Introduction



Mathematical modeling plays a crucial role in understanding the project keeping precision l'esprit. We utilised modelling to gain an understanding of portions of the project we could not experimentally characterise. We aimed to build predictions that we could cross-validate with wetlab data. For Duonco, we segregated the entire project into its constituent steps to look at the processes we could model.


picture

Figure 1: Project Plan


We began by identifying seven areas of the project we could work on. From our interactions with experts, we realistically narrowed down to five. These avenues have been described below.



Protein Production



One of the main questions we wanted to answer was: how could we predict the rate of production of our heterologous proteins inside the bacteria? This is important to predict density of the affibody and THP on Affi-OMVs and THP-OMVs respectively. To answer this question, we initially made a diagram of all the genes and proteins relevant to expression of the recombinant proteins from the plasmid. Our gene of interest is inserted into the E.coli expression vector pGEX-4T-1. Protein expression is under the control of the IPTG inducible tac promoter.

picture

Figure 2. Gene circuit of our project


We wished to predict protein output (the dependent variable) and a function of IPTG concentration and time (independent variables). We applied the following assumptions while building our model:


Assumptions

  • IPTG is transported by both the permease LacY and by free diffusion
  • No catabolite repression occurs as cells were grown in LB Broth, which has no glucose
  • The tac promoter (pTac) drives transcription at a rate 11 × of LacUV5
  • The repressor (LacI) dimerises and the dimer alone binds to the operator site LacO. We neglect tetramerisation of LacI
  • LacI dimer binds with 1:1 stochiometry to operator (LacO) site
  • IPTG is not metabolised by beta-galactosidase (LacZ) and equation for LacZ are neglected
  • 2 IPTG molecules bind to a single LacI dimer
  • Cell death and growth is neglected, and DNA content remains the same.
  • Copy number of pGEX-4T1 plasmid with pBR322 origin is 20
  • The leaky nature of the operon is neglected


Equations for the genomic copy are as given below:

(1.a) Constitutive transcription of LacI is modelled as with zero-order kinetics \[ \phi \overset{k_{SMR} }\to M_{R}\] \[ \frac{dM_{R} }{dt} = k_{sMR} - \lambda_{M_{R} } M_{R}\]
(1.b) Translation of LacI mRNA to the repressor protein R, follows first order kinetics \[ M_{R} \overset{k_{SR} }\to M_{R} + R\]
(1.c) Dimerization of R follows second order kinetics \[ 2R\underset{k_{-2R} }{\stackrel{k_{2R} }{\rightleftharpoons} }R_{2}\] \[ \frac{dR}{dt} = k_{sR}M_{R} + 2k_{-2R} R_{2} - 2 k_{2R}{R}^{2} - \lambda_{R} R\]
Since the concentration of the operator is constant, it is equal to that of the combination of free operator and repressor bound operator \[ O_{T} = O + R_{2}O\] \[ \frac{dR_{2} }{dt} = k_{2R}{R}^2 - k_{-2R} R_{2} - k_{r} R_{2} O + k_{-r} (O_{T} - O) - k_{dr1} R_{2} {I}^{2} + k_{-dr1} [I_{2}R_{2}] - \lambda_{R_{2} } R_{2}\]
(2.a) The dimer \(R_{2}\) binds with the operator O in a 1-1 manner \[ R_{2} + O \underset{k_{-r} }{\stackrel{k_{r} }{\rightleftharpoons} } R_{2}O\]
(3.a) 2 IPTG (I) molecules bind with 1 dimer \(R_{2}\) \[ 2I + R_{2} \underset{k_{-dr1} }{\stackrel{k_{dr1} }{\rightleftharpoons} } I_{2}R_{2}\]
(3.b) IPTG can also bind to the repressor-operator\(R_{2}O\) complex such that 2 IPTG molecules bind to 1\(R_{2}O\) \[ 2I + R_{2}O \underset{k_{-dr2} }{\stackrel{k_{dr2} }{\rightleftharpoons} } I_{2}R_{2} + O\] \[ \frac{dO}{dt} = -k_{r} R_{2} O + k_{-r} (O_{T} - O) + k_{dr2} (O_{T} - O) {I}^{2} - k_{dr1} R_{2} {I}^{2}\]
(4.a) Transcription of the genomic LacY gene under the lac promoter is dependent on the concentration of free operator \[ O \overset{k_{S1MY} }\to O + mRNA_{Y}\] \[ \frac{dM_{Y} }{dt} = k_{s0MY} (O_{T} - O) + k_{s1MY} O - \lambda_{MY} M_{Y}\]
(4.b) Leaky transcription of LacY can be modelled as dependent on concentration of bound receptor \(R_{2}O\) \[ R_{2}O \overset{k_{SOMY} }\to R_{2}O + mRNA_{Y}\]
(4.c) Translation of LacY protein(Y) follows first order kinetics \[ M_{Y}\overset{k_{SY} }\to M_{Y}+Y\]
(5.a) The facilitated tranportation of IPTG by Y from the extracellular region where its concentration is \(I_{ex}\) to the intracellular region where its concentration is I. This reaction is modelled with Michaelis Menten equations \[ Y + I_{ex} \underset{k_{-p} }{\stackrel{k_{p} }{\rightleftharpoons} }YT_{ex} \stackrel{kft}\to Y + I\] \[ \frac{d YI_{ex} }{dt} = -(k_{ft} + k_{-p}) YI_{ex} + k_{p} Y I_{ex} - \lambda_{YI_{ex} YI_{ex} }\] \[ \frac{dY}{dt} = k_{sY} M_{Y} + (k_{ft} + k_{-p}) YI_{ex} - k_{p} Y I_{ex} - \lambda_{Y} Y\]
(5.b) IPTG diffuses across the membrane \[ I_{ex} \underset{k_{t} }{\stackrel{k_{t} }{\rightleftharpoons} }I_{in}\] \[ \frac{dI_{in} }{dt} = -2 k_{dr1} R_{2} {I}^{2} + 2 k_{-dr1} [I_{2}R_{2}] - 2 k_{dr2} (O_{T} - O) {I}^{2} + 2 k_{-dr2} O [I_{2}R_{2}] + k_{ft} YI_{ex} + k_{t} (I_{ex} - I) + 2 \lambda_{[I_{2}R_{2}]} [I_{2}R_{2}] + \lambda_{YI_{ex} } Y_{I_{ex} }\]
(6.a) The degradation equations for various molecular species are denoted below: \[ M_{R}\overset{\lambda_{M_{R} } }\to\phi\] \[ M_{Y}\overset{\lambda_{M_{Y} } }\to\phi\] \[ R\overset{\lambda_{R} }\to\phi\] \[ R_{2}\overset{\lambda_{R2} }\to\phi\] \[ Y\overset{\lambda_{Y} }\to\phi\] \[ YI_{ex}\overset{\lambda YI_{ex} }\to I\] \[ I_{2}R_{2}\overset{\lambda_{I_{2}R_{2} } }\to I\] \[ mRNA_{Y}\overset{\lambda M_{Y} }\to \phi\] \[ C\overset{\lambda c}\to \phi\]


Equations for Plasmid Genes


(7.a) Transcription of LacI under the LacI promoter in the plasmid \[ \phi \overset{k_{SMR} }\to M_{R}\] \[ \frac{dM_{Rp} }{dt} = k_{sMR} - \lambda_{M_{R} } M_{Rp}\]
(7.b) Translation of LacI mRNA to the repressor protein, denoted Rp for the plasmid \[ M_{Rp} \overset{k_{SR} }\to M_{Rp} + Rp\]
(7.c) Dimerization of Rp for the plasmid \[ 2Rp\underset{k_{-2R} }{\stackrel{k_{2R} }{\rightleftharpoons} }R_{2p}\] \[ \frac{dRp}{dt} = k_{sR}M_{Rp} + 2k_{-2R} R_{2p} - 2 k_{2R}{Rp}^{2} - \lambda_{R} Rp\]
Here too, the concentration of the operator is constant, it is equal to that of the combination of free operator and repressor bound operator \[ O_{Tp} = Op + R_{2p}Op\] \[ \frac{dR_{2p} }{dt} = k_{2R}{Rp}^2 - k_{-2Rp} R_{2p} - k_{r} R_{2p} Op + k_{-r} (O_{Tp} - Op) - k_{dr1} R_{2p} {Ip}^{2} + k_{-dr1} [I_{2}R_{2}p] - \lambda_{R_{2} } R_{2p}\]
(8.a) The dimer \(R_{2p}\) binds with the operator Op of plasmid in a 1-1 manner \[ R_{2p} + Op \underset{k_{-r} }{\stackrel{k_{r} }{\rightleftharpoons} } R_{2p}Op\]
(9.a) IPTG(denoted I) binds with the dimer R_{2p} such that 2 IPTG molecules bind with 1 dimer \(R_{2}\) \[ 2I + R_{2p} \underset{k_{-dr1} }{\stackrel{k_{dr1} }{\rightleftharpoons} } I_{2}R_{2}p\]
(9.b) IPTG can also bind to the repressor-operator\(R_{2}Op\) complex such that 2 IPTG molecules bind to \(R_{2}Op\) \[ 2Ip + R_{2}Op \underset{k_{-dr2} }{\stackrel{k_{dr2} }{\rightleftharpoons} } I_{2}R_{2}p + Op\] \[ \frac{dOp}{dt} = -k_{r} R_{2p} Op + k_{-r} (O_{Tp} - Op) + k_{dr2} (O_{Tp} - Op) {I}^{2} - k_{dr1} R_{2p} {I}^{2}\]
(10.a) Transcription of ClyA(denoted c) under the pTac promoter is dependent on concentration of free operator and the rate constant is 11 times \(k_{S1MY}\) \[ O \overset{k_{S1MYp} }\to O + M_{Yp}\] \[ \frac{dM_{Yp} }{dt} = k_{SIMPp} O + K_{leakyp}\]
(10.b) Leaky transcription of ClyA \[ R_{2p}Op \overset{k_{leakyp} }\to R_{2p}Op + M_{Yp}\]
(11.a) Translation of \(M_{Yp}\) to the protein ClyA \[ M_{Yp} \overset{KM_{Yp} }\to M_{Yp} + C\] \[ \frac{dC}{dt} = k_{M_{Yp} } M_{Yp}\]

Symbol Species denoted
MR LacI repressor mRNA
R LacI repressor monomer
R2 LacI repressor dimer
O lacO operator
R2O Repressor-operator complex
I Intracellular inducer IPTG
Iex Extracellular inducer IPTG
I2R2 Repressor-inducer complex
MY LacY permease mRNA
Y LacY permease
YIex Permease-inducer complex
C Cly-A Protein complex
\(\phi\) Generic source or sink

The constants and their values used are given in a table here.

picture

Figure 3: Protein concentration vs IPTG concentration


We obtained a plot of indicating the relationship between the concentration of the inducer IPTG and the output protein. We found that the curve saturates ~1 mM IPTG, and utilised this value while inducing our own bacterial cultures. We found sufficient protein production following this.


OMV Internalisation



One of the processes we aimed to modeling OMV internalisation. OMV internalisation is defined as the process of OMVs entering mammalian cells by receptor mediated endocytosis. In this case, the receptors are HER2 and CX3CR1. Building a model which can recapitulate the essential features of the process can help us predict the concentrations of the enzyme and prodrug to load. This model can also be used to gain estimates on what percentage of administered OMVs internalise, and what the the maximum dosage of OMVs should be.
The following equations can be written to describe the process

  • Initial vesicular association with the membrane can be descibed as given below \[ C+C_{bs} \underset{k_{d} }{\stackrel{k_{a} }{\rightleftharpoons} } C_{b}\]
  • Where \(C\) stands for the free vesicles, \(C_{bs}\) for the number of free cell-surface receptors and \(C_{b}\) for the number of vesicles with single ligand bound to surface receptor. \(k_{a}\) and \(k_{d}\) represent the association and dissociation rate constants
  • Let the equation describing vesicular internalisation given by the equation below \[ C_{b} \overset{k_{int} }\to C_{i}\]
  • The factors affecting the equilibrium reaction can be denoted as below \[ \frac{dC_{b} }{dt} = k_{a}C_{bs}C - k_{d}C_{b} - k_{int}C_{b}\]
  • Similarly, the free receptor change could be taken into account as given below \[ \frac{C_{bs} }{dt} = -k_{a}C_{bs} + k_{d}C_{b}\]
  • Internalisation can be expressed by the equation below \[ \frac{dC_{i} }{dt} = k_{int}C_{b}\]

Variables


Symbol Species denoted
\(C\) Concentration of Vesicles
\(C_{b}\) Concentration of bounded vescicles
\(C_{bs}\) Concentration of free binding sites
\(C_{i}\) Concentration of internalised vesicles

Solving the above system of equations numerically gives us the plots below:

picture

Figure 4: Internalisation of OMVs at low concentrations follows a linear pattern

picture

Figure 5: Internalisation saturates off at a point in the graph



Electroporation


Electroporation is the method of using high voltage electric pulses to increase cell permeability facilitating delivery of drug and genetic material into the cell. In our project, this technique plays a crucial role in loading the prodrug into the OMVs. The challenge was to optimise the size of the pore so as to enhance the rate of drug carried in each OMV without compromising the drug loading process or membrane integrity. Hence, a mathematical model was created to calculate the optimum pore size for effective cargo loading. This was done with the help of one-dimensional differential equations that took the pore size and carrying capacity as the variables.


Pore Density


We assume the OMVs to be spherical in nature, occupying a radius R. The electric field applied induces pores on the surface. Their exact size and radius distribution depends on the electric field being applied. The pore density N is governed by the well known Krassova and Filev equation, given below \[ \frac{d N}{d t} = \alpha\exp\left(\frac{V_{m} }{V_{e p} }\right)^{2}\left[1-\frac{N}{N_{0}\exp\left(q\left(\frac{V_{m} }{V_{e p} }\right)^{2}\right)}\right]\]

Where {V_{m} } is the transmembrane potential of the spherical cell in a uniform electric field defined as: \[ {V_{m} } = 1.5E \times r_{c} cos \psi\]

We isolate particulars of these equation using the below constants \[ K = \alpha{e}^{\frac{V_{m}^{2} }{V_{ep}^{2} } }\] \[ C = \frac{ {e}^{-q{\frac{V_{m}^{2} }{V_{ep}^{2} } } } }{N_{o} }\]

The equation then becomes: \[ \frac{dN_{ep} }{dt} = K(1 - N_{ep}C)\] Solving the above ordinary differential equation yeilds us: \[ -{\frac{ln(1 - N_{ep}C)}{C} }{\rvert}_{N_{epo} }^{ {N_{ep} }^{t} }\] \[ -KCt = ln{(\frac{1 - {N_{ep} }^{t}C}{1 - {N_{ep} }^{o}C})}\] \[ e^{-kct} = \frac{1 - {N_{ep} }^{t}C}{1 - {N_{ep} }^{o}C}\] \[ e^{-kct}{(1 - {N_{ep} }^{o}C)} = {1 - {N_{ep} }^{t}C}\] The constants for our case are given below: \[ \alpha = 1\times10^{9}\] \[ V_{m} = 0.08\] \[ V_{ep} = 0.258\] \[ {(\frac{V_{m} }{V_{p} })} = 0.3100775194\] \[ {(\frac{V_{m} }{V_{p} })}^{2} = 0.096148\] \[ e^{ {(\frac{V_{m} }{V_{p} })}^{2} } = 1.10092\] \[ e^{-{(\frac{V_{m} }{V_{p} })}^{2} } = 1.10092\] \[ r_{m} = 0.8\times{10}^{-9}m\] \[ r_{a} = 0.51\times{10}^{-9}m\] \[ \frac{r_{m} }{r_{\alpha} } = 1.5686\] \[ q = 2.46059\] \[ K = \alpha{e}^{\frac{V_{m}^{2} }{V_{ep}^{2} } } = 1.10092\] \[ C = \frac{ {e}^{-q{\frac{V_{m}^{2} }{V_{ep}^{2} } } } }{N_{o} }= {(\frac{0.789322}{N_{o} })}\]
We plug in these values for our original equation. It leads us to the plot shown below. \[ {N_{ep} }^{t} = {\frac{1 - e^{-kct}{(1 - {N_{ep} }^{o}C)} }{C} }\]


picture

Figure 6: Pore density evolution with time. As expected, the pore density saturates off to a particular value depending on the field strength(characterized by membrane voltage Vm). One also expects the pore density to exponentially decay off to zero as we remove the force field.


Prodrug-Enzyme Interaction



The prodrug is the precursor drug that is inactive but, when acted upon by the cognate enzyme, is activated. The damage to healthy cells is prevented by this inactive nature of prodrug, as the specific combination of HER2 and CX3CR1 found on cancerous cells will make sure that the prodrug and enzyme are selectively internalised. The prodrug-enzyme kinetics are important in understanding amount of prodrug and enzymes required to trigger cell death.

The prodrug in our project is 5-fluorouracil (5 FU) and the enzyme is cytosine deaminase. 5 FU is activated to 5-fluorocytosine by cytosine deaminase.

Assumptions

  • Effect of serum concentration of 5-FC on measured tissue concentration of 5-FC is negligible
  • Effect of bound 5-FC and 5-FU on measured tissue concentrations of 5-FC and 5-FU is negligible
  • Distribution of 5-FC and 5-FU between interstitial and intercellular spaces is effectively instantaneous with a distribution volume of 1.0
  • The distribution of 5-FC, 5-FU and the intermediate F Nuc within the compartments is homogenous
  • Measured levels of 5-FU and FNuc are both, unidirectional
  • FNuc is effecively trapped within the tumor tissue

Rate \[ {k_{1} }^{app}\] \[ {k_{3} }^{app}\] \[ \frac{k_{1} \times k_{3} }{k_{2}+k_{3} }\] \[ {k_{1} }^{app}\]
1 0.316 0.0029 0.0012 0.0090
2 0.831 0.0023 0.0025 0.0018
3 0.728 0.0010 0.0010 0.0009
4 0.287 0.0022 0.0008 0.0010
5 0.267 0.0028 0.0010 0.0011

\[ k_{i} = \frac{ {k_{1} }^{app}\times{k_{3}^{app} } }{ {k_{2} }^{app} + {k_{3}^{app} } }\] \[ Rate = -\frac{d[A]}{dt} = k[A]\] \[ \frac{d[A]}{A} = -kdt\] \[ \int_{[A]_{o} }^{[A]}\frac{d[A]}{[A]} = -\int_{to}^{t}kdt\] \[ \int_{1}^{[A]}\frac{d[A]}{[A]} = -\int_{to}^{t}kdt\] \[ ln[A] - ln[A_{o}] = -kt\] \[ ln[A] = -kt + ln[A_{o}]\] \[ t_{1/2} = \frac{ln2}{k}\] \[ t_{1/2} = 551.26\; \pm 404.92s\]


We calculated the half-life of 5-FU to be 551.2 s


Ligand Receptor Interaction



Ligand-receptor interactions are a major class of protein-protein interactions that regulate life processes and helps maintain specificity of various proteins and their functions. The conformational change that the ligand undergoes effectively acts as a lock and key mechanism which discriminates between proteins and makes sure that only the specific ligand binds to the receptor site. In the context of our project, the ligand is the affibody or the THP and the receptors are HER2 and CX3CR1 respectively. Understanding ligand-receptor interactions and quantifying their rates are crucial for designing the prodrug delivery mechanism and ensuring the selective targeting of cancerous cells with minimum damage to healthy cells. The rate at which the ligand binds to the receptor, the mechanism of binding and the probability of a ligand failing to bind give us important information in deciding the optimum dosage of the prodrug to be loaded. It also helps in finding the rate of reaction and the time taken for the successful completion of the binding. Such studies can be done in-silico with Autodock Vina, a popular molecular docking software.

We went on to conduct docking studies for our project. One of the main inputs we required was the structure file of our THP. Since we were unable to find the THP structure anywhere, a protein structure prediction was done using AlphaFold, an artificial intelligence program developed by DeepMind for the prediction of protein structure. The predicted structures are shown below.


picture picture picture picture

Figure 6: Structures of THP

We required a ligand-receptor system to perform the docking and the following molecules coupled with the above molecule made the system complete.

picture picture picture

Figure 7:Other Proteins: Affibody(Left), CX3CR1(Center), HER2(Right)

We attempted docking using Autodock Vina. However, after multiple attempts over multiple days, we were unable to proceed with the same. After troubleshooting the issue, we came to the conclusion that the molecules we used were too big for the software to handle, thus, our dream of a theoretical free energy estimate was lost. This problem was rectified using literature sources to find our k values.



References


  1. Code for plots - Protein production, OMV Internalization, Electroporation

  2. Team iGEM IISER_TVM 2021 - Moldemort- a novel class of eco-friendly antifungal therapeutics against Invasive Fungal Infections

  3. Eigenbrot C, Ultsch M, Dubnovitsky A, Abrahmsén L, Härd T. Structural basis for high-affinity HER2 receptor binding by an engineered protein. Proc Natl Acad Sci U S A. 2010 Aug 24;107(34):15039-44. doi: 10.1073/pnas.1005025107. Epub 2010 Aug 9. PMID: 20696930; PMCID: PMC2930565.

  4. Jones Emily J., Booth Catherine, Fonseca Sonia, Parker Aimee, Cross Kathryn, Miquel-Clopés Ariadna, Hautefort Isabelle, Mayer Ulrike, Wileman Tom, Stentz Régis, Carding Simon R. The Uptake, Trafficking, and Biodistribution of Bacteroides thetaiotaomicron Generated Outer Membrane Vesicles .Frontiers in Microbiology, 2020, Vol 11, 10.3389/fmicb.2020.00057

  5. Ying CT, Wang J, Lamm RJ, Kamei DT. Mathematical Modeling of Vesicle Drug Delivery Systems 2: Targeted Vesicle Interactions with Cells, Tumors, and the Body. Journal of Laboratory Automation. 2013;18(1):46-62. doi:10.1177/2211068212458265

  6. Stamatakis, M., & Mantzaris, N. V. (2009). Comparison of Deterministic and Stochastic Models of the lac Operon Genetic Network. Biophysical Journal, 96(3), 887–906. https://doi.org/10.1016/j.bpj.2008.10.028

  7. de Boer, H. A., Comstock, L. J., & Vasser, M. (1983). The tac promoter: A functional hybrid derived from the trp and lac promoters. Proceedings of the National Academy of Sciences, 80(1), 21–25. https://doi.org/10.1073/pnas.80.1.21