You have 3 free guides left 😟
Unlock your guides
You have 3 free guides left 😟
Unlock your guides

Reaction rates and mechanisms are crucial for understanding enzyme catalysis. These concepts explain how factors like , pH, and influence reaction speeds. They also describe the mathematical relationships between reactants and products, helping us predict and control enzymatic processes.

Studying reaction rates and mechanisms reveals the inner workings of enzymes. By examining steady-state and pre-steady-state kinetics, we can uncover the step-by-step processes enzymes use to catalyze reactions. This knowledge is essential for developing new drugs and improving industrial applications of enzymes.

Factors influencing reaction rates

Temperature and pH effects

Top images from around the web for Temperature and pH effects
Top images from around the web for Temperature and pH effects
  • Higher temperatures generally increase reaction rates up to an optimal temperature
    • Beyond the optimal temperature, rates decrease due to enzyme denaturation
  • pH alters the ionization state of enzymes and substrates
    • Enzymes typically have an optimal pH range for maximum activity (pepsin functions best at acidic pH in the stomach)

Concentration and molecular crowding effects

  • Substrate concentration influences reaction rates
    • Higher concentrations increase rates until enzyme saturation is reached ()
  • Enzyme concentration directly affects reaction rates
    • Higher concentrations lead to faster rates until substrate becomes limiting
  • Molecular crowding in cellular environments can impact reaction rates
    • Alters enzyme and substrate diffusion and increases effective concentrations
  • Viscosity of the cellular environment can influence reaction rates by affecting the diffusion of enzymes and substrates (cytoplasm is more viscous than water)

Inhibitors and activators

  • Presence of inhibitors can decrease reaction rates by binding to enzymes and modulating their activity
    • Competitive inhibitors bind to the active site and compete with the substrate (pepstatin A inhibits pepsin)
    • Non-competitive inhibitors bind to allosteric sites and alter enzyme conformation (phosphorylation of glycogen phosphorylase)
  • Presence of activators can increase reaction rates by binding to enzymes and modulating their activity
    • Allosteric activators bind to regulatory sites and enhance enzyme activity (fructose-2,6-bisphosphate activates phosphofructokinase)

Rate laws and reaction order

Rate law equations

  • Rate laws for enzymatic reactions describe the relationship between and reactant concentrations
    • Typically expressed as v=k[E]m[S]nv = k[E]^m[S]^n, where vv is reaction rate, kk is , [E][E] is enzyme concentration, [S][S] is substrate concentration, and mm and nn are reaction orders
  • Reaction order refers to the power to which the concentration of a reactant (enzyme or substrate) is raised in the rate law equation
    • reactions have rates independent of reactant concentrations, with a rate law of v=kv = k
    • reactions have rates directly proportional to one reactant concentration, with a rate law of v=k[A]v = k[A], where [A][A] is the concentration of the reactant
    • reactions have rates proportional to the product of two reactant concentrations or the square of one reactant concentration, with a rate law of v=k[A][B]v = k[A][B] or v=k[A]2v = k[A]^2

Michaelis-Menten kinetics and Lineweaver-Burk plots

  • Michaelis-Menten kinetics describes the rate law for many enzymatic reactions
    • Hyperbolic relationship between reaction rate and substrate concentration, characterized by the parameters VmaxV_{max} (maximum reaction rate) and KmK_m (Michaelis constant)
    • VmaxV_{max} represents the maximum rate achieved by the system at saturating substrate concentrations
    • KmK_m is the substrate concentration at which the reaction rate is half of VmaxV_{max}, and is an inverse measure of the substrate's affinity for the enzyme
  • Lineweaver-Burk plots (double-reciprocal plots) can be used to linearize Michaelis-Menten kinetics data
    • Allows for the determination of VmaxV_{max} and KmK_m values from the y-intercept and slope, respectively
    • Useful for comparing kinetic parameters of different enzymes or substrates

Reaction mechanisms and rate-determining steps

Elementary steps and rate-determining step

  • Reaction mechanisms for enzymatic reactions describe the sequence of leading from reactants to products
    • Includes the formation of enzyme-substrate complexes and any species
    • Example: The mechanism of chymotrypsin involves the formation of an acyl-enzyme intermediate
  • The (RDS) is the slowest elementary step in a reaction mechanism
    • Determines the overall reaction rate
    • The rate law for an enzymatic reaction is determined by the rate-determining step, with the reaction order for each reactant corresponding to its stoichiometric coefficient in the RDS

Steady-state approximation and kinetic isotope effects

  • Steady-state approximation assumes that the concentrations of reaction intermediates remain constant over time
    • Simplifies the kinetic analysis of complex reaction mechanisms
    • Allows for the derivation of the Michaelis-Menten equation
  • Kinetic isotope effects can be used to identify the rate-determining step
    • Compares reaction rates with isotopically labeled reactants (deuterium or 13C)
    • The RDS often involves bond-breaking or bond-forming steps that are sensitive to isotopic substitution
    • Primary kinetic isotope effects are observed when the isotopically labeled atom is directly involved in the RDS (C-H bond cleavage in the RDS of alcohol dehydrogenase)

Transient kinetic methods

  • Pre-steady-state kinetics can be used to study the formation and decay of reaction intermediates before the steady-state is reached
    • Provides insights into the elementary steps of a reaction mechanism
  • Transient kinetic methods, such as stopped-flow and rapid quench-flow techniques, can be used to study the kinetics of fast enzymatic reactions
    • Allows for the detection of short-lived reaction intermediates
    • Stopped-flow: Rapid mixing of reactants and monitoring of the reaction progress by spectroscopic methods (fluorescence or absorbance)
    • Rapid quench-flow: Rapid mixing of reactants followed by quenching of the reaction at specific time points, allowing for the isolation and characterization of intermediates

Steady-state vs Pre-steady-state kinetics

Steady-state kinetics

  • Describes the behavior of an enzymatic reaction once the concentrations of reaction intermediates (e.g., enzyme-substrate complexes) have reached a constant level
    • Typically occurs after an initial pre-steady-state phase
  • In steady-state kinetics, the rates of formation and decay of reaction intermediates are equal
    • Results in a constant concentration of intermediates over time
  • Characterized by the Michaelis-Menten equation, which relates reaction rate to substrate concentration
    • Parameters VmaxV_{max} and KmK_m describe the maximum rate and substrate affinity, respectively

Pre-steady-state kinetics

  • Describes the behavior of an enzymatic reaction during the initial phase, before the concentrations of reaction intermediates have reached a steady state
  • In pre-steady-state kinetics, the concentrations of reaction intermediates change rapidly over time
    • Intermediates are formed and consumed during the early stages of the reaction
  • Can provide information on the individual rate constants for the formation and decay of enzyme-substrate complexes and other reaction intermediates
  • Transient kinetic methods, such as stopped-flow and rapid quench-flow techniques, are used to study pre-steady-state kinetics
    • Rapidly mix reactants and monitor the time course of the reaction on millisecond to second timescales

Importance of studying both steady-state and pre-steady-state kinetics

  • Analysis of pre-steady-state kinetics can reveal the presence of multiple reaction intermediates, conformational changes in the enzyme, and other mechanistic details that are not apparent from steady-state kinetics alone
  • Combining steady-state and pre-steady-state kinetics provides a comprehensive understanding of enzyme catalysis
    • Elucidates the overall reaction mechanism, rate-determining steps, and regulatory mechanisms
    • Helps in the design of enzyme inhibitors and the engineering of enzymes with desired catalytic properties
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.
Glossary
Glossary