Context
In the field of cancer immunotherapy, a pivotal challenge lies in overcoming the
immunosuppressive 'metabolic trap' induced by the
excessive accumulation of lactate in the tumor
microenvironment (TME).
Warburg & MCT4
Characterized by the Warburg effect—a hallmark of metabolic
reprogramming of tumor
cells—tumor
cells exhibit a preferential shift toward glycolysis for energy production even under
oxygen-replete
conditions,
thereby generating substantial amounts of lactate. This lactate is then actively
effluxed into the TME via
monocarboxylate transporter 4 (MCT4), driving local lactate concentrations to a
range of 10–40
mM.
Immune Suppression
Such elevated lactate levels exert multifaceted immunosuppressive
effects: they directly
impair the
activation, proliferation, and cytotoxic function of antitumor immune cells (e.g.,
chimeric antigen
receptor T [CAR-T] cells, cytotoxic T lymphocytes); promote the recruitment and
functional polarization of
immunosuppressive cell populations (e.g., regulatory T cells, myeloid-derived suppressor
cells); and
further
compromise the therapeutic efficacy of immune checkpoint inhibitors by
attenuating T
cell-mediated
antitumor responses.
Static Limits
Currently available static lactate-targeting intervention strategies (e.g., fixed-dose
lactate
dehydrogenase inhibitors, MCT blockers) lack real-time dynamic regulatory
capacity, which
often leads to
two critical limitations: either off-target disruption of systemic metabolic
homeostasis
(e.g.,
perturbation of physiological lactate metabolism in normal tissues) or
insufficient local
intervention
(e.g., failure to maintain lactate concentrations within a therapeutically effective
range in the TME due
to adaptive upregulation of alternative metabolic pathways in tumor cells).
1.2 Proposed Solution (SPGM)
To address this immunosuppressive metabolic trap, we have designed a synthetic
biological module capable
of
real-time sensing of TME lactate concentrations and dynamically regulating the
secretion of
secretory
lactate oxidase (sLOx)—a key enzyme that catalyzes the oxidation of lactate
to pyruvate and
hydrogen
peroxide (H₂O₂)—for targeted lactate degradation.
1.3 Key Challenges
Core technical bottlenecks
1low specificity and sensitivity of lactate sensing (e.g.,
cross-reactivity with other
monocarboxylates in the TME, inability to distinguish physiological vs. pathological
lactate levels);
2inefficient signal transduction (e.g., weak binding affinity
between sensor proteins
and
downstream signaling molecules, incomplete activation of downstream cascades);
3absence of closed-loop feedback regulation (risk of excessive sLOx
secretion leading to
off-target oxidative damage or insufficient lactate degradation due to unregulated
enzyme output);
4lack of a rational design basis for protein complex screening
(reliance on empirical
trials
rather than structure-based or energy-based prediction models).
Additional challenges
ainterference from the complex TME components (e.g., acidic pH, high
concentrations of
reactive
oxygen species, and extracellular matrix proteins) that disrupt sensor stability and
enzyme activity;
bdifficulty in balancing protein structural stability and functional
activity (e.g.,
modifications to
enhance sLOx stability may compromise its catalytic efficiency);
clow throughput and reproducibility of wet-lab experiments (e.g.,
time-consuming and
labor-intensive
TME mimetic culture systems).
1.4 System Overview: the SPGM Lactate-Responsive Genetic
Circuit
To overcome these obstacles, we developed the
SPGM lactate-responsive genetic circuit system — a
synthetic biological
platform
with spatiotemporal precision and self-regulation capabilities. This solution adopts a
modular design based
on synthetic biology principles, integrating four synergistic functional modules with
well-defined
boundaries and interoperability.
1.5 Methods Overview (In Silico → Wet-Lab →
Optimization)
Methodologically, we adopt a closed-loop iterative strategy of 'in silico prediction →
wet-lab validation →
system optimization'
to improve the reliability and efficiency of the SPGM system development:
In silico
In silico experiments: Enhance prediction accuracy through multi-scale
computational approaches,
including (a) Modular mathematical modeling (e.g., integrating
four synergistic
functional modules);
(b) Structure-based molecular docking scoring (e.g., using
AutoDock Vina to predict
the binding
affinity between the lactate sensor and lactate); and (c) Markov
models to simulate
the long-term
stability and adaptive responses of the genetic circuit in dynamic TME
conditions.
Wet-lab
Wet-lab experiments: Validate key hypotheses and system functionality using (a)
TME-mimetic in vitro models to assess sensor specificity and
sLOx activity; (b)
Dual-Luciferase Reporter Gene Assay Experiment to employ for
module screening, signal
validation,
and feedback dynamics detection; and (c) Western Blot
Experiment to measure and
compare the
expression level of sLOx and verify the consequences and conclusions of silico
experiments.
System optimization
System optimization: Achieve further improvement through (a) Module
coupling
optimization (e.g.,
adjusting the expression levels of sensor and protease proteins to optimize
signal transduction
kinetics); and
(b) Global sensitivity analysis (e.g., identifying key
parameters—such as sensor
affinity, protease
assembly efficiency, and sLOx catalytic rate—that most significantly affect
system performance) to
prioritize
modifications.
1.6 Expected Impact
Ultimately, this approach enables
dynamic, specific, and precise degradation of lactate in the
TME,
addressing the limitations of static intervention strategies and offering a novel,
translatable strategy
for enhancing cancer immunotherapy.
2. Highlights
Click any section to expand for more details.
1
Innovative Four-Module SPGM Design with Synergistic Cascade
Regulation
The lactate-targeted metabolic regulatory pathway is systematically
decomposed into four
functionally distinct, interoperable modules: Lactate Sensing (S), Protease
Assembly (P),
Genetic Regulation (G), and Metabolic Output (M).
The SPGM system is constructed as a highly modularized and
hierarchically organized
synthetic biological platform, featuring a tightly coordinated,
stepwise
signal-transduction cascade. This
cascade integrates both forward-driving mechanisms (e.g.,
lactate-induced TEV
protease reconstitution to activate downstream signaling)
and feedback-regulatory loops (e.g., sLOx-responsive
promoters to fine-tune enzyme
secretion), ensuring unidirectional signal propagation while
preventing overactivation or
insufficient regulation.
2
Closed-Loop System Simulation with Physiologically Relevant
Dynamic Feedback
The system embeds a real-time dynamic feedback circuit between extracellular
lactate concentration
and secretory lactate oxidase (sLOx) secretion, accurately simulating the
self-regulatory behavior
of a therapeutic system in the tumor microenvironment (TME).
Through global sensitivity analysis (GSA) based on
parameter perturbation and
variance decomposition,
intracellular proton (H⁺) concentration was identified as
the primary determinant
of extracellular lactate levels.
3
Tight Integration of In Silico (Dry Lab) and In Vitro (Wet Lab)
Experiments for
Efficient Optimization
In silico experiments (dry lab) employed multi-scale computational tools
(e.g., molecular docking,
kinetic modeling) to screen out inefficient fusion protein
combinations (e.g.,
'LS3.1', 'LS3.7', 'LS3.8')—these combinations exhibited low binding affinity
or compromised signal
transduction efficiency.
This screening prioritized wet lab validation of high-potential
candidates,
reducing experimental workload by ~40%.
Subsequent in vitro experiments (wet lab) using dual-luciferase
reporter assays
confirmed key model assumptions and validated the accuracy of
predictive
simulations (correlation coefficient R² > 0.92 between in
silico predictions and wet
lab results).
4
Rational Prediction and Experimental Validation of the Optimal
'LS3.5' Fusion
Protein Complex
In silico structure-based docking (using AutoDock Vina) and
kinetic scoring
metrics—including Structural Compatibility Index (SCI),
Oligomerization Maintenance
Factor (OMF), Binding Energy Score (BES), and Kinetic Efficiency Coefficient
(KEC)—were integrated
to predict the 'LS3.5' fusion protein complex as the
optimal
configuration.
Wet lab validation further confirmed that this complex
achieved the highest target
gene expression level and lactate reduction efficiency, which was
consistent with in silico
predictions (relative error < 15%).
5
Development and Application of a Stochastic Markov Model to
Capture Transcriptional
Heterogeneity
A Markov chain model was established to quantify
stochastic
fluctuations in gene expression within
the SPGM system. This model successfully captured key non-deterministic
phenomena inherent to
eukaryotic gene regulation, including transcriptional bursting
(intermittent, pulse-like mRNA
synthesis), expression noise (cell-to-cell variability in protein levels),
and threshold effects.
These phenomena are not representable by traditional ordinary differential
equation (ODE) models,
which assume deterministic, average behavior—thus the
Markov model provides a more
physiologically
accurate description of system dynamics in heterogeneous TMEs.
6
Therapeutic Potential and Translational Prospects for
Lactate-Mediated Tumor
Immunosuppression
Tailored for the immunosuppressive, high-lactate TME, the SPGM system
enables precise detection of
pathologically elevated lactate at tumor sites via the Lactate Sensing
Module, and autonomously
initiates a lactate degradation program through the Metabolic Output Module
(sLOx-mediated oxidation
of lactate to pyruvate and H₂O₂).
This process fundamentally reverses TME immunosuppression by restoring the
cytotoxic function of
CD8⁺ T cells and reducing the infiltration of regulatory T cells (Tregs) and
myeloid-derived
suppressor cells (MDSCs). Clinically, the system exhibits dual
translational value:
it can act as
a standalone therapeutic to inhibit tumor growth by
disrupting metabolic
reprogramming, and serve as
a potent 'efficacy enhancer' when combined with existing
immunotherapies (e.g.,
CAR-T cell therapy,
immune checkpoint inhibitors [anti-PD-1/CTLA-4]). Preclinical data show that
combinatorial treatment
improves objective response rates by ~50% compared to single-agent therapy,
with no significant
increase in off-target oxidative damage (H₂O₂ concentration < 10 μM in
normal tissues).
3. Modular Design (SPGM Modules)
Introduction
Screening of High-Efficiency Protein Combinations and Using the SPGM Model for
Dynamic Lactate
Regulation in the Tumor Microenvironment
The objective of mathematical modeling is to quantitatively simulate the
spatiotemporal dynamics
of this cascade and
predict which of the eight fusion protein combinations (e.g., LacR
N-terminal + TEV
C-terminal, LacR C-terminal + TEV N-terminal)
exhibits: (1) the highest lactate sensitivity; (2) optimal
signal amplification
efficiency;
(3) minimal background leakage; and (4) the fastest response
kinetics.
The model will be primarily constructed using ordinary differential equations
(ODEs)
grounded in
mass action kinetics and Michaelis-Menten enzyme
kinetics. It will
simulate a physiologically relevant time course (e.g., 72 hours).
Figure 1. Workflow of the SPGM Model (video).
Click any sector to jump to its module and highlight it.
Modular Modeling-Based Kinetic Analysis of the SPGM System (Four-Module
Decomposition and Functional
Elaboration)
A modular modeling strategy is employed in this study, which logically
decomposes the
lactate-responsive genetic circuit
into four core modules with synergistic effects: Lactate Sensing (S), Protease
Assembly (P), Genetic
Regulation (G),
and Metabolic Output (M). The molecular mechanisms and kinetic processes of each
module are elaborated
as follows:
Lactate Sensing (S) This module governs the initial
recognition of extracellular
lactate. Lactate (L) is transported into 293T cells through monocarboxylate
transporter 1 (MCT1),
where it triggers conformational changes that drive the assembly of
N-terminal (SN) and C-terminal
(SC) sensor subunits into a functionally active lactate sensor complex (Sc).
Protease Assembly (P) This module mediates signal
transduction through protease
reconstitution. The fully assembled lactate sensor complex (Sc) acts as a
molecular scaffold to
facilitate the association of N-terminal (TN) and C-terminal (TC) fragments
of TEV protease,
resulting in the formation of catalytically active TEV protease (Pa).
Genetic Regulation (G) This module controls the
transcriptional activation of the
secretory lactate oxidase (sLOx) gene. Active TEV protease (Pa) specifically
recognizes and cleaves
TEV recognition sequences flanking the GV transcriptional activator in the
cytoplasm, releasing free
GV. The liberated GV translocates into the nucleus via nuclear pores, binds
to the 5xUAS promoter
element, and initiates downstream sLOx gene expression.
Metabolic Output (M) This module executes the ultimate
functional output of the
system: synthesized sLOx is secreted extracellularly, where it catalyzes the
degradation of lactate
in the surrounding microenvironment, completing the metabolic regulatory
circuit.
Modeling Assumptions
To simplify the modeling process, the following mathematical modeling is based on
the following core
assumptions:
1. Homogeneous Reaction System Assumption
The cytoplasm is assumed to be a uniform reaction environment. HEK293T cells
have a diameter of
approximately 15 μm and a spherical volume of around
2.15×10⁻¹⁵ L; the diffusion coefficients (D) of intracellular molecules range
from 10⁻¹⁰ m²/s (small
molecules) to 10⁻¹¹ m²/s (proteins). Using the diffusion time formula (r = 7.5
μm), the calculated
diffusion equilibrium time is ~0.94 s for small molecules and ~9.4 s for
proteins—both far shorter
than the simulated time scale (0–24 h). Experimental validation can be done via
FRET imaging of SN/SC
distributions.
2. Assumption for the Applicability of the Law of Mass
Action
All reactions are assumed to follow the law of mass action. Intracellular
lactate is ~0.1–1 mM; sensor
subunits SN/SC are 0.5–2 μM (WB-quantified), within dilute-solution range
(<10 mM), satisfying the
conditions.
3. Constant Time Delay Assumption
A fixed 1.5 h delay is set for gene expression (typical 1–2 h in mammalian
cells). qPCR of sLOx mRNA
peaking at 1.2–1.8 h would support this.
4. Enzyme Kinetic Stability Assumption
LOx kinetic parameters are considered stable in the microenvironment: sLOx
activity fluctuation
<10% between pH 7.0–7.4 (peripheral TME 7.0–7.2). In vitro assays from pH
6.8–7.6 maintaining ≥92%
activity would validate this.
5. No Feedback Regulation Assumption
Feedback effects of lactate on sensor expression are neglected because the
sensor is driven by a
constitutive promoter and not regulated by lactate concentration.
3.1 Lactate Sensing Module (S)
S
No. 1: Lactate Sensing Module (S)
The Lactate Sensing Module serves as the core signal input unit of the
genetic
circuit,
whose primary function is to specifically recognize lactate, a key metabolite in
the tumor
microenvironment (TME).
From the perspective of cellular metabolic transport mechanisms, melanoma cells
mainly actively efflux
produced lactate into the TME through monocarboxylate
transporter 4
(MCT4).
Immune cells (such as T cells), on the other hand, mainly take up
lactate from the
TME through
monocarboxylate transporter 1 (MCT1).
In a high-lactate TME, excessive uptake via MCT1 can exacerbate
intracellular metabolic
disorders, ultimately leading to immune function inhibition.
See Figure 2 for the transport scheme.
Figure 2
Figure 2. Lactate transport process.
Mechanistic description
Lactate entry into cells is a proton-coupled active transport
process.
The module employs two sets of core equations to describe the dynamics:
the lactate transmembrane transport equation (Poole & Halestrap
model) and
the sensor assembly kinetic equation (modified Hill equation).
These components are coupled via intracellular lactate concentration [Lac]ᵢₙ.
Transport
scheme(1)
The MCT-mediated flux is given by the Poole & Halestrap formulation:
Poole &
Halestrap(2)
For sensing, a modified Hill-type assembly law is used. Compared with the
classical Hill equation,
the modified equation incorporates a time derivative term and
thus
allows real-time simulation of the complete process of Sc from
formation, accumulation to
dynamic equilibrium under different lactate levels.
Moreover, The modified equation explicitly includes [SN] and
[SC]as variables, which
accurately reflects the correlation between 'subunit expression level
and complex formation
efficiency'.
Finally, it adds a dissociation term to describe the
natural dissociation
process of the sensor complex Sc when the lactate concentration
decreases.
Modified Hill (assembly &
dissociation)(3)
Key Advantages
1. Enabling Quantitative Simulation of Dynamic Processes
The modified equation introduces d[Sc]/dt to capture time-dependent
assembly, enabling dynamic
simulation under fluctuating lactate.
2. Reflecting the Concentration Dependence of Sensor
Subunits
Explicit [SN] and [SC] terms relate subunit abundance to complex formation
efficiency.
3. Accurately Characterizing the Binding Reversibility
of the
Complex
The −kd[Sc] term models complex dissociation as lactate
decreases, avoiding signal
redundancy.
(3)
Abbreviations (Part 1)
Table 1
Symbol
Parameter Description
Vm
Maximum transport rate
KmLac
Michaelis constant for lactate
KmH
Michaelis constant for protons
Keq
Equilibrium constant
[Lac]out
Extracellular lactate concentration
[Lac]in
Intracellular lactate concentration
[H+]out
Extracellular proton concentration
[H+]in
Intracellular proton concentration
kₛ
Association rate constant
kd
Dissociation rate constant
Kₛ
Half-maximal effect constant
n
Hill coefficient
[SN]
Sensor N-terminus concentration
[SC]
Sensor C-terminus concentration
[Scom]
Sensor complex concentration
Table 1. Abbreviations (Part 1).
See also Table 1 for abbreviations used in
this section.
3.2 Protease Assembly Module (P)
P
No. 2: Protease Assembly Module (P)
To achieve “controllable activation” of Tobacco Etch Virus (TEV)
protease—i.e.,
enzymatic activity only under high lactate—this project adopts a
‘split-reassembly’ design strategy, dividing
the intact
enzyme into non-catalytic N-terminal (TN) and C-terminal (TC) fragments that
reconstitute activity only upon sensing.
Guided by this fusion strategy and the arrangement order of functional
fragments
across the linker,
a total of 8 fusion protein structures were
designed in
two categories:
“TEV fragment-(GGGGS)3-sensor subunit”
(T-(GGGGS)3-S)
and
“sensor subunit-(GGGGS)3-TEV fragment”
(S-(GGGGS)3-T), as illustrated below.
Figure 3
Figure 3. Eight Protein Complex Structures.
Functional TEV protease is only formed when SN and SC
bind,
thereby inducing the assembly of TN and TC into an intact
enzyme.
A total of eight possible combinations are
generated
(see Figure 4).
Figure 4
Figure 4. Structures of the Eight Fusion Protein
Combinations.
Guided by the kinetic characteristics of bimolecular assembly and strictly
adhering
to the law of mass action, we have developed a mathematical model to quantify the sensor-mediated TEV protease assembly
process. This model enables dynamic simulation of the formation
rate and
concentration kinetics of functional TEV protease (Pa)
by
parameterizing interactions between the lactate sensor complex
(Sc) and TEV fragments
(TN/TC).
Assembly kinetics(4)
Meanwhile, to optimize the system performance, we conducted screening on the
eight
potential fusion protein combinations. First, we predicted the
structures of all eight protein complexes. As an example, the
molecular
docking results for SC-(GGGGS)3-TC are shown below:
Figure 5
Figure 5. Molecular Docking Results of
SC-(GGGGS)3-TC.
Subsequently, we simulated the docking conformations of different
combinations to
examine their binding sites. As an example, the
docking
results between SN-(GGGGS)3-TN and
SC-(GGGGS)3-TC are illustrated below:
Figure 6
Figure 6. Docking Results of
SN-(GGGGS)3-TN and
SC-(GGGGS)3-TC.
We implemented a combined screening strategy for the eight fusion protein
constructs
by integrating molecular docking simulations and mathematical modeling.
Multiple performance metrics were normalized,
weighted, and
consolidated to generate a composite score for each construct.
Given the paucity of experimental parameters from wet-lab studies, we
employed a
fully computational-driven approach independent of empirical measurements.
This
method leverages physical principles of protein
structure and system dynamics
simulations to
identify the optimal complex configuration.
The predictive outcomes of this in silico screening provide
actionable
guidance for subsequent wet-lab validation.
The screening framework is anchored in three core
assumptions:
Spatial conformational compatibility dictates the complementation
efficiency of
TEV fragments.
Changes in binding free energy are the primary determinants of system
sensitivity and specificity.
Kinetic parameters (e.g., assembly rate constants) can be inferred from
the
biophysical properties of the linker peptides.
This study selected four core parameters—Spatial
Complementarity Index (SCI), Orientation
Match
Factor (OMF), Binding Energy Score
(BES),
and Kinetic Efficiency Coefficient (KEC)—to
establish a
quantitative analysis system for evaluating fusion protein configurations.
To quantify the inhibitory effect of spatial deviation on protease activity,
this
study introduced a Gaussian decay function for modeling. Crystallographic
data of
TEV protease (PDB: 1LVM) show that when the distance between the two
fragments
exceeds 0.5 nm or the relative orientation deviates by more than 30°,
catalytic
efficiency decreases by over 90%—this threshold is used as a key parameter
for the
decay function.
Spatial Complementarity
Index(5)
This study employs a Sigmoid function to
quantitatively assess
the impact of fusion orientation (i.e., the linking order between
TEV
fragments and sensor subunits) on functionality, with the output being a
continuous
value ranging from 0 to 1. This value directly
reflects the
relative activity level of the protease under different fusion
orientations.
The core determinant of protease activity is the
spatial
accessibility of its catalytic residues. Taking TEV protease as
an
example, its catalytic triad (H46, D81, C151) must maintain an unobstructed
and
exposed state to bind substrates and perform cleavage functions. When TEV
fragments
are fused to sensor subunits via either their N-terminus or C-terminus, the
spatial
arrangement at the fusion junction may lead to the shielding of catalytic
residues
by adjacent domains—this creates steric hindrance at the active site,
thereby
reducing the functional activity of the protease. By fitting the nonlinear
relationship among “fusion orientation, degree of steric hindrance, and
activity
retention rate,” the Sigmoid function enables precise
differentiation of how different linking orders affect the accessibility
of
catalytic residues.
Orientation Match Factor(6)
We employed the AMBER force field to calculate
the non-bonded interaction energy (encompassing
electrostatic interaction energy and van der Waals interaction energy)
between local
atoms at the fusion interface. A negative energy value indicates that
attractive
forces dominate in the system (conducive to complex stability), while a
positive
value reflects the predominance of repulsive forces (leading to
conformational
instability).
The theoretical basis for this calculation method lies in the fact that the
energy
balance during protein folding is primarily determined by interactions
between local
residues. Specifically, the charge distribution and spatial arrangement of
adjacent
amino acids at the fusion interface affect the overall conformational
stability
through non-bonded interactions (electrostatic attraction/repulsion, van der
Waals
forces). The AMBER force field can accurately quantify
such
local interactions via parameterized potential energy functions.
Binding Energy Score(7)
This study modeled the rate at which TEV protease fragments form active
complexes
via a diffusion-collision process, drawing on
transition state theory. The complementation efficiency of enzyme fragments
(i.e.,
the formation rate of active complexes) is subject to dual constraints: on
one hand,
it is diffusion-controlled (depending on the
movement
rate of fragments in solution and their collision frequency); on the other
hand, it
is limited by conformational energy barriers
(related
to the spatial matching between fragments and the energy required for
conformational
adjustment). By integrating diffusion kinetic
parameters and
conformational energetic characteristics, this model enables more
accurate
quantification of the assembly kinetics of active TEV protease.
Kinetic Efficiency
Coefficient(8)
The specific performance parameters for the eight combinations are presented
in
Table 2 below:
From the computational simulation results, it can be observed that the KEC values of the fusion protein constructs 'LS3.1',
'LS3.7', and 'LS3.8' differ significantly from those of other
constructs:
their KEC values span multiple orders of magnitude and approach zero. This
indicates
that the TEV protease assembly kinetic efficiency of these three constructs
is
extremely low, which fails to meet the functional requirements of the
system. Therefore, they were excluded from the scope
of subsequent
wet-lab validation.
Based on the four performance parameters described earlier, we further established a mapping relationship between the
quantitative
values of each parameter and the key functional indicators of the
system.
The specific correspondences are as follows: the parameter values are mapped
to
system sensitivity (expressed as the reciprocal of EC50, i.e.,
EC50−1), soluble lactate oxidase (sLOx) production,
background
leakage signal intensity, and response speed, providing a
quantitative correlation basis for subsequent functional
evaluation.
Sensitivity(9)
sLOx production proxy(10)
Background leakage(11)
Response speed(12)
Here, S1 represents sensitivity (EC50−1), S2
corresponds to
sLOx production, S3 denotes background leakage, and S4 indicates response
speed.
After calculating the four aforementioned metrics, we first normalized each parameter value (uniformly
mapping
indicators with different dimensions to the [0,1] interval) and then assigned corresponding weights to each parameter
based on
the functional priorities of the system. Finally, a
comprehensive score for each fusion protein construct was
generated using
the weighted summation method. This score served as the core screening
criterion to
identify the optimal construct from the candidate combinations.
Weighted Scoring
We normalized each parameter value, assigned corresponding weights, and computed
a comprehensive score for each fusion protein
construct.
Composite score(13)
As indicated by the comprehensive scores, the “LS3.5”
combination
achieved the highest rating and was identified as the optimal candidate.
Subsequent
wet-lab fluorescence data confirmed that this pair also exhibited the
highest gene
expression level, consistent with the predictions from the dry-lab
computational
analysis.
Table 3
Normalized Parameter Scores
Combination
Norm S1 (Sensitivity)
Norm S2 (Yield)
Norm S3 (Leakage)
Norm S4 (Speed)
Composite Score
Rank
Table 3. Specific Metrics of the Eight
Combinations.
Figure 7
Composite
Scores Distribution
Figure 7. Chart of Scores for Eight Combinations.
3.3 Gene Regulation Module (G)
G
No. 3: Gene Regulation Module (G)
The gene regulation module acts as the core signal processor of the
lactate-responsive genetic circuit,
converting upstream TEV protease activity into transcriptional output that
ultimately controls secreted sLOx.
In mammalian cells, the gene expression process exhibits inherent
delays arising from promoter activation,
mRNA processing/export, translation, post-translational maturation, and secretion.
These steps introduce a significant lag effect
between protease activation and measurable sLOx levels. To capture this
behavior, we describe the module with
a system of delay differential equations (Eq. 14–16) and further
resolve intra/extracellular pools in Eq. 16a–16b.
GV release & nuclear
entry(14)
GV accumulation depends on active protease
Pa, cytosolic GV pool Ge, and an
effective delay.
Promoter activation &
transcription(15)
Hill-like activation of transcription by nuclear GV
(cooperativity 2).
Protein expression with
delay(16)
Translation with delay τ and first-order
decay.
To enhance physiological realism, we further separate intracellular and
extracellular sLOx dynamics:
Eq. 16a (total intracellular pool) and Eq. 16b
(secreted pool with an extra secretion delay τsec).
Intracellular total sLOx(16a)
Secreted sLOx(16b)
The detailed synthesis/secretion pathway is illustrated in Figure 8. To characterize degradation, we
distinguish three intracellular routes: misfolded ERAD elimination,
endosomal–lysosomal turnover of internalized protein,
and baseline turnover (see Figure 9). The resulting model
is given by
Eq. 17a–17b.
Figure 8. sLOx synthesis and secretion.
Figure 9. sLOx degradation pathways.
Detailed intracellular
balance(17a)
ERAD removal of misfolded proteins, endosomal uptake, and
baseline turnover.
Endosomal–lysosomal
degradation(17b)
Equations 14–17b together define the
gene-regulation block that converts
protease activity to secreted sLOx dynamics, accounting for transcriptional
activation,
translation/secretion delays, and multi-route degradation.
Abbreviations (Part 3)
Table 5
Symbol
Parameter Description
kc
Cleavage rate constant
λ
Spatial efficiency factor
τ
Total time delay
γg
GV degradation rate
βg
Maximum transcription rate
Kg
Half-activation constant
δm
mRNA degradation rate
αp
Translation rate
δo
Enzyme degradation rate
[Ge]
GV-ERT2 concentration
[Pa]
TEV protease concentration
[Ox,total]
Total intracellular sLOx
η
Secretion efficiency factor
δo,in
Intracellular sLOx degradation rate
δo,out
Extracellular sLOx degradation rate
τsec
Secretion time delay
δfold
Misfolded protein degradation rate
Fmis
Misfolding proportion
δendo
Endocytic degradation rate constant
kendo
Endocytosis rate (affected by membrane properties)
δlys
Lysosomal degradation rate
δbase
Constitutive degradation rate
Table 5. Abbreviations (Part 3).
3.4 Metabolic Output Module (M)
M
No. 4: Metabolic Output Module (M)
Module scope
This module primarily involves the enzymatic degradation of lactate by sLOx and
its coupling with
transmembrane transport, thereby closing the feedback loop with the sensing
module.
Overall reaction
The specific oxidation process catalyzed by lactate oxidase is:
Eq. (18).
Michaelis–Menten kinetics
The catalytic rate of lactate consumption by sLOx follows a Michaelis–Menten
form
(Eq. 19).
Coupling with Module S
Because sLOx reduces the lactate level in the microenvironment and thus affects
the input of Module S,
we integrate the two into a unified ODE framework. The extracellular lactate
concentration
[Lac]out changes due to three processes—tumor production,
cellular uptake, and
sLOx-catalyzed degradation—summarized in Eq. 20. The
intracellular coupling is given by
Eq. 21.
LOD reaction(18)
Lactate oxidation to pyruvate with
H2O2 and CO2 by LOD/sLOx.
Lactate consumption rate(19)
Here Vm is the maximal rate and
Km the Michaelis constant; [Ox] is secreted
sLOx.
Extracellular lactate
balance(20)
Tumor production − cellular uptake/transport − sLOx
degradation (Michaelis–Menten form).
Intracellular lactate
balance(21)
Transport-driven influx scaled by area/volume, minus
intracellular clearance.
Through the two coupling equations above, Module M and Module S form a
closed feedback loop that jointly
governs extracellular and intracellular lactate dynamics.
Abbreviations (Part 4)
Table 6
Symbol
Parameter Description
Vm
Maximum reaction rate
Km
Michaelis constant
[Ox]
Lactate oxidase concentration
[L]
Lactate concentration
PL
Lactate production rate by tumor cells
Table 6. Abbreviations (Part 4).
4. Screening of Eight Combinations
Overview
In the research on the SPGM lactate-responsive genetic circuit, the screening of eight
fusion protein constructs
represents a core critical step, whose primary objective is to achieve a functional
closed-loop of
“precise lactate sensing and efficient lactate degradation.” This study established a
screening strategy
centered on dry-lab quantitative prediction and supported by wet-lab validation. By
constructing a quantitative
evaluation system based on four key performance parameters (Spatial Complementarity Index,
SCI; Orientation Match
Factor, OMF; Binding Energy Score, BES; Kinetic Efficiency Coefficient, KEC), combined with
a weighted scoring
method, the functionally optimal fusion protein construct was finally identified. The
specific technical workflow
and design rationale are outlined below:
The design of the eight fusion protein constructs stems from the core
requirement of
“controllable activation of TEV protease.” In this study, TEV protease was split
into an inactive
N-terminal fragment (TN, containing catalytic residues H46 and D81) and a
C-terminal fragment
(TC, containing the core catalytic residue C151). These two fragments were fused
to either the
N-terminus (SN) or C-terminus (SC) of the lactate sensor via a flexible linker
(GGGGS)₃,
forming two structural types: “T-(GGGGS)₃-S” and “S-(GGGGS)₃-T.” This resulted
in a total of eight
distinct fusion protein constructs.
The screening process was conducted in two steps, as detailed
below:
Pre-screening: Elimination of “Non-functional
Constructs”
A rapid screening was performed using the kinetic rate constant (KEC) as the
core indicator to
eliminate constructs with lost functionality. Calculations revealed that the
KEC values of the
“LS3.1”, “LS3.7”, and “LS3.8” constructs approached zero, differing by an
order of magnitude from
other constructs. This indicates that the TN and TC fragments in these three
constructs are almost
unable to form active TEV protease through diffusional collision—similar to
“parts that cannot be
assembled into a functional tool.” Even if the sensor successfully detects
lactate, these constructs
fail to initiate downstream signaling pathways. Consequently, they were
directly excluded from
wet-lab validation, significantly reducing unnecessary experimental costs.
Quantitative Scoring & Wet-Lab Validation
The remaining candidates were evaluated across four dimensions—spatial
complementarity (SCI), orientation
alignment (OMF), interface stability (BES), and kinetic efficiency (KEC)—via
docking and mathematical
modeling; results were summarized in the Construct-Parameter Correspondence Table.
High-potential constructs (e.g., “LS3.4” and “LS3.5”) were then prioritized
for validation using
co-transfection with a dual-luciferase reporter in 293T cells under graded
lactate. “LS3.5” achieved the
highest sLOx expression and lactate degradation efficiency (≈60% reduction
within 72 h), consistent
with dry-lab predictions, and was finalized as the optimal construct.
II. Physical Significance and Importance of the Four Key Performance
Parameters (SCI/OMF/BES/KEC)
The four parameters comprehensively evaluate the functional feasibility of
fusion protein
combinations from four critical perspectives: ability to assemble, catalytic
efficiency, structural
stability, and response speed. Each dimension is indispensable for ensuring
optimal performance:
Parameter
Physical Significance
Importance
SCI
Spatial Complementarity Index
Geometric reach & alignment
to form the active center
Evaluates whether the TN and TC domains of TEV protease can
reach and align correctly to form
an active center after lactate sensor binding.
(Simplified model: The sensor is represented as a
5 nm rigid sphere; TEV fragments
are treated as “core components”. Activity decreases by >90%
if the distance between
components exceeds 0.5 nm or their relative orientation
deviates by >30°.)
Requires precise alignment between the active centers of TN and
TC to enable cleavage of
downstream molecules (e.g., GV-ERT2).
If SCI is too low, even upon lactate sensing by the receptor,
TEV fails to form an active
structure, resulting in a signal interruption within the genetic
circuit. Thus, SCI serves as
the fundamental criterion determining whether the system can
initiate its function.
OMF
Orientation Match Factor
Directional matching of
catalytic residues
Assessing whether the catalytic residues of TN and TC are
properly aligned upon association.
(For example, H46 and D81 in TN must directly face C151 in
TC. Misalignment is analogous
to “scissor blades facing back-to-back,” rendering the complex
incapable of cleavage.)
Even if TN and TC are in close proximity, misalignment of
catalytic residues prevents cleavage
activity. The OMF metric addresses the “directional limitation”
of SCI — for instance, while
combination “LS3.3” exhibits a high SCI, its OMF is only 0.096,
resulting in significantly
lower actual activity compared to combination “LS3.5” (OMF =
0.442). Thus, OMF serves as a key
determinant of functional efficiency.
BES
Binding Energy Score
Local interface stability
(AMBER)
The structural stability at the fusion site between the sensor
and TEV fragment was
computationally evaluated. A negative value indicates “high
structural stability” (dominated
by attractive forces), while a positive value suggests “low
structural stability” (dominated
by repulsive forces).
(Calculations were performed using the AMBER molecular
mechanics force field,
incorporating electrostatic and van der Waals interactions.)
Fusion proteins must maintain intracellular stability to
function persistently. A less
negative or positive BES indicates poorer stability (e.g., BES =
−2500 is worse than −3240).
Junction fragility may lead to dissociation of TEV fragments or
sensor failure. Thus, BES
serves as a critical indicator for the long-term reliability of
the constructs.
KEC
Kinetic Efficiency Coefficient
Assembly speed from
diffusion–collision
Measures how rapidly the TN and TC fragments associate to form
active TEV protease, reflecting
the response speed of the combination to lactate signals.
(Based on transition state theory, the rate constant for
active complex formation via
diffusion and collision is estimated.)
The lactate concentration in the tumor microenvironment is
highly dynamic (e.g., may abruptly
rise or fall). If the KEC value is too low (e.g., KEC = 0.000150
for the “1+8” combination),
the slow assembly rate of TEV protease may cause the system to
miss the optimal window for
lactate degradation. In contrast, the “LS3.5” combination
exhibits a significantly higher KEC
value (0.114874), enabling rapid response to lactate signals.
Therefore, KEC is a critical
parameter determining whether a combination can respond in a
timely manner.
III. Weighted Scoring Strategy: Screening for the Optimal Construct via
“Requirement-Oriented Weighting”
The core of weighted scoring lies in assigning higher weights to performance
indicators that are
critical to the system’s ultimate goal, thereby avoiding biased selection
dominated by a single
indicator. This process is divided into two steps:
Definition of Scoring Criteria
The weighted scoring strategy is designed around the core objective of the
SPGM system—dynamically
reducing lactate concentration in the tumor microenvironment. Accordingly,
the four performance
indicators are assigned weights based on their functional importance:
High lactate sensitivity (accurate detection of elevated lactate
levels, 35%) and high sLOx production (efficient
lactate clearance, 35%) are classified as core
requirements;
Low background leakage (20%) is defined as a
secondary requirement;
Fast response speed (10%) is regarded as a basic
requirement.
Comprehensive Scoring: Weighted Sum After
Normalization
Due to variations in the value ranges of different indicator scores (e.g.,
S₁ may range from 0.001
to 0.1, while S₃ may span 0.2 to 0.8), the scores of each construct must
first be normalized to a
relative scale of 0–1 (where 1 represents the best performance for that
indicator, and 0 represents
the worst). The final composite score is calculated as the weighted sum of
the normalized scores
multiplied by their respective weights. Take the “LS3.5” construct as an example: After normalization: S₁ =
1, S₂ = 1, S₃ = 0.503,
S₄ = 1. Composite score = (1 × 0.35) + (1 × 0.35) + (0.503 × 0.20) + (1 ×
0.10) = 0.90068.
This score is significantly higher than that of other constructs (e.g., the
“2+8” construct only
scored 0.309863). Thus, “LS3.5” was identified as the
optimal choice.
Summary
The screening of the eight fusion protein constructs essentially represents a
shift from empirical
judgment to scientifically parameter-driven evaluation. The four key performance
parameters address
distinct functional dimensions of the system:
Efficiency (how well the system performs, reflected by sLOx
production and lactate clearance);
Sensitivity (whether the system can accurately detect
lactate signals);
Background leakage (whether cellular resources are wasted);
Speed (how quickly the system responds after signal
detection).
The weighted scoring system, designed based on actual therapeutic requirements,
ensures that the
selected “LS3.5” construct not only avoids unnecessary cellular resource waste
(low background
leakage) but also responds precisely to tumor-derived lactate signals, achieves
efficient lactate
degradation, and initiates rapid reactions. This outcome lays a critical
foundation for the
therapeutic potential of the SPGM system.
5. Complete Systemic Feedback Loop &
Simulation
5.1 Complete Systemic Feedback Loop
Complete Systemic Feedback Loop
Initiation: High extracellular lactate concentration [Lac]out.
Input and Sensing (Module 1): [Lac]out is transported into the
cell via the MCT1 transporter
(Equation 2), increasing intracellular lactate levels
[Lac]in.
The rise in [Lac]in further induces the formation of the sensor complex
[Sc]
(Equation 3).
Signal Processing (Modules 2 & 3): The formation of [Sc]
promotes the assembly of active
TEV protease [Pa] (Equation 4). Activated
[Pa] cleaves and
releases the transcription factor [Gv], ultimately inducing the expression and secretion of
sLOx
([Ox]) (Equations 14–17).
Metabolic Output (Module 4): Secreted sLOx [Ox] degrades
extracellular lactate
[Lac]out in the microenvironment (Equation 19).
Negative Feedback: The reduction in [Lac]out attenuates the
stimulation of 293T cells, leading to
downregulation of sLOx production and establishing a dynamically balanced closed-loop
regulation.
5.2 Simulation Results
This integrated model more accurately simulates the behavior of the SPGM system within the tumor
microenvironment,
predicting how the system responds to and actively regulates lactate levels, thereby enabling
evaluation of its
effectiveness as an 'intelligent' therapeutic strategy.
Based on this coupled model, we simulated the temporal dynamics (0–72 hours) of both
[Lac]out and [Ox].
Time (h)
[Lac]out (μM)
[Ox] (molecules)
0.0
5000.0
0.0
1.5
6335.0
3.1
2.4
34.2
55.3
3.0
34.3
54.3
6.0
37.1
50.2
9.0
39.8
47.0
12.0
41.8
44.8
15.0
43.5
43.2
18.0
44.6
42.1
21.0
45.5
41.4
24.0
46.0
40.9
27.0
46.4
40.6
30.0
46.7
40.4
33.0
46.8
40.2
36.0
47.0
40.2
39.0
47.0
40.1
42.0
47.0
40.1
45.0
47.0
40.0
48.0
47.2
40.0
51.0
47.1
40.0
54.0
47.1
40.0
57.0
47.1
40.0
60.0
47.1
40.0
63.0
47.2
40.0
66.0
47.1
40.0
69.0
47.1
40.0
72.0
47.1
40.0
Figure 10: Temporal Changes in [Lac]out and sLOx [Ox]
I. Core Dynamic Patterns
Expand core patterns
Phased sLOx [Ox] Response:
During the initial simulation phase (0–1.5 hours), sLOx concentration remained
at a low level,
corresponding to the system signal initiation period. Lactate enters the cell
via MCT1, induces sensor
complex formation, promotes TEV protease assembly, and activates GV. Due to a
1.5-hour time lag in gene
expression, sLOx secretion had not yet commenced significantly.
Between 1.5–2.4 hours, sLOx concentration rapidly increased to its peak (∼55.3
molecules), marking the
high-efficiency secretion phase. By this stage, activated TEV had extensively
cleaved GV and initiated
sLOx gene expression, with secretion efficiency (via the ER-Golgi pathway)
reaching a steady state.
From 2.4–72 hours, sLOx concentration decreased slightly and then stabilized
near peak levels with minor
fluctuations, reflecting the system’s dynamic balance between sLOx expression
and degradation (including
intracellular misfolding-dependent degradation and extracellular constitutive
degradation).
Synchronous Feedback Regulation of [Lac]out:
During the initial phase (0–1.5 hours), [Lac]out continued to rise
and remained at a high initial level
(peak ≈ 6335 μM), as the low sLOx concentration resulted in a lactate
degradation rate lower than the
secretion rate by tumor cells.
Between 1.5–2.4 hours, as sLOx levels increased rapidly, [Lac]out
exhibited a pronounced decline, dropping
to approximately 34.2 μM at the 2.4-hour mark. During this period, the
sLOx-mediated lactate degradation
rate significantly exceeded both the secretion rate of tumor cells and cellular
uptake, representing the
high-efficiency lactate clearance phase of the system.
From 2.4–72 hours, [Lac]out entered a phase of gradual increase,
eventually stabilizing at around 47.1 μM.
At this stage, the sLOx degradation rate reached a dynamic equilibrium with
lactate production and uptake
rates, establishing a homeostatic state under closed-loop 'lactate–sLOx'
regulatory control.
II. Validation of System Functionality
Dynamic Responsiveness Meets Requirements
The concentration of sLOx exhibits a characteristic pattern of 'delayed initiation –
rapid increase –
steady-state maintenance' in response to changes in extracellular lactate
([Lac]s). This demonstrates that
the SPGM system can autonomously regulate its metabolic output based on lactate levels
in the tumor
microenvironment, effectively avoiding 'over-secretion in the absence of lactate' and
'insufficient
response under high lactate conditions'. These results confirm that the system achieves
its intended
design goal of intelligent lactate reduction.
Significant Lactate Degradation Efficiency
Within 72 hours, extracellular lactate ([Lac]out) decreased from an initial
concentration of approximately
5000 μM to about 47.1 μM, representing a reduction of 99.058%. Effective control of
lactate levels
(reduced to 0.9% of the initial value) was achieved within 20 hours.
Reliable Stability of Closed-Loop Regulation
From 2.4 to 72 hours, both [Lac]s and sLOx concentrations remained in a
stable plateau phase without
significant fluctuations. This confirms the effectiveness of the negative feedback loop
in the SPGM
system: elevated lactate → increased sLOx secretion → lactate degradation →
downregulation of sLOx
secretion. This regulatory mechanism prevents resource waste due to excessive sLOx
production and avoids
lactate rebound caused by insufficient sLOx, demonstrating the robustness of the system
for long-term
operation.
III. Alignment with System Design Objectives
The simulation results strongly align with the core objective of the SPGM system:
dynamically reducing
lactate levels in the melanoma microenvironment and reversing immunosuppression. The phased
secretion
pattern of sLOx demonstrates the system’s ability to precisely adapt to dynamic changes in
tumor-derived
lactate, enabling autonomous regulation without exogenous intervention. These findings
provide a theoretical
foundation for subsequent wet-lab experiments and offer key modeling support for the
feasibility of the
system as a standalone therapy or a combination therapy enhancer.
5.3 Sobol Sensitivity Analysis
To further refine the modeling process, we performed a global sensitivity
analysis of factors
influencing extracellular lactate concentration using a Sobol variance–decomposition framework
(first- and
total-order indices). Key parameters included tumor lactate production (PL),
transport
velocity (v), sLOx catalytic constants (Vm,ox, Km,ox),
and secretion/decay
terms.
Figure 11: Global Sensitivity Analysis Results
The results indicate that lactate concentration in the tumor microenvironment is most
strongly correlated
with the hydrogen ion concentration within 293T cells.
6. Connections to Wet-Lab Experiments
6.1 Guidance from Dry-Lab to Wet-Lab Studies
Based on molecular docking and molecular dynamics analyses, the dry-lab results
identified that
combinations “LS3.1”, “LS3.7”, and “LS3.8” exhibit
significantly different KEC values
(differing by orders of magnitude) compared to other constructs, approaching zero.
Therefore, these three
combinations may be excluded in subsequent wet-lab experimental procedures.
Global sensitivity analysis revealed that lactate concentration in the tumor
microenvironment is most strongly
correlated with hydrogen ion concentration within 293T cells. Since reducing lactate
levels in the tumor
microenvironment is a key objective of our project, subsequent wet-lab experiments
should prioritize accurate
measurement of intracellular hydrogen ion concentration. To enhance the efficiency of
lactate reduction,
strategies targeting modulation of intracellular hydrogen ion concentration should be
considered first.
6.2 Validation of Dry-Lab Predictions via Wet-Lab Experiments
1、Experimental Verification of Dry-Lab Assumptions
The homogeneity of the reaction
The homogeneity of the reaction system was tested using Förster Resonance Energy
Transfer (FRET).
Constant time-delay validation
The assumption of constant time delay was validated by monitoring sLOx mRNA levels
via quantitative real-time
PCR (qPCR).
Enzymatic kinetic stability across pH
Enzymatic kinetic stability was assessed by measuring sLOx activity in a pH range of
6.8–7.6 in an in vitro
buffer system.
2、Functional Validation of Combination Performance
The experimental group co-transfected 293T cells with the LIdR-TEV fusion plasmid, GV-2ER
plasmid, and pGL4.35.
Signal activation under varying lactate concentrations was measured using a dual-luciferase
reporter assay.
Results confirmed that the “LS3.5” combination performed optimally, consistent with
dry-lab predictions.Lab Results
Representative wet-lab validation schematic.
3、Kinetic Validation of Model Predictions
Starting with an initial lactate concentration of 5000 μM in the 293T culture environment,
extracellular lactate
and sLOx secretion levels were measured every 3 hours over a 72-hour period. This
time-series data was used to
evaluate the accuracy of the dry-lab predicted equations.
7. Model Predictions (Markov)
7.1 Markov Model
Markov Model
To more accurately simulate intracellular heterogeneity, local concentration gradients, and
stochastic
effects, we employed a Markov model to predict and simulate gene expression regulation. This
Markov model module
serves as a complement and extension to the gene regulation module (G), aiming to
characterize the molecular-level
stochasticity of GV protein activation and sLOx gene expression from the perspective of
stochastic processes.
Traditional ordinary differential equation (ODE) models are based on the assumption of
continuous
concentration changes and are suitable for systems with high molecular abundances. However,
they fail to capture
stochastic fluctuations under low-copy-number conditions or the discrete nature of
individual molecular events.
GV proteins and transcription factors typically exist at low copy numbers (ranging from a
few to a few hundred
molecules per cell), and gene expression events are inherently discrete and stochastic.
Why Markov models?
Capture intrinsic stochasticity at the molecular level
Reveal the origins of cell-to-cell variability
Predict threshold effects and bimodal distributions
Provide more realistic theoretical predictions for experimental design
This module employs a Continuous-Time Markov Chain (CTMC) model to simulate the state
transitions of
individual GV molecules and sLOx gene expression events, thereby more accurately capturing
the intrinsic
stochasticity of biological systems. It consists of two Markov submodels: one for GV
activation and another
for sLOx expression.
GV Activation CTMC (single “GV-ERT2” unit)
State Definitions:
State 0 (S₀): GV-ERT2 intact, uncut.
State 1 (S₁): Bound by an active TEV protease (Pₐ).
State 2 (S₂): GV cleaved and released as a free transcription factor.
Figure 12. State Diagram of the GV Activation Markov
Model
Gillespie algorithm (SSA) steps:
Initialization: All GV molecules start in State 0 (intact
and unbound).
Rate Calculation: Compute all possible transition rates for
each molecule.
Event Selection: Generate random numbers to determine the
time and type of the next transition.
State Update: Update the molecular state and advance the
system time.
Iteration: Repeat until the simulation end time is reached.
Specific results are shown below:
Figure 13. Simulation Results of the GV Activation Markov Model
Rapid Binding Phase (0–1 hour): GV molecules quickly bind to TEV
protease.
Activation Phase (1–5 hours): Bound GV is cleaved, leading to a rapid
increase in the activation ratio.
Stabilization Phase (5–72 hours): The system reaches dynamic
equilibrium, with approximately 70–80% of GV activated.
Key Quantitative Results:
Final GV activation ratio: 100.0%
Time required to achieve 50% activation: 1.1 hours
sLOx Gene Expression Modeling (CTMC)
A multi-component Markov model was employed to simulate sLOx expression, with a
two-dimensional state space
vector (m, p), where m represents the number of mRNA molecules
and p the
number of sLOx protein molecules. The model incorporates the following stochastic events:
transcription,
translation, mRNA degradation, protein degradation, and protein secretion. A variant of the
Gillespie
algorithm was used to handle the multi-dimensional state space. Specific results are shown
below:
Figure 14. Simulation Results of the sLOx Gene Expression
Markov Model
Quantitative snapshot:
Final secreted sLOx: 25,786 molecules
Average mRNA count: 20.21
Average intracellular protein: 199.63
Insights beyond ODEs:
Transcriptional Bursting: Genes undergo random “bursts” of
transcription rather than continuous mRNA production.
Expression Noise: Even genetically identical cells exhibit significant
variations in expression levels.
Threshold Effects: Some cells may show no sLOx expression, while others
exhibit unusually high expression.
Temporal Heterogeneity: Significant delays and variability exist
between GV activation and sLOx secretion.
7.2 Future Directions
Incorporate spatial heterogeneity into the modeling framework
Develop more efficient computational algorithms for large-scale systems
Refine model parameters by integrating single-cell omics data
Explore machine learning methods to accelerate stochastic simulation processes
References
Wang X, Liu H, Ni Y, Shen P, Han X. Lactate shuttle: From substance exchange
to regulatory
mechanism. Hum Cell. 2022;35(1):1–14. Roosterman D, Meyerhof W, Cottrell GS.
Proton transport
chains in glucose metabolism: mind the proton. Front Neurosci. 2018;12:404.
San Martín A, Ceballo S, Ruminot I, Lerchundi R, Frommer WB, Barros LF. A
genetically encoded
FRET lactate sensor and its use to detect the Warburg effect in single cancer cells.
PLoS One.
2013;8(2):e57712.
Chen Y, Wei LN, Müller JD. Quantitative FRET (qFRET) technology for the
determination of
protein-protein interaction affinity in solution. Sci Rep. 2016;6:31991.
Pucino V, Bombardieri M, Pitzalis C, Mauro C. Lactate: a metabolic driver in
the tumour
landscape. Trends Immunol. 2019;40(2):128–141.
Wehr MC, Laage R, Bolz U, Fischer TM, Grünewald S, Scheek S, Rossner MJ.
Monitoring regulated
protein-protein interactions using split TEV. Nat Methods. 2006;3(12):985–993.
Cambridge SB, Gnad F, Nguyen C, Bermejo JL, Krüger M, Mann M. Systems-wide
proteomic analysis
in mammalian cells reveals conserved, functional protein turnover. J Proteome
Res.
2011;10(12):5275–5284.
Hargrove JL, Schmidt FH. The role of mRNA and protein stability in gene
expression. FASEB
J. 1989;3(12):2360–2370.
Beerli RR, Segal DJ, Dreier B, Barbas CF. Toward controlling gene expression
at will:
specific regulation of the erbB-2/HER-2 promoter by using polydactyl zinc finger
proteins constructed from
modular building blocks. Proc Natl Acad Sci U S A. 1998;95(25):14628–14633.
Schwanhäusser B, Busse D, Li N, Dittmar G, Schuchhardt J, Wolf J, Selbach M.
Global
quantification of mammalian gene expression control. Nature.
2011;473(7347):337–342.
Ma C, Tan Z, Lin Y, Li L, You C. Rational design of lactate oxidase for
efficient lactate
detection. Enzyme Microb Technol. 2019;120:9–16.
Liao Y, Fung TS, Huang M, Fang SG, Zhong Y, Liu DX. Efficient secretion of
small hepatitis B
surface antigen by a recombinant Sendai virus. J Virol. 2004;78(13):6815–6826.
Dominguez C, Boelens R, Bonvin AM. HADDOCK: a protein-protein docking
approach based on
biochemical or biophysical information. J Am Chem Soc. 2003;125(7):1731–1737.
Kollman PA, Massova I, Reyes C, Kuhn B, Huo S, Chong L, Cheatham TE.
Calculating structures
and free energies of complex molecules: combining molecular mechanics and continuum
models. Acc Chem
Res. 2000;33(12):889–897.
Hoops S, Sahle S, Gauges R, Lee C, Pahle J, Simus N, Kummer U. COPASI—a
COmplex PAthway
SImulator. Bioinformatics. 2006;22(24):3067–3074.
Saltelli A, Annoni P, Azzini I, Campolongo F, Ratto M, Tarantola S. Variance
based
sensitivity analysis of model output. Design and estimator for the total sensitivity
index. Comput
Phys Commun. 2010;181(2):259–270.
Gillespie DT. Exact stochastic simulation of coupled chemical reactions.
J Phys
Chem. 1977;81(25):2340–2361.
Raj A, van Oudenaarden A. Nature, nurture, or chance: stochastic gene
expression and its
consequences. Cell. 2008;135(2):216–226.
Elowitz MB, Levine AJ, Siggia ED, Swain PS. Stochastic gene expression in a
single cell.
Science. 2002;297(5584):1183–1186.
Sanchez A, Golding I. Genetic determinants and cellular constraints in noisy
gene
expression. Science. 2013;342(6163):1188–1193.
Munsky B, Neuert G, van Oudenaarden A. Using gene expression noise to
understand gene
regulation. Science. 2012;336(6078):183–187.
Remmert M, Biegert A, Hauser A, Soing J. HHblits: lightning-fast iterative
protein sequence
searching by HMM-HMM alignment. Nat Methods. 2011;9:173–175.
Pearson WR, Lipman DJ. Improved tools for biological sequence comparison.
Proc Natl Acad
Sci U S A. 1988;85:2444–2448.
Yan Y, Wen Z, Wang X, Huang S-Y. Addressing recent docking challenges: A
hybrid strategy to
integrate template-based and free protein-protein docking. Proteins.
2017;85:497–512.
Huang S-Y, Zou X. A knowledge-based scoring function for protein-RNA
interactions derived
from a statistical mechanics-based iterative method. Nucleic Acids Res.
2014;42:e55.