1
|
Chae SJ, Shin S, Lee K, Lee S, Kim JK. From homogeneity to heterogeneity: Refining stochastic simulations of gene regulation. Comput Struct Biotechnol J 2025; 27:411-422. [PMID: 39906159 PMCID: PMC11791169 DOI: 10.1016/j.csbj.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 02/06/2025] Open
Abstract
Cellular processes are intricately controlled through gene regulation, which is significantly influenced by intrinsic noise due to the small number of molecules involved. The Gillespie algorithm, a widely used stochastic simulation method, is pervasively employed to model these systems. However, this algorithm typically assumes that DNA is homogeneously distributed throughout the nucleus, which is not realistic. In this study, we evaluated whether stochastic simulations based on the assumption of spatial homogeneity can accurately capture the dynamics of gene regulation. Our findings indicate that when transcription factors diffuse slowly, these simulations fail to accurately capture gene expression, highlighting the necessity to account for spatial heterogeneity. However, incorporating spatial heterogeneity considerably increases computational time. To address this, we explored various stochastic quasi-steady-state approximations (QSSAs) that simplify the model and reduce simulation time. While both the stochastic total quasi-steady state approximation (stQSSA) and the stochastic low-state quasi-steady-state approximation (slQSSA) reduced simulation time, only the slQSSA provided an accurate model reduction. Our study underscores the importance of utilizing appropriate methods for efficient and accurate stochastic simulations of gene regulatory dynamics, especially when incorporating spatial heterogeneity.
Collapse
Affiliation(s)
- Seok Joo Chae
- Department of Mathematical Sciences, KAIST, Daejeon, 34141, Republic of Korea
- Biomedical Mathematics group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Department of Bioengineering, Rice University, Houston, 77005, TX, United States of America
| | - Seolah Shin
- Biomedical Mathematics group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Department of Applied Mathematics, Korea University, Seoul, 02841, Republic of Korea
| | - Kangmin Lee
- Department of Mathematical Sciences, KAIST, Daejeon, 34141, Republic of Korea
- Biomedical Mathematics group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Seunggyu Lee
- Biomedical Mathematics group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Department of Applied Mathematics, Korea University, Seoul, 02841, Republic of Korea
| | - Jae Kyoung Kim
- Department of Mathematical Sciences, KAIST, Daejeon, 34141, Republic of Korea
- Biomedical Mathematics group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
2
|
Simon L, Biswas A. A physiologically based pharmacokinetic model for the transdermal uptake of semivolatile organic compounds from the atmosphere and through clothing. JOURNAL OF OCCUPATIONAL AND ENVIRONMENTAL HYGIENE 2024; 21:778-787. [PMID: 39357064 DOI: 10.1080/15459624.2024.2398024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
This study focuses on the semivolatile organic compound (SVOC) absorption through clothing and the skin. SVOCs are ubiquitous in daily life, in products like personal care items, plastics, and building materials. Understanding their permeation through the skin barrier is crucial for evaluating potential health risks of complete exposure. A PBPK model was developed to comprehend the dynamic interplay between SVOCs and human skin and to estimate tissue distribution throughout the body. The framework incorporated parameters such as skin permeability, physicochemical properties of the chemicals, and the impact of protective clothing and adsorbents. This model predicted the rate and extent of SVOC absorption under diverse scenarios. The PBPK predictions matched the experimental amount of mono-ethyl phthalate (MEP), a phthalate metabolite, when urine samples were collected for bare-skinned and clothed participants. Urine concentrations of MEP during a 6-hr exposure and for the next 48 hr show that clean clothing effectively decreased dermal uptake and the buildup of chemicals in the body. Additional removal of MEP was achieved through adsorption on activated carbon fabric. An increase in the maximum monolayer adsorption capacity or the Langmuir equilibrium constant further reduced the amount of MEP in the urine.
Collapse
Affiliation(s)
- Laurent Simon
- Otto H York Department and Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | - Abishek Biswas
- Otto H York Department and Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey
| |
Collapse
|
3
|
Murugan R. Theory on the rate equations of Michaelis-Menten type enzyme kinetics with competitive inhibition. PLoS One 2024; 19:e0302679. [PMID: 39024204 PMCID: PMC11257316 DOI: 10.1371/journal.pone.0302679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/09/2024] [Indexed: 07/20/2024] Open
Abstract
We derive approximate expressions for pre- and post-steady state regimes of the velocity-substrate-inhibitor spaces of the Michaelis-Menten enzyme kinetic scheme with fully and partial competitive inhibition. Our refinement over the currently available standard quasi steady state approximation (sQSSA) seems to be valid over wide range of enzyme to substrate and enzyme to inhibitor concentration ratios. Further, we show that the enzyme-inhibitor-substrate system can exhibit temporally well-separated two different steady states with respect to both enzyme-substrate and enzyme-inhibitor complexes under certain conditions. We define the ratios fS = vmax/(KMS + e0) and fI = umax/(KMI + e0) as the acceleration factors with respect to the catalytic conversion of substrate and inhibitor into their respective products. Here KMS and KMI are the Michaelis-Menten parameters associated respectively with the binding of substrate and inhibitor with the enzyme, vmax and umax are the respective maximum reaction velocities and e0, s0, and i0 are total enzyme, substrate and inhibitor levels. When (fS/fI) < 1, then enzyme-substrate complex will show multiple steady states and it reaches the full-fledged steady state only after the depletion of enzyme-inhibitor complex. When (fS/fI) > 1, then the enzyme-inhibitor complex will show multiple steady states and it reaches the full-fledged steady state only after the depletion of enzyme-substrate complex. This multi steady-state behavior especially when (fS/fI) ≠ 1 is the root cause of large amount of error in the estimation of various kinetic parameters of fully and partial competitive inhibition schemes using sQSSA. Remarkably, we show that our refined expressions for the reaction velocities over enzyme-substrate-inhibitor space can control this error more significantly than the currently available sQSSA expressions.
Collapse
Affiliation(s)
- Rajamanickam Murugan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
4
|
Shin S, Chae SJ, Lee S, Kim JK. Beyond homogeneity: Assessing the validity of the Michaelis-Menten rate law in spatially heterogeneous environments. PLoS Comput Biol 2024; 20:e1012205. [PMID: 38843305 PMCID: PMC11185478 DOI: 10.1371/journal.pcbi.1012205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/18/2024] [Accepted: 05/24/2024] [Indexed: 06/19/2024] Open
Abstract
The Michaelis-Menten (MM) rate law has been a fundamental tool in describing enzyme-catalyzed reactions for over a century. When substrates and enzymes are homogeneously distributed, the validity of the MM rate law can be easily assessed based on relative concentrations: the substrate is in large excess over the enzyme-substrate complex. However, the applicability of this conventional criterion remains unclear when species exhibit spatial heterogeneity, a prevailing scenario in biological systems. Here, we explore the MM rate law's applicability under spatial heterogeneity by using partial differential equations. In this study, molecules diffuse very slowly, allowing them to locally reach quasi-steady states. We find that the conventional criterion for the validity of the MM rate law cannot be readily extended to heterogeneous environments solely through spatial averages of molecular concentrations. That is, even when the conventional criterion for the spatial averages is satisfied, the MM rate law fails to capture the enzyme catalytic rate under spatial heterogeneity. In contrast, a slightly modified form of the MM rate law, based on the total quasi-steady state approximation (tQSSA), is accurate. Specifically, the tQSSA-based modified form, but not the original MM rate law, accurately predicts the drug clearance via cytochrome P450 enzymes and the ultrasensitive phosphorylation in heterogeneous environments. Our findings shed light on how to simplify spatiotemporal models for enzyme-catalyzed reactions in the right context, ensuring accurate conclusions and avoiding misinterpretations in in silico simulations.
Collapse
Affiliation(s)
- Seolah Shin
- Department of Applied Mathematics, Korea University, Sejong, Republic of Korea
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seok Joo Chae
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Mathematical Sciences, KAIST, Daejeon, Republic of Korea
| | - Seunggyu Lee
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, Republic of Korea
- Division of Applied Mathematical Sciences, Korea University, Sejong, Republic of Korea
| | - Jae Kyoung Kim
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Mathematical Sciences, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
5
|
Eilertsen J, Schnell S, Walcher S. The Michaelis-Menten Reaction at Low Substrate Concentrations: Pseudo-First-Order Kinetics and Conditions for Timescale Separation. Bull Math Biol 2024; 86:68. [PMID: 38703247 PMCID: PMC11069484 DOI: 10.1007/s11538-024-01295-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
We demonstrate that the Michaelis-Menten reaction mechanism can be accurately approximated by a linear system when the initial substrate concentration is low. This leads to pseudo-first-order kinetics, simplifying mathematical calculations and experimental analysis. Our proof utilizes a monotonicity property of the system and Kamke's comparison theorem. This linear approximation yields a closed-form solution, enabling accurate modeling and estimation of reaction rate constants even without timescale separation. Building on prior work, we establish that the sufficient condition for the validity of this approximation iss 0 ≪ K , where K = k 2 / k 1 is the Van Slyke-Cullen constant. This condition is independent of the initial enzyme concentration. Further, we investigate timescale separation within the linear system, identifying necessary and sufficient conditions and deriving the corresponding reduced one-dimensional equations.
Collapse
Affiliation(s)
- Justin Eilertsen
- Mathematical Reviews, American Mathematical Society, 416 4th Street, Ann Arbor, MI, 48103, USA
| | - Santiago Schnell
- Department of Biological Sciences and Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | | |
Collapse
|
6
|
Eilertsen J, Schnell S, Walcher S. The unreasonable effectiveness of the total quasi-steady state approximation, and its limitations. J Theor Biol 2024; 583:111770. [PMID: 38423205 DOI: 10.1016/j.jtbi.2024.111770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
In this note, we discuss the range of parameters for which the total quasi-steady-state approximation of the Michaelis-Menten reaction mechanism holds validity. We challenge the prevalent notion that total quasi-steady-state approximation is "roughly valid" across all parameters, showing that its validity cannot be assumed, even roughly, across the entire parameter space - when the initial substrate concentration is high. On the positive side, we show that the linearized one-dimensional equation for total substrate is a valid approximation when the initial reduced substrate concentration s0/KM is small. Moreover, we obtain a precise picture of the long-term time course of both substrate and complex.
Collapse
Affiliation(s)
- Justin Eilertsen
- Mathematical Reviews, American Mathematical Society, 416 4th Street Ann Arbor, MI, 48103, United States of America.
| | - Santiago Schnell
- Department of Biological Sciences and Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, United States of America.
| | | |
Collapse
|
7
|
Byun JH, Jeon HS, Yun HY, Kim JK. Validity conditions of approximations for a target-mediated drug disposition model: A novel first-order approximation and its comparison to other approximations. PLoS Comput Biol 2024; 20:e1012066. [PMID: 38656966 PMCID: PMC11090311 DOI: 10.1371/journal.pcbi.1012066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 05/13/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Target-mediated drug disposition (TMDD) is a phenomenon characterized by a drug's high-affinity binding to a target molecule, which significantly influences its pharmacokinetic profile within an organism. The comprehensive TMDD model delineates this interaction, yet it may become overly complex and computationally demanding in the absence of specific concentration data for the target or its complexes. Consequently, simplified TMDD models employing quasi-steady state approximations (QSSAs) have been introduced; however, the precise conditions under which these models yield accurate results require further elucidation. Here, we establish the validity of three simplified TMDD models: the Michaelis-Menten model reduced with the standard QSSA (mTMDD), the QSS model reduced with the total QSSA (qTMDD), and a first-order approximation of the total QSSA (pTMDD). Specifically, we find that mTMDD is applicable only when initial drug concentrations substantially exceed total target concentrations, while qTMDD can be used for all drug concentrations. Notably, pTMDD offers a simpler and faster alternative to qTMDD, with broader applicability than mTMDD. These findings are confirmed with antibody-drug conjugate real-world data. Our findings provide a framework for selecting appropriate simplified TMDD models while ensuring accuracy, potentially enhancing drug development and facilitating safer, more personalized treatments.
Collapse
Affiliation(s)
- Jong Hyuk Byun
- Department of Mathematics and Institute of Mathematical Science, Pusan National University, Busan, Republic of Korea
- Institute for Future Earth, Pusan National University, Busan, Republic of Korea
| | - Hye Seon Jeon
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Hwi-yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Kyoung Kim
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Mathematical Sciences, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
8
|
Lim R, Martin TLP, Chae J, Kim WJ, Ghim CM, Kim PJ. Generalized Michaelis-Menten rate law with time-varying molecular concentrations. PLoS Comput Biol 2023; 19:e1011711. [PMID: 38079453 PMCID: PMC10735182 DOI: 10.1371/journal.pcbi.1011711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 12/21/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
The Michaelis-Menten (MM) rate law has been the dominant paradigm of modeling biochemical rate processes for over a century with applications in biochemistry, biophysics, cell biology, systems biology, and chemical engineering. The MM rate law and its remedied form stand on the assumption that the concentration of the complex of interacting molecules, at each moment, approaches an equilibrium (quasi-steady state) much faster than the molecular concentrations change. Yet, this assumption is not always justified. Here, we relax this quasi-steady state requirement and propose the generalized MM rate law for the interactions of molecules with active concentration changes over time. Our approach for time-varying molecular concentrations, termed the effective time-delay scheme (ETS), is based on rigorously estimated time-delay effects in molecular complex formation. With particularly marked improvements in protein-protein and protein-DNA interaction modeling, the ETS provides an analytical framework to interpret and predict rich transient or rhythmic dynamics (such as autogenously-regulated cellular adaptation and circadian protein turnover), which goes beyond the quasi-steady state assumption.
Collapse
Affiliation(s)
- Roktaek Lim
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong
- Department of Physics, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | | | - Junghun Chae
- Department of Physics, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Woo Joong Kim
- Department of Physics, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Cheol-Min Ghim
- Department of Physics, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Pan-Jun Kim
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong
- Center for Quantitative Systems Biology & Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Kowloon, Hong Kong
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Kowloon, Hong Kong
- Abdus Salam International Centre for Theoretical Physics, Trieste, Italy
| |
Collapse
|
9
|
Vu NAT, Song YM, Tran QT, Yun HY, Kim SK, Chae JW, Kim JK. Beyond the Michaelis-Menten: Accurate Prediction of Drug Interactions through Cytochrome P450 3A4 Induction. Clin Pharmacol Ther 2022; 113:1048-1057. [PMID: 36519932 DOI: 10.1002/cpt.2824] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
The US Food and Drug Administration (FDA) guidance has recommended several model-based predictions to determine potential drug-drug interactions (DDIs) mediated by cytochrome P450 (CYP) induction. In particular, the ratio of substrate area under the plasma concentration-time curve (AUCR) under and not under the effect of inducers is predicted by the Michaelis-Menten (MM) model, where the MM constant ( K m $$ {K}_{\mathrm{m}} $$ ) of a drug is implicitly assumed to be sufficiently higher than the concentration of CYP enzymes that metabolize the drug ( E T $$ {E}_{\mathrm{T}} $$ ) in both the liver and small intestine. Furthermore, the fraction absorbed from gut lumen ( F a $$ {F}_{\mathrm{a}} $$ ) is also assumed to be one because F a $$ {F}_{\mathrm{a}} $$ is usually unknown. Here, we found that such assumptions lead to serious errors in predictions of AUCR. To resolve this, we propose a new framework to predict AUCR. Specifically, F a $$ {F}_{\mathrm{a}} $$ was re-estimated from experimental permeability values rather than assuming it to be one. Importantly, we used the total quasi-steady-state approximation to derive a new equation, which is valid regardless of the relationship between K m $$ {K}_{\mathrm{m}} $$ and E T $$ {E}_{\mathrm{T}} $$ , unlike the MM model. Thus, our framework becomes much more accurate than the original FDA equation, especially for drugs with high affinities, such as midazolam or strong inducers, such as rifampicin, so that the ratio between K m $$ {K}_{\mathrm{m}} $$ and E T $$ {E}_{\mathrm{T}} $$ becomes low (i.e., the MM model is invalid). Our work greatly improves the prediction of clinical DDIs, which is critical to preventing drug toxicity and failure.
Collapse
Affiliation(s)
- Ngoc-Anh Thi Vu
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Yun Min Song
- Department of Mathematical Sciences, KAIST, Daejeon, Korea.,Biomedical Mathematics Group, Institute for Basic Science, Daejeon, Korea
| | - Quyen Thi Tran
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Korea.,Department of Bio-AI convergence, Chungnam National University, Daejeon, Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Jung-Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Korea.,Department of Bio-AI convergence, Chungnam National University, Daejeon, Korea
| | - Jae Kyoung Kim
- Department of Mathematical Sciences, KAIST, Daejeon, Korea.,Biomedical Mathematics Group, Institute for Basic Science, Daejeon, Korea
| |
Collapse
|
10
|
Atypical kinetics of cytochrome P450 enzymes in pharmacology and toxicology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:131-176. [PMID: 35953154 DOI: 10.1016/bs.apha.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atypical kinetics are observed in metabolic reactions catalyzed by cytochrome P450 enzymes (P450). Yet, this phenomenon is regarded as experimental artifacts in some instances despite increasing evidence challenging the assumptions of typical Michaelis-Menten kinetics. As P450 play a major role in the metabolism of a wide range of substrates including drugs and endogenous compounds, it becomes critical to consider the impact of atypical kinetics on the accuracy of estimated kinetic and inhibitory parameters which could affect extrapolation of pharmacological and toxicological implications. The first half of this book chapter will focus on atypical non-Michaelis-Menten kinetics (e.g. substrate inhibition, biphasic and sigmoidal kinetics) as well as proposed underlying mechanisms supported by recent insights in mechanistic enzymology. In particular, substrate inhibition kinetics in P450 as well as concurrent drug inhibition of P450 in the presence of substrate inhibition will be further discussed. Moreover, mounting evidence has revealed that despite the high degree of sequence homology between CYP3A isoforms (i.e. CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different susceptibilities and potencies of mechanism-based inactivation (MBI) with a common drug inhibitor. These experimental observations pertaining to the presence of these atypical isoform- and probe substrate-specific complexities in CYP3A isoforms by several clinically-relevant drugs will therefore be expounded and elaborated upon in the second half of this book chapter.
Collapse
|
11
|
Srinivasan B. Explicit Treatment of Non-Michaelis-Menten and Atypical Kinetics in Early Drug Discovery*. ChemMedChem 2020; 16:899-918. [PMID: 33231926 DOI: 10.1002/cmdc.202000791] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 12/27/2022]
Abstract
Biological systems are highly regulated. They are also highly resistant to sudden perturbations enabling them to maintain the dynamic equilibrium essential to sustain life. This robustness is conferred by regulatory mechanisms that influence the activity of enzymes/proteins within their cellular context to adapt to changing environmental conditions. However, the initial rules governing the study of enzyme kinetics were mostly tested and implemented for cytosolic enzyme systems that were easy to isolate and/or recombinantly express. Moreover, these enzymes lacked complex regulatory modalities. Now, with academic labs and pharmaceutical companies turning their attention to more-complex systems (for instance, multiprotein complexes, oligomeric assemblies, membrane proteins and post-translationally modified proteins), the initial axioms defined by Michaelis-Menten (MM) kinetics are rendered inadequate, and the development of a new kind of kinetic analysis to study these systems is required. This review strives to present an overview of enzyme kinetic mechanisms that are atypical and, oftentimes, do not conform to the classical MM kinetics. Further, it presents initial ideas on the design and analysis of experiments in early drug-discovery for such systems, to enable effective screening and characterisation of small-molecule inhibitors with desirable physiological outcomes.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Mechanistic Biology and Profiling Discovery Sciences, R&D, AstraZeneca, 310, Milton Rd, Milton CB4 0WG, Cambridge, UK
| |
Collapse
|
12
|
Kim JK, Tyson JJ. Misuse of the Michaelis-Menten rate law for protein interaction networks and its remedy. PLoS Comput Biol 2020; 16:e1008258. [PMID: 33090989 PMCID: PMC7581366 DOI: 10.1371/journal.pcbi.1008258] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
For over a century, the Michaelis-Menten (MM) rate law has been used to describe the rates of enzyme-catalyzed reactions and gene expression. Despite the ubiquity of the MM rate law, it accurately captures the dynamics of underlying biochemical reactions only so long as it is applied under the right condition, namely, that the substrate is in large excess over the enzyme-substrate complex. Unfortunately, in circumstances where its validity condition is not satisfied, especially so in protein interaction networks, the MM rate law has frequently been misused. In this review, we illustrate how inappropriate use of the MM rate law distorts the dynamics of the system, provides mistaken estimates of parameter values, and makes false predictions of dynamical features such as ultrasensitivity, bistability, and oscillations. We describe how these problems can be resolved with a slightly modified form of the MM rate law, based on the total quasi-steady state approximation (tQSSA). Furthermore, we show that the tQSSA can be used for accurate stochastic simulations at a lower computational cost than using the full set of mass-action rate laws. This review describes how to use quasi-steady state approximations in the right context, to prevent drawing erroneous conclusions from in silico simulations.
Collapse
Affiliation(s)
- Jae Kyoung Kim
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - John J. Tyson
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
- Division of Systems Biology, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|