03.01.2014 Views

Computer Simulation Methods 3

Computer Simulation Methods 3

Computer Simulation Methods 3

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Computer</strong> simulation<br />

methods<br />

(3)<br />

Dr. Vania Calandrini


in the previous lecture:<br />

setting up a simulation<br />

boundaries<br />

monitoring the equilibration<br />

short-range and long-range interactions<br />

minimum image, cutoff and related problems/solutions<br />

Ewald summation<br />

Particle Mesh Ewald summation


Molecular dynamics<br />

Classical Molecular<br />

Dynamics<br />

technique for computing the equilibrium and transport properties of a<br />

classical many-body system.<br />

the nuclear motion of the constituents particles obeys the laws<br />

of classical mechanics (hυ υ< 6ps -1 at 300 K )<br />

These vibrations are not well treated by<br />

classical physics. They require quantum<br />

mechanics simulations.<br />

These data are used as input in molecular<br />

dynamics simulations to parametrize the<br />

characteristic frequencies of the potential.


In molecular dynamics, successive configurations are generated by integrating Newtonʼs<br />

laws of motion. The result is a trajectory that specifies how the positions and velocities of<br />

the particles in the system vary with time.<br />

1) a body continues to move in a straight line at constant velocity unless a force acts upon it<br />

2) force equals the rate of change of momentum<br />

3) to every action there is an equal and opposite reaction<br />

The trajectory is obtained by solving the differential equations embodied in Newtonʼs second<br />

law:<br />

m i¨r i ≡ ṗ i = − ∂V =Fi = F i<br />

∂r i<br />

ṙ i = p i<br />

m i<br />

conservative system :<br />

each atom is represented<br />

by a mass point<br />

ri<br />

V = V (r 1 , r 2 ,...,r n )


Time and Spatial scales<br />

MD simulations<br />

(atomic details):<br />

Biological<br />

Systems:<br />

~ 10 -12 to 10 -7 s<br />

~ 10 -10 to 10 -8 m<br />

~ 100 000 particles


Molecular dynamics with continuous<br />

Potentials<br />

Under the influence of a continuous potential the motions of all the particles are coupled<br />

together, giving rise to a many body problem that cannot be solved analytically=> then the<br />

equations of motion are integrated numerically<br />

Numerical integration of motion equations : Finite difference methods<br />

the integration is broken down into many small stages, each separated in time by a fixed δt: forces acting on<br />

each particle at time t are computed knowing the positions of all the particles. Then the accelerations together<br />

with the positions and velocities at time t are used to compute the new positions and velocities at time t+δt.<br />

During each time step the force is assumed to be constant.<br />

Taylor series<br />

expansions:<br />

Any finite difference integrator is naturally an approximation for a system developing<br />

continuously in time.


The integrator is responsible for the accuracy of the simulation results.<br />

Main requirements:<br />

• Stable: it has to conserve energy<br />

• Robust, in the sense that it allows for large time steps in order to propagate the system<br />

efficiently through phase space


Verlet algorithm :<br />

adding up (1) and (2):<br />

(1)<br />

(2)<br />

Taylor’s<br />

expansions<br />

Δt→0<br />

or:<br />

next current previous<br />

requires the storage of old and current positions<br />

subtracting (2) from (1) one can derive the velocity from the knowledge of the trajectory:<br />

or:


Verlet algorithm :<br />

requires the storage of old and current positions<br />

the velocities are not available until the positions have been computed at the next step<br />

it is not a self starting algorithm: at t=0 you have only one set of positions. To obtain the<br />

positions et t=-δt, one can use eq. 2 truncated after the first term:<br />

low precision<br />

time reversible = reversing the momenta of all particles at a given instant, the system<br />

trace back its trajectory in phase space


Leap frog algorithm :<br />

positions and velocities not synchronized


velocity Verlet algorithm :<br />

(1)<br />

v(t + ∆t<br />

2 )=v(t)+∆t 2<br />

f(t)<br />

m + O(∆t2 )<br />

Δt/2→0<br />

Taylor’s expansion<br />

(2)<br />

r(t + ∆t) =r(t)+v(t + ∆t<br />

2 )∆t + O(∆t2 )<br />

from the mean<br />

value theorem or<br />

central difference<br />

(3)<br />

v(t + ∆t) =v(t + ∆t<br />

2 )+∆t 2<br />

f(t + ∆t)<br />

m<br />

f(t + ∆t) =−<br />

- Velocity Verlet requires only the storage of current positions.<br />

- Verlet and velocity Verlet schemes produce exactly the same trajectory<br />

approximation of derivatives by finite differences :<br />

forward difference<br />

backward difference<br />

central difference<br />

δV (r(t + ∆t))<br />

δr<br />

+ O(∆t)<br />

from the<br />

backward<br />

difference


High-order schemes: predictor-correctors<br />

The basic idea is to use r(t) and its first n derivatives at time t to make a prediction for<br />

r(t+ Δt) and its first n derivatives at time t+ Δt. We then compute the forces (i.e.<br />

accelerations) at the predicted positions and find that these forces are different from<br />

the predicted ones. So we adjust our predictions for the accelerations to match the<br />

facts. Moreover, on the basis of the observed discrepancy between the predicted and<br />

observed accelerations, we also try to improve our estimate of the positions and the<br />

remaining n−1 derivatives. the “recipe” used in applying this correction is a<br />

compromise between accuracy and stability.<br />

see Understanding Molecular <strong>Simulation</strong> by Daan Frenkel<br />

and Berend Smit for some specific examples of a predictor<br />

corrector algorithm.


Comments:<br />

• Although, computational speed seems important, it is usually not very relevant, because<br />

the fraction of time spent on integrating is usually small (as opposed to computing<br />

interactions) at least for simple molecular system.<br />

• Accuracy: we can consider the integration error as the source of the divergence between<br />

the “true” trajectory and the computed trajectory compatible with the same initial conditions<br />

(note that this problem need not to be serious in molecular dynamics (MD) simulations<br />

since the aim of an MD simulation is to predict the average behavior and not what will<br />

precisely happen to a system in a known initial condition. It has been proven that MD<br />

simulations provide good statistical predictions for many systems.)<br />

• Algorithms that allow the use of large time steps require the storage of increasingly higher<br />

order derivatives.<br />

• Energy conservation: higher order algorithms tend to have very good energy<br />

conservation for short times, while the overall energy drifts for long times. In contrast Verlet<br />

algorithms tend to have only moderate short term energy conservation but little long-term<br />

drift.<br />

•Time reversibility (= reversing the momenta of all particles at a given instant, the system<br />

trace back its trajectory in phase space): many algorithms such as predictor-correctors are<br />

not time reversible.Verlet scheme is time reversible.


Tutorials


Required programs:<br />

NAMD<br />

VMD<br />

a parallel molecular dynamics code designed for high-performance simulation of<br />

large biomolecular systems.<br />

VMD is a molecular visualization program for displaying, animating, and analyzing<br />

large biomolecular systems using 3-D graphics and built-in scripting. It can be used<br />

to setup simulations and analyze trajectories .<br />

by the Theoretical and Computational Biophysics Group at the Beckman Institute for<br />

Advanced Science and Technology of the University of Illinois at Urbana-Champaign<br />

you need also a text editor and a plotting program


Tutorial<br />

http://www.ks.uiuc.edu/Training/Tutorials/namd-index.html<br />

NAMD Tutorial<br />

the examples in the tutorial will focus on the study of a small protein, ubiquitin<br />

purpose:<br />

- basic steps of a molecular dynamics simulation, i.e., preparation, minimization, and<br />

equilibration of your system.<br />

- typical simulation techniques and analysis of equilibrium properties<br />

- descriptions of all files needed for the simulations


To do:<br />

do this analysis for<br />

water box


careful<br />

reading


in this lecture:<br />

Molecular dynamics with continuous Potentials:<br />

time and spatial scales<br />

finite difference methods:<br />

Verlet algorithm<br />

velocity Verlet algorithm<br />

Leap frog algorithm<br />

Tutorial (using NAMD and VMD):<br />

basic steps of a molecular dynamics simulation, i.e., preparation, minimization, and<br />

equilibration of the system and analysis of equilibrium properties


for any question:<br />

v.calandrini@grs-sim.de

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!