.601.01

1.73

2.40

1.70

1.60 1.00

C D

αβ

significantly, from +2.8 kcal/mol in the phenol-catalyzed model to -0.5 kcal/mol in the

HheC active site model. The slight preference for Cβ in the HheC active site model is

in line with experimental results.57 The reported regioselectivity of HheC-mediated

azidolysis of RSO is 79%(β):21%(α).57 This corresponds approximately to a

difference in barriers of -0.8 kcal/mol.

For cyanolysis of RSO, we calculated a similar pattern. With the phenol-catalyzed

model (Figure 5.7), the barriers for attack at Cα and Cβ are 10.5 and 11.0 kcal/mol,

i.e. a slight preference for the Cα is observed. With the HheC active site model

(Figure 5.6), the barriers for cyanolysis are 10.6 and 7.0 kcal/mol respectively, for

attack at Cα and Cβ. The regioselectivity has thus changed from +0.5 in the phenol-

catalyzed model to -3.6 kcal/mol in the HheC active site model (Table 5.6).

Page 55: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

45

Table 5.6 Calculated barriers (kcal/mol)a for azidolysis and cyanolysis of RSO.

Model (# atoms) Nucleophile Barrier Cα Barrier Cβ Regioselectivity Phenol-catalyzed (33) N3¯ 10.7 13.5 +2.8 HheC active site (130) N3¯ 8.6 8.1 -0.5 Phenol-catalyzed (32) CN¯ 10.5 11.0 +0.5 HheC active site (129) CN¯ 10.6 7.0 -3.6 a Solvation effects calculated with ε = 80 for the phenol-catalyzed and ε = 4 for HheC active site models.

For both the azidolysis and the cyanolysis of RSO, HheC promotes attack at Cβ with

several kcal/mol compared to the phenol-catalyzed epoxide opening. We were

interested in establishing what effects cause this change in regioselectivity. For this

purpose, different functional groups in the active site model were mutated individually

to establish their influence on the regioselectivity of RSO conversion (Paper IV).

5.3.3 Regioselectivity of HheC mutants

Analysis of the optimized structures for the cyanolysis and azidolysis of RSO in the

wild type HheC active site model indicate that there are multiple effects that might

mediate the preference for attack at the Cβ. These include the position and orientation

of the nucleophile and the substrate in the active site, steric effects mediated by

surrounding residues, and electrostatic stabilization of attack at Cβ. These possible

effects were analyzed in depths in the following.

One residue that might influence the regioselectivity of epoxide-opening is the

backbone carbonyl of Pro175. The Pro175 CO appears to interact with the Cβ-

hydrogen of RSO (Scheme 5.8), which might aid in stabilization of the positive charge

that evolves at the β-carbon during epoxide opening. The optimized distances of the

wild type model support this idea. In the reactant structure with azide, the distance

from the Pro175 CO to the Cβ-hydrogen is 2.54 Å. At the transition state for attack of

azide at Cα, this distance is 2.40 Å, while at the transition state for Cβ attack, the

distance has been shortened to 2.06 Å, indicating an interaction. For cyanolysis, the

Page 56: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

46

O

N

H

H

OH R

αβPro175

Scheme 5.8 Interaction of the backbone CO of Pro175 with the Cβ hydrogen of RSO.

Pro175-CO to Cβ-hydrogen distance is 2.29 Å in the reactant structure, 2.31 Å at the

transition state for Cα attack and 2.15 Å at the transition state for Cβ attack. The

interaction of Pro175 with the Cβ-hydrogen of RSO might facilitate attack at Cβ,

thereby reducing the preference for the Cα atom. We tested this hypothesis by

mutating the backbone carbonyl of Pro175 into CH2. The transition states for

cyanolysis at Cα and Cβ in the Pro175CO→CH2 model are shown in Figure 5.8. The

barrier for attack at Cα is 10.8 kcal/mol, which is almost the same as in the wild type

model (10.6 kcal/mol). However, the barrier for attack at Cβ is raised by 1.5 kcal/mol

compared to the wild type model (Table 5.7). This decreases the preference for attack

at Cβ by 1.3 kcal/mol, although it is not sufficient to reverse the regioselectivity.

Other effects that might influence the regioselectivity are the position and orientation

of the substrate and the nucleophile. For example, in the optimized reactant of the

wild-type model, the distance from the cyanide carbon to the Cα and Cβ carbons of

RSO are respectively 4.44 Å and 3.47 Å. For a reaction in solution, where the

nucleophile and the substrate are not restricted in their movements, this distance

difference might be less critical. However, in the HheC active site, large movements

of the substrate or the nucleophile might be energetically costly, as they could result in

changes in the hydrogen bonding pattern or in steric interactions with surrounding

residues. We tested what effect the hydrogen bonds to RSO and cyanide have on the

regioselectivity of attack. The epoxide forms hydrogen bonds with the hydroxyl

groups of Ser132 and Tyr145. Tyr145 can be considered essential for the reaction

mechanism and it was thus only tried to investigate how mutation of Ser132 affects

the regioselectivity of epoxide opening by mutating Ser132 into an alanine residue.

Page 57: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

47

Figure 5.8 Transition states for RSO cyanolysis in the Pro175-CO→CH2 model, A) Attack at Cβ, B) Attack at Cα. The mutated backbone carbon atom is marked with ∆.

1.92

*

*

*

*

*

*

*

*

*

*

Phe12

Arg149Tyr145

Pro175

Leu178

Tyr177

Tyr187

Asn176

Ser132

1.02

2.09

1.82

2.00

1.05

1.72

2.35

1.80

1.43

Phe186*

2.00

*

*

*

**

*

*

*

*

*

Phe12

Arg149

Tyr145

Pro175

Leu178

Tyr177

Tyr187

Asn176

Ser1321.02

2.10

1.82

2.06

1.07

1.85

2.72

1.71

1.38

Phe186

**

*

BA

The barrier for attack at Cβ in the Ser132→Ala model was calculated to be 9.2

kcal/mol, while for nucleophilic attack of cyanide at Cα, we calculate a barrier of 12.1

kcal/mol. The regioselectivity is -2.9 kcal/mol, which is 0.7 kcal/mol less than in the

wild type model (Table 5.7). Thus, the interaction of the substrate with Ser132

contributes little to the preferred β-regioselectivity.

Table 5.7 Calculated barriers (kcal/mol)a for cyanolysis of RSO with different models. Model (# atoms) Barrier Cα Barrier Cβ Regioselectivity Phenol-catalyzed (32) 10.5 11.0 +0.5 HheC wild type (129) 10.6 7.0 -3.6 HheC Ser132 → Ala (128) 12.1 9.2 -2.9 HheC Pro175-CO → CH2 (130) 10.8 8.5 -2.3 HheC Leu178NH → O (128) 9.1 6.5 -2.6 HheC RmTyr187 (116) 7.9 7.8 -0.1 a Solvation effects calculated with ε = 80 for the phenol-catalyzed and ε = 4 for HheC active site models.

Page 58: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

48

The position of cyanide in the halide binding site might have an effect on the

regioselectivity. In the HheC active site model, cyanide forms hydrogen bonds with

the backbone nitrogen of Leu178 and the water molecule (Figure 5.6). A weak

interaction is also observed with the backbone NH of Tyr177. We tested if the

hydrogen bond from cyanide to the backbone nitrogen of Leu178 has a significant

effect on the regioselectivity of epoxide opening (Paper IV). In the Leu178NH→O

model, the backbone amide was mutated into an ester bond, i.e. the NH was replaced

by oxygen. This eliminates the backbone hydrogen bond to cyanide. The barrier for

attack at Cα is somewhat reduced in this model (9.1 kcal/mol, compared to 10.6

kcal/mol in the wild type, Table 5.7) and the regioselectivity is reduced to -2.6

kcal/mol. This indicates that the Leu178NH hydrogen bond has an effect on the

regioselectivity of attack.

The position of the nucleophile could also be affected by steric hindrance caused by

surrounding residues. One residue that might have an effect on the movement of the

nucleophile is Tyr187. It has previously been suggested that Tyr187 contributes to the

rigidity of the halide binding site, probably through its interaction with Asn176.52,58

We prepared an HheC active site model in which Tyr187 was removed (Figure 5.9).

In the optimized structures of the RmTyr187 model, the position of cyanide has

changed slightly, resulting in a more favourable interaction with the backbone NH of

Tyr177. In the reactant of the wild type model, the distance from the nitrogen of

cyanide to the hydrogen of the Tyr177 amide is 2.73 Å, while in the RmTyr187

model, this distance is only 2.10 Å. This indicates that in the wild type model, Tyr187

hinders cyanide from positioning itself close to the Tyr177 backbone. In the

RmTyr187 model, the computed barriers are 7.9 and 7.8 kcal/mol for attack at Cα and

Cβ, respectively (Table 5.7). Thus, the regioselectivity is reduced to only -0.1

kcal/mol, implying that cyanolysis would lead to a racemic mixture of cyanoalcohols.

The regioselectivity of the RmTyr187 mutant is thus quite close to the regioselectivity

of the phenol-catalyzed model.

We also tested the effect of Tyr187 on RSO azidolysis. The computed barriers for

azidolysis in the RmTyr187 model are 5.6 kcal/mol for attack at Cα and 8.1 kcal/mol

Page 59: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

49

Figure 5.9 Transition states for cyanolysis of RSO in the HheC RmTyr187 model, A) Attack at Cβ, B) Attack at Cα.

ATyr145

1.77

*

**

***

*

*

*

Phe12

Arg149

Pro175

Leu178

Tyr177

Phe186

Asn176

Ser132

1.42

2.14

2.31

1.03 1.86

*

*1.86

1.74

*

2.101.96

1.06

2.30

Tyr145

*

*

*

*

*

*

*

*

Phe12

Arg149

Pro175

Leu178Tyr177

Phe186

Asn176

Ser132

1.92

1.96

2.65

1.301.86

1.87

**

*

1.03

2.19

*

2.072.12

1.12

1.81

B

for attack at Cβ. The resulting regioselectivity is +2.5 kcal/mol, which is similar to the

value obtained with the phenol-catalyzed model (+2.8 kcal/mol, Table 5.6). For both

azidolysis and cyanolysis, Tyr187 is thus identified as being critical for the observed

change in regioselectivity in going from the phenol-catalyzed to the HheC active site

model.

It can be argued that the effect of Tyr187 in the active site model is exaggerated due to

the freezing scheme employed. In our model, Tyr187 is truncated and fixed at Cγ,

while in the native enzyme, the side chain would be extended with Cβ and might

exhibit larger flexibility. However, the interaction of Tyr187 with Asn176 (and also

with Trp249) observed in the HheC crystal structure indicates that Tyr187 has little

flexibility also in the native enzyme.52,58

From the above analysis, the factors governing the regioselectivity of HheC-mediated

epoxide opening can be summarized as follows (Scheme 5.9):

Page 60: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

50

Nu-

O

OR

halide binding site

αβ

Tyr187

Pro175

H2O

Leu178 Tyr177

Ser132 Tyr145

Scheme 5.9 Illustration of effects governing the regioselectivity of HheC-mediated epoxide opening.

• The nucleophile and the substrate are bound in the active site through multiple

hydrogen bonds. This restricts their movement relative to each other. Removal

of for example the hydrogen bond from cyanide to the Leu178 backbone

reduces the regioselectivity of RSO cyanolysis by 1.0 kcal/mol.

• The backbone carbonyl of Pro175 stabilizes opening at Cβ. Mutation of the

Pro175 carbonyl reduces the regioselectivity of RSO cyanolysis by 1.3

kcal/mol.

• Tyr187 imposes steric hindrance on the nucleophile, making attack at Cα less

favourable. Removal of Tyr187 reduces the regioselectivity of RSO cyanolysis

by 3.5 kcal/mol, resulting in equally preferred attack of cyanide at Cβ and Cα.

Page 61: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

51

Chapter 6

Conclusions

The presented theoretical studies of epoxide-transforming enzymes give a deeper

understanding of the mechanistic strategies employed in epoxide conversion. Several

elements can be identified that are common for all three enzymes. Also, the theoretical

results make it possible to evaluate the effects that contribute to the observed

regioselectivities.

6.1 Mechanistic strategies of epoxide-transforming enzymes At first glance, the mechanisms and catalytic residues employed by the studied

enzymes appear to be quite different. LEH employs a concerted general acid/general

base mechanism mediated by an Asp-Arg-Asp triad. The nucleophile in LEH is an

activated water molecule (Paper I). sEH exhibits an Asp-His-Asp catalytic triad in

combination with two tyrosine residues and employs a covalent reaction mechanism.

The nucleophile in sEH is the side chain carboxy of Asp333 (Paper II and III). HheC

employs an Arg-Tyr-Ser catalytic triad to perform a concerted, acid-catalyzed

reaction. A variety of nucleophiles are accepted by HheC, including azide, cyanide,

and nitrite (Paper IV).

Comparison of the studied mechanisms reveals certain mechanistic features, which are

shared by all three enzymes. For example, in all cases, epoxide opening takes place

through an SN2 mechanism, in which epoxide opening and nucleophilic attack occur

concertedly (Paper I, II and IV). An SN1 mechanism, in which epoxide opening and

nucleophilic attack occur step-wise, is not observed for any of the studied enzymes or

substrates. For all three enzymes, a general acid is present, which donates a proton to

the emerging oxyanion. For HheC and LEH, proton donation occurs concertedly with

epoxide opening. If the general acid is eliminated, the barriers for epoxide opening

Page 62: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

52

increase substantially. For example, the ring opening of (1R,2S)-1-methylcyclohexene

oxide (3,4 P) was studied with an LEH active site model in which Asp101 was

deprotonated (Paper I). In that situation, the barrier for epoxide opening increases

from 14.9 kcal/mol to 44.1 kcal/mol (Paper I). If both tyrosines in sEH are mutated to

phenylalanines, the barrier for MSO opening increases from 7.8 kcal/mol to 24.8

kcal/mol (Paper III). In each case, removal of the general acid renders the enzyme

catalytically inactive. The theoretical results are consistent with experimental studies

of sEH and LEH mutants. If both tyrosines are mutated in the bacterial EchA, the

mouse sEH or the potato StEH, virtually no catalytic activity is detected.80,81,85

Mutation of Asp101 to alanine or asparagine in LEH also results in a catalytically

inactive enzyme.64

A feature shared by HheC and sEH is the presence of an additional hydrogen bond

donor to the epoxide oxygen, besides the general acid. In HheC this is an optimally

positioned serine residue, and in sEH a second tyrosine is present. In silico mutation

of the second tyrosine in sEH to phenylalanine results in a barrier increase from 7.8 to

13.2 or 15.7 kcal/mol for the epoxide opening reaction, respectively, for the

Tyr465Phe and the Tyr381Phe mutations (Paper III). In HheC, mutation of Ser132 to

alanine results in a barrier increase from 7.0 to 9.2 kcal/mol for attack of cyanide at

Cβ of RSO (Paper IV). The additional hydrogen bond donor does thus provide a

barrier reduction of several kcal/mol for both sEH and HheC.

6.2 Regioselectivities of epoxide-transforming enzymes The regioselectivities of epoxide opening are substantially different for the studied

enzymes. sEH mediates preferred attack at the more substituted, benzylic carbon of

(1S,2S)-β-methylstyrene oxide (MSO) and (S)-styrene oxide (SSO) (Paper II), while

HheC mediates preferred attack at the less substituted, terminal carbon of (R)-styrene

oxide (Paper IV). LEH mediates preferred attack at either the more or the less

substituted carbon, depending on the substrate (Paper I). In each case, very different

factors govern the regioselectivity. The regioselectivity of epoxide opening is in

general influenced by steric and by electronic factors. For LEH, the regioselectivity of

limonene-1,2-epoxide and 1-methylcyclohexene oxide conversion is dominated by

conformational factors (Paper I). The structure of the formed transition state

Page 63: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

53

determines if attack occurs at either the more substituted (C1) or the less substituted

(C2) carbon. In each case, attack will be favoured at the atom that leads to a chair-like

transition state (Paper I). In sEH, electronic factors seem to dominate the opening of

MSO and SSO. The stabilization provided by the phenyl substituent of the substrate

results in attack at the benzylic carbon, a preference that is further enhanced by

presence of the two tyrosine residues (Paper III). When both tyrosines in sEH are

mutated to phenylalanine, steric factors play a more dominant role and preferred

attack at the terminal carbon of SSO is observed (Paper III). For HheC, both steric and

electronic factors can be identified that mediate preferred attack at the terminal carbon

of RSO. For example, the backbone carbonyl of Pro175 interacts with the hydrogen

on the terminal carbon of RSO, providing electronic stabilization to the emerging

positive charge. In silico mutation of the Pro175 backbone carbonyl to a CH2 reduces

the preference of cyanide for the β-carbon by 1.3 kcal/mol (from -3.6 kcal/mol in the

wild type model to -2.3 in the Pro175-CO → CH2 model, Paper IV). In addition, steric

factors reduce the advantage for attack at the benzylic position of RSO. For example

Tyr187 seems to hinder the movement of the nucleophile (Paper IV). If Tyr187 is

removed from the model, the regioselectivity is reduced to only -0.1 kcal/mol.

The observed effects on the regioselectivity of epoxide opening can be summarized in

regard to their nature. Electronic effects include stabilization mediated by substituents

on the substrate, the presence of hydrogen bond donors/general acids to the epoxide

oxygen, and stabilization effects provided by the surroundings such as the backbone

carbonyl of Pro175 in HheC. Steric effects on the regioselectivity are caused by the

substitution on the substrate (attack at an less substituted carbon is sterically

preferred), conformations of the formed transition states (such as for limonene-1,2-

epoxide), and the orientations imposed on the substrates/nucleophiles, when they are

bound in the active site.

6.3 Final Conclusions The presented results show how a theoretical approach can give valuable information

about enzymatic mechanisms. For example, the detailed analysis of individual effects

influencing the regioselectivity of epoxide opening in HheC would not be possible in

Page 64: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

54

a similar fashion in an experimental setting. Many mutations which easily are

introduced in silico would be almost impossible to introduce in vitro/in vivo. Also the

characterization of short-lived transition states is not (yet) possible with conventional

experimental methods. Only indirect strategies can be employed, such as structural

characterization of enzymes in complex with transition state analogues. In the

computational approach, it is possible to optimize transition state structures, which are

of great aid in explaining reaction mechanisms. For example, the discrimination

between different mechanistic pathways in LEH-mediated conversion of limonene-

1,2-epoxide can be explained with the conformations of the potential transition states.

Overall, this thesis confirms the usefulness of a computational approach in

investigating enzymatic reaction mechanisms.

Page 65: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

55

References

1. Koch, W.; Holthausen, M.C. A Chemist’s Guide to Density Functional Theory. 2nd ed., 2001, Wiley-VCH Verlag GmbH.

2. Hohenberg, P.; Kohn, W. Phys. Rev. 1964, 136, B864 - B871. 3. Kohn, W.; Sham, L. J. Phys. Rev. 1965, 140, A1133-A1138. 4. Jensen, F. Introduction to Computational Chemistry, 1999, John Wiley & Sons

Ltd, England. 5. Vosko S.H.;Wilk L.; Nusair M. Can. J. Phys. 1980, 58, 1200-1211. 6. Becke, A. D. Phys. Rev. A 1988, 38, 3098-3100. 7. Lee, C.; Yang, W.; Parr, R.G. Phys. Rev. B 1988, 37, 785-789. 8. Becke, A. D. J. Chem. Phys. 1993, 98, 5648-52. 9. Bauschlicher, C.W. jr., Chem. Phys. Lett. 1995, 246, 40-44. 10. Bauschlicher, C.W. jr., Patridge, H. Chem. Phys. Lett. 1995, 240, 533-540. 11. Curtiss, L.A.; Redfern, C.A.; Raghavachari, K. J. Chem. Phys. 2005, 123,

124107. 12. Glukhovtsev, M.N.; Bach, R.D.; Pross, A.; Radom, L. Chem. Phys. Lett. 1996,

260, 558-564. 13. Baker, J.; Muir, M.; Andzelm, J. J. Chem. Phys. 1995, 102, 2063-2079. 14. Guner, V.; Khuong, K.S.; Leach, A.G.; Lee, P.S.; Bartberger, M.D.; Houk,

K.N. J. Phys. Chem. A. 2003, 107, 11445-11459. 15. Siegbahn, P.E.M. J. Biol. Inorg. Chem. 2006, 11, 695–701. 16. Lundberg, M, Siegbahn, P.E.M. J. Chem. Phys. 2005, 122, 224103. 17. Yhang, Z., Zang, W. J. Chem. Phys. 2004, 109, 2604-2608. 18. Csonka, G.I.; , Johnson, B.G. Theor. Chem. Acc. 1998, 99, 158-165. 19. Patchkovskii,S.; Ziegler, T. J. Chem. Phys. 2002, 116, 7806. 20. Gaussian 03, Frisch, M. J. et al. Gaussian, Inc., Wallingford CT, 2004. 21. (a) Klamt, A.; Schüürmann, G., J. Chem. Soc. Perkin. Trans. 1993, 2, 799-805,

(b) Andzelm, J.; Kölmel, C.; Klamt, A., J. Chem. Phys. 1995, 103, 9312-20, (c) Barone, V.; Cossi,, M. J. Phys. Chem. A 1998, 102, 1995-2001. (d) Cossi, M.; Rega, N.; Scalmani, G.; Barone, V. J.Comput Chem. 2003, 24, 669-81.

22. Himo, F. Theor. Chem. Acc. 2006, 116, 232–240. 23. March, J., Advanced Organic Chemistry: Reactions, Mechanism and Structure,

4th ed., John Wiley & Sons, New York, 1992. 24. Badalassi, F.; Klein,G.; Crotti,P.; Reymond, J.-L. Eur. J. Org. Chem. 2004,

2557-2566. 25. Smith, J. G. Synthesis 1984, 629. 26. Blumenstein, J.J.; Ukachukwu, V.C.; Mohan, R.S.; Whalen, D.L. J. Org.

Chem. 1993, 58, 924.

Page 66: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

56

27. Audier, H. E.; Dupin, J. F.; Jullien, J. Bull. Soc. Chim. Fr. 1968, 9, 3850-55. 28. Chini, M.; Crotti, P.; Macchia F. Tetrahedron Lett. 1990, 31, 5641-5644. 29. Van de Weghe, P.; Collin, J. Tetrahedron Lett. 1995, 36, 1649-52. 30. Fringuelli, F.; Piermatti, O.; Pizzo, F.; Vaccaro, L. J. Org. Chem. 1999, 64,

6094-6096. 31. Jacobsen, E.N. Acc. Chem. Res. 2000, 33, 421-431. 32. Onaka, M.; Ohta, A.; Sugita, K.; Izumi, Z. Applied Catalysis A: General 1995,

125, 203-216. 33. Mirmashhori, B.; Azizi, N.; Saidi, M.R. J. Mol. Catal. A: Chemical 2006, 247,

159–161. 34. Gassman, P.G.; Guggenheim, T.L. J. Am. Chem. Soc. 1982, 104, 5849-5850. 35. Chini, M.; Crotti, P.; Favero, L.; Macchia F. Tetrahedron Lett. 1991, 32, 4775-

4778. 36. Srinivas, B.; Pavan Kumar, V.; Sridhar, R.; Surendra, K.; Nageswar, Y.V.D.;

Rama Rao, K. J. Mol. Catal. A: Chemical 2007, 261, 1–5. 37. Iranpoor, N.; Firouzabadi, H.; Shekarize, M. Org. Biomol. Chem. 2003, 1, 724-

727. 38. Mitchell, D.; Koenig, T.M. Tetrahedron Lett. 1992, 33, 3281-84. 39. Bonini, C.; Righi, G. Synthesis 1994, 225-238. 40. Orru, RVA; Faber, K.; Curr. Opin. Chem. Biol. 1999, 3,16–21. 41. Rink, R.; Fennema, F.; Smids, M.; Dehmel, U.; Janssen, D.B. J. Biol. Chem.

1997, 272, 14650-14657. 42. Nardini, M.; Ridder, I.S.; Rozeboom, H.J., Kalk, K.H. ; Rink, R.; Janssen, D.

B.; Dijsktra, B. W. J. Biol. Chem. 1999, 274, 14579-14586. 43. Rink, R.; Janssen, D. B.. Biochemistry 1998, 37, 18119-18127. 44. Rui, L.; Cao, L.; Chen, W.; Reardon, K.F.; Wood, T.K.; Appl. Environ.

Microbiol. 2005, 71, 3995–4003. 45. Cao, L..; Lee, J.; Chen, W.; Wood, T.K. Biotech. Bioeng. 2006, 94, 522-529. 46. Pedragosa-Moreau, S.; Archelas, A.; Furstoss, R. J. Org. Chem . 1993, 58,

5533-5536. 47. Barbirato, F.; Verdoes, J.C.; de Bont, J. A. M.; van der Werf, M. J. FEBS Letters 1998, 438, 293-296. 48. van der Werf, M.J.; Orru, R.V.A.; Overkamp, K.M.; Swarts, H.J.; Osprian, I.;

Steinreiber, A.; de Bont, J.A.M.; Faber, K., Appl. Microbiol. Biotechnol. 1999, 52, 380-385.

49. Nakamura, T.; Nagasawa, T.; Yu, F.; Watanabe, I.; Yamada, H. App. Environ. Microbiol. 1994, 60, 1297-1301

50. Van den Wijngaard, A.J.; Reuvekamp, P.T.W., Janssen, D.B. J. Bacteriol. 1991, 173, 124-129.

51. van Hylckama Vlieg, J.E.T.; Tang, L.; Lutje Spelberg, J.H.; Smilda, T., Poelarends; G.J.; Bosma, T.; van Merode, A.E.; Fraaije, M.W.; Janssen, D.B., J. Bacteriol. 2001, 183, 5058-66.

52. de Jong R.M., Tiesinga J.J., Rozeboom H.J., Kalk K.H., Tang L., Janssen D.B., Dijkstra B.W., EMBO J. 2003, 22, 4933-44.

Page 67: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

57

53. Janssen, D.B.; Majerić-Elenkov, M.; Hasnaoui, G.; Hauer, B.; Lutje Spelberg, J.H. Biochem. Soc. Trans. 2006, 34, 291–295. 54. Majerić-Elenkov, M.; Hauer, B.; Janssen, D.B. Adv. Synth. Catal. 2006, 348,

579-585. 55. Hasnaoui, G.; Lutje Spelberg, J.H.; de Vries, E.; Tang, L.; Hauer, B.; Janssen,

D.B. Tetrahedron: Asymmetry 2005, 16, 1685–1692. 56. Nakamura, T.; Nagasawa, T.; Yu, F.; Watanabe, I.; Yamada, H. Biochem.

Biophys. Res. Commun. 1991, 180, 124-130. 57. Lutje Spelberg, J.H.; van Hylckama Vlieg, J.E.T.; Tang, L.; Janssen, D.B.;

Kellogg, R.M. Org. Lett. 2001, 3, 41-43. 58. Tang, L.; Pazmiño, D.E.T, Marco W. Fraaije, M.W.; de Jong, R.M.; Dijkstra,

B.W.; Janssen, D.B. Biochemistry 2005, 44, 6609-6618. 59. Majerić-Elenkov, M.; Tang, L.; Hauer, B.; Janssen, D.B. Org. Lett. 2006, 8,

4227-29. 60. Haak, R.M.; Tarabiono, C., Janssen, D.B.; Minnaard, A.J.; de Vries, J.G.;

Feringa, B.L. Org. Biomol. Chem. 2007, 5, 318–323. 61. Gomez, G.A., Morisseau, C.; Hammock, B.D.; Christianson, D.W.

Biochemistry 2004, 43, 4716-4723. 62. van der Werf, M. J.; Overkamp, K.M.; Bont, J.A.M., J. Bacteriol. 1998, 180,

5052-5057. 63. van der Werf, M.; Swarts, H.; de Bont, J.A.M., Appl. Environ. Microbiol.

1999, 65, 2092-2102. 64. Arand, M.; Hallberg, B.M.; Zou, J.; Bergfors, T.; Oesch, F.; van der Werf, M.;

de Bont, J. A. M.; Jones, T.A.; Mowbray, S.L., EMBO Journal 2003, 22, 2583-2592.

65. van der Werf, M.; de Bont, J.A.M.; Swarts, H.J., Tetrahedron: Asymmetry, 1999, 10, 4225-4230.

66 . Fretland, A.J.; Omiecinski, C.J. Chem. Biol. Interact. 2000, 129, 41-59. 67. Dietze, E.C.; Magdalou, J.; Hammock, B.D. Int. J. Biochem. 1990, 22, 461- 470. 68. Borhan, B.; Jones, A. D.; Pinot, F.; Grant, D.F.; Kurth, M.J.; Hammock, B.D.

J. Biol. Chem. 1995, 270, 26923-26930. 69. Müller, F.; Arand, M.; Frank, H.; Seidel, A.; Hinz, W.; Winkler, L.; Hänel, K.;

Blée, E., Beetham, J.K., Hammock, B.D.; Oesch, F. Eur. J. Biochem. 1997, 245, 490-496.

70. Hammock, B.D.; Pinot, F.; Beetham, J.K.; Grant, D.F.; Arand, M.E.; Oesch, F. Biochem. Biophys. Res. Commun. 1994, 3, 850-856.

71. Lacouciere, G.M.; Armstrong; R.N. J. Am. Chem. Soc. 1993, 115, 10466-10467.

72. Laughlin, L.T.; Tzeng, H.-F.; Lin, S.; Armstrong, R. N. Biochemistry 1998, 37, 2897-2904. 73. Arand, M.; Wagner, H.; Oesch, F. J. Biol. Chem. 1996, 271, 4223-4229.

Page 68: Quantum Chemical Studies of Epoxide- Transforming Enzymes12099/...Density functional theory is employed to study the reaction mechanisms of different epoxide-transforming enzymes

58

74. Pinot, F.; Grant, D. F.; Beetham, J. K.; Parker, A. G.; Borhan, B.; Landt, S.; Jones, A. D.; Hammock, B. D. J. Biol. Chem. 1995, 270, 7968-7974. 75. Rink, R.; Fennema, F.; Smids, M.; Dehmel, U.; Janssen, D.B. J. Biol. Chem.

1997, 272, 14650-14657. 76. Tzeng, H.-F.; Laughlin, L. T.; Armstrong, R. N. Biochemistry 1998, 37, 2905-

2911. 77. Bell, P.A.; Kasper, C.B. J. Biol. Chem. 1993, 268, 14011-14017. 78. Argiriadi, M.A.; Morisseau, C.M.; Goodrow, M. H.; Dowdy, D.L.; Hammock,

B.D.; Christianson, D.W. J. Biol. Chem. 2000, 275, 15265-15270. 79. Zou, J.; Hallberg, B.M.; Bergfors, T.; Oesch, F.; Arand, M.; Mowbray, S.L.;

Jones, T.A. Structure 2000, 8, 111-122. 80. Rink, R.; Kingma, J.; Lutje Spelberg; J.H.; Janssen, D.B. Biochemistry 2000,

39, 5600-5613. 81. Yamada, T.; Morisseau, C.; Maxwell, J.E.; Argiriadi, M.A.; Christianson,

D.W.; Hammock, B.D. J. Biol. Chem. 2000, 275, 23082-23088. 82. Mowbray, S.L.; Elfström, L.T.; Ahlgren, K.M.; Andersson, C.E.; Widersten,

M. Protein Sci. 2006, 15, 1628-1637. 83. Lacouciere, G.M; Armstrong, R.N., Chem. Res. Toxicol. 1994, 7, 121-124. 84. Elfström; L.T.; Widersten, M. Biochemistry 2006, 45, 205-212. 85. Elfström, L.T.; Widersten, M. Biochemical J. 2005, 390, 633-40. 86. Schiøtt, B.; Bruice, T. C. J. Am. Chem. Soc. 2002, 124, 14558-14570. 87. Moussou, P.; Archelas, A.; Baratti, J.; Furstoss, R. J. Mol. Catal. B 1998, 5,

213-217. 88. Weijers, C.A.G.M.; de Bont, J.A.M., J. Mol. Catal. B: Enzymatic 1999, 6, 199-

214. 89. Bellucci, G.; Chiappe, C.; Cordoni, A.; Marioni, F. Tetrahedron Lett. 1994, 35,

4219-4222. 90. Williamson, K.C.; Morisseau, C.; Maxwell, J.E.; Hammock. B.D.

Tetrahedron: Asymmetry. 2000, 11, 4451-4462. 91. Pedragosa-Moreau, S.; Archelas, A.; Furstoss, R. Bioorg. Med. Chem. 1994, 2,

609-616. 92. Mischitz, M; Mirtl, C.; Saf, R.; Faber, K. Tetrahedron: Asymmetry 1996, 7,

2041-2046. 93. Monterde, M. I.; Lombard, M.; Archelas, A.; Cronin, A.; Arand, M.; Furstoss,

R. Tetrahedron: Asymmetry 2004, 15, 2801–2805.