iGEM Freiburg 2024
iGEM Freiburg 2024  | CAPTURE
  • Description Engineering Success Results Contribution
  • Protocols Notebook Safety
  • Part Collection Plasmid Design
  • Measurement
  • Human Practices
  • Team Members Attributions
  • Achievements

Engineering Success

Design. Build. Test. Learn. Repeat.
  • AMP Plasmid
    • Sushi S1
    • CONGA
  • Lipid-based Nanocarriers
    • Production
    • Fusion
  • Outer Membrane Vesicles
    • Constitutive Promotor
    • Nonspecific Fusion
  • References

The basis of every iGEM project is to implement biological advances with tools from engineering to address global and local issues. We use the principles of engineering to continuously improve and advance our project step by step. Our guiding principle is the DBTL cycle: Design, Build, Test, Learn.

A project starts with the design phase. In this phase, multiple questions regarding the purpose of the project must be answered: What are the specifications? What should the end product look like and what should it be able to do? Next, the biological part or system has to be built and implemented, followed by an extensive testing phase. The functionality of the system’s components is assessed as well as each component is tested for its functionality and utility before moving on to the final stage, where the team can learn from the experimental data and use it to improve the biological system. The team enters the next design phase and the engineering cycle begins anew.

With our project, we aim to combat antibiotic resistance in Pseudomonas aeruginosa using antimicrobial peptides (AMPs). We choose to deliver a potent AMP encoded on a plasmid to the target bacterium using target-specific transport vesicles. Thus, our project is divided into three main tracks: the plasmid design and the two distinct delivery systems: Lipid-based nanocarriers and outer membrane vesicles (OMVs). All three tracks went through the DBTL cycle several times.

AMP Plasmid

CAPTURE utilizes the ability of antimicrobial peptides (AMPs) to bind negatively charged bacterial membranes, disrupting the membrane and eventually causing bacterial cell death. By delivering a plasmid which encodes the AMP specifically to the target pathogen we can circumvent the peptide’s high susceptibility to extracellular proteases and high production costs by forcing the bacteria to produce the peptide themselves.

The Design of the plasmid needs to be precisely adjusted to the target pathogen to ensure synthesis of the peptide only in the malignant bacteria (read more about plasmid specificity in Plasmid Design). Furthermore, the mechanism of action of the AMP needs to be taken into consideration to adjust the cellular localization of the peptide, ensuring the best possible effect.

Cycle 1: Sushi S1

eng-cycle

Design

During the initial planning process of our project, we decided to use an antimicrobial peptide called Sushi S1 which derives from the lipopolysaccharide (LPS)-binding region of Factor C of horseshoe crabs. Our literature research showed that Sushi S1 has one of the lowest minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC) against gram-negative bacteria like E. coli in general [1] and our target pathogen Pseudomonas aeruginosa in particular [2]. By choosing a highly effective AMP, we want to make sure that as much bacteria as possible are killed upon delivery of the plasmid and induction of peptide expression.

In previous publications the functionality of Sushi S1 was only tested by applying the synthesized peptide to the bacteria [3]. It was shown that the peptide binds with high affinity to Lipopolysaccharides (LPS) on the surface of the outer membrane of gram-negative bacteria [4,5]. Since we have not found any papers trying to express Sushi S1 inside the targeted bacterium, we decided to first test what cellular localization of the peptide has the biggest impact on the bacterial growth.

We designed three versions of our plasmid allowing us to test the effect of Sushi S1 at three different locations: in the cytoplasm, the periplasm or the extracellular space. In the framework of iGEM we first focused on experiments with the gram-negative bacterium E. coli.

Build

Using the pET-22b(+) plasmid backbone we cloned three variants of Sushi S1:

  • Cytoplasmic: pelB signal sequence was deleted from plasmid backbone
  • Periplasmic: pelB signal sequence [6]
  • Extracellular: insertion of heat-stable toxin II (HSTII) secretion sequence [7]

For cloning purposes and plasmid amplification we used E. coli Top 10. The nucleotide sequence of Sushi S1 was ordered as a synthesized G-block fragment from IDT. Our cloning strategy for the various plasmids consisted of a combination of restriction digest, Gibson assembly and Site Directed Mutagenesis (get more details about the cloning process in our Notebook).

Test

We performed growth curve experiments in E. coli BL21(DE3) and E. coli BL21(DE3)-pRARE2-LysS and measured OD600 over time after induction of Sushi S1 expression (read more about our growth curve experiments on the Results page). We compared the three localizations with the growth of two different controls (empty pET-22b(+) plasmid and non-induced cultures).

Experimental Setup

Figure 1 Expression of Sushi S1 fused to different localization sequences in E. coli BL21(DE3) OD600 values of bacterial cultures after peptide induction with 0.5 mM IPTG, cultured at 30°C and 200 rpm in LB-medium (n = 2). pelB-Sushi: periplasmic Sushi S1, HSTII-Sushi: extracellular Sushi S1, Sushi: cytoplasmic Sushi S1. Controls: empty pET-22b(+) plasmid and uninduced bacterial cultures.

Our initial findings revealed:

  • Sushi S1 expression inhibited bacterial growth for all three localization sequences at least to some degree.
  • The extracellular version HSTII‑Sushi S1 showed the strongest growth impairment.
  • Induction of the empty pET-22b(+) plasmid also impaired growth. Therefore, a new control was required.

To address the issue of a new control and to verify our results we switched to pET-22b(+) plasmids containing the fluorescent protein mCherry as negative control. At this point we also changed our medium from LB‑medium to Mueller-Hinton broth (MH medium), as further literature research showed that it is recommended for testing antimicrobial susceptibility [8].

Although the results for HSTII-Sushi S1 were reproducible and bacterial growth was significantly reduced compared to mCherry expression, we observed that OD600 values started to increase again after approximately 6 hours.

Experimental Setup

Figure 2 Expression of HSTII-Sushi S1 in E. coli BL21(DE3) OD600 values of bacterial cultures after peptide induction with 0.5 mM IPTG, cultured at 30°C and 200 rpm in MH-medium (n=2). Sushi: extracellular HSTII-Sushi S1. Controls: pET-22b(+) plasmid containing HSTII-mCherry sequence and uninduced bacterial cultures.

To test if we could eliminate this effect, we repeated the experiment in E. coli BL21(DE3) with pRARE2 LysS plasmid. In this strain, Sushi S1 expression inhibited growth for 24 h, but the mCherry control also showed strong growth inhibition.

Learn

From our expression experiments with Sushi S1 in E. coli we can draw two main conclusions:

  • Effectiveness of AMP delivery concept
    Our chosen AMP Sushi S1 reduced bacterial growth at a significant level when produced within the target organism. Although the bacteria started growing again after about 6 hours, we were able to show that the concept of delivering a plasmid encoding a potent AMP to the bacteria works.

  • Optimal peptide localization (signal peptide selection)
    The secreted form of Sushi S1 (HSTII-Sushi S1) showed the strongest impact on bacterial growth. This aligns with literature data, showing that Sushi S1 binds to Lipopolysaccharides (LPS) on outer bacterial membranes.

Based on these findings, we decided to focus on HSTII-Sushi S1 in all following expression experiments in E. coli.

Cycle 2: CONGA

eng-cycle

Design

With the intention to test if growth inhibition can be observed for a longer period of time we replaced Sushi S1 with another antimicrobial peptide.

In the scope of our integrated human practice, we had the chance to talk to Prof. William Wimley, who works on synthetically developing highly potent AMPs (more about our meeting with Prof. Wimley on the Human Practices page). He drew our attention to his most recent publication in which he characterized his new and very effective AMP D-CONGA Q7 [9]. Despite the fact that D-CONGA Q7 consists only of the D-enantiomers of amino acids, he encouraged us to try the peptide’s effectiveness when expressed in the targeted bacteria.

Based on our Sushi S1 results and discussion with Prof. William Wimley, we decided to test the peptide D-CONGA Q7 for our purpose. Since the expression of Conga by E. coli would produce the L-enantiomer of the peptide (L-CONGA), we additionally wanted to test the synthesized version of the peptide comparing its potency to D-CONGA Q7.

For the expression experiments we focused on two localizations: Cytoplasmic and extracellular (HSTII-Conga). However, our primary focus was on the extracellular version, given the results from our Sushi S1 experiments.

To be able to compare the effect of the two AMPs Sushi S1 and Conga, the experimental setup for both peptides was kept identical.

Build

Prof. Wimley kindly provided us with a small sample of his AMP D-CONGA Q7 to compare the effectiveness of both enantiomers of the peptide. L-CONGA was generously synthesized by the group of Prof. Maja Köhn and donated to our team.

For testing the expression of Conga in bacterial cultures we modified existing Sushi S1 plasmids using Site Directed Mutagenesis to insert the synthesized Conga sequence to generate two new constructs:

  • HSTII-Conga for extracellular localization
  • Conga without signal sequence for cytoplasmic localization

To analyze the effect of peptide expression E. coli BL21(DE3) standard strain and pRARE2 LysS strain were transformed with finished plasmids.

Test

Liquid bacterial cultures of E. coli BL21 and Pseudomonas fluorescens were incubated with synthesized Conga peptides and plated on MH plates to examine the impact of the AMP enantiomers on bacterial growth. After incubation with D-CONGA Q7 neither E. coli nor P. fluorescens showed any growth on plates. However, L-CONGA seemed to only impair growth of E. coli to a smaller degree than the D-enantiomer.

Experimental Setup

Figure 3 Conga Killing Assay E. coli BL21(DE3) and Pseudomonas fluorescens cultures were incubated in MH-medium with AMPs for 3 hours at 200 rpm and 37°C or 30°C respectively. 5 µl drops were plated on MH-Agar. D- and L-CONGA final concentration: 40 µM. controls: kanamycin (40 µg/ml and 80 µg/ml respectively), acetic acid (solvent of peptides, 0.025%), control (untreated bacterial cultures).

To analyze the impact of expressed Conga we performed growth curve experiments similar to Sushi S1. After the induction we measured OD600 over time in both E. coli BL21(DE3) standard strain and pRARE LysS containing strain. We used mCherry expression and uninduced cultures as control.

Experimental Setup

Figure 4 Expression of extracellular Sushi S1 and Conga in E. coli BL21(DE3) OD600 values of bacterial cultures after peptide induction with 0.5 mM IPTG, cultured at 30°C and 200 rpm in MH-medium (n=2). Controls: pET-22b(+) plasmid including mCherry and uninduced bacterial cultures.

Similar to the effect that we observed for Sushi S1, Conga inhibited growth for roughly 6 hours. Unfortunately our results of the pRARE LysS strain were inconclusive due to control issues (cultures of mCherry expression showed similar growth inhibition). The comparison between the intracellular and extracellular localization of Conga revealed that the extracellular HSTII-Conga showed stronger inhibition than the cytoplasmic version.

Learn

After testing the efficacy of two highly potent antimicrobial peptides in E. coli, we can conclude that the principle of expressing the AMPs within the targeted pathogen is promising.

  • We observed that the L-enantiomer of Conga effectively inhibited growth of E. coli.
  • Sushi S1 and Conga appeared to have an almost identical effect on bacterial growth. They acted as inhibitors for 6 hours, but lost their effect after a longer period of time.
  • The extracellular localization was more effective for both AMPs.

Due to time constraints, we could only test these two peptides in two different bacterial strains. Further research is needed and could include:

  • Identification of source of the control issues. During our experiments using the E. coli BL21(DE3) strain containing the pRARE2 LysS plasmid, we observed that the synthesis of our control protein mCherry significantly reduced bacterial growth. We suspect that mCherry secretion could cause problems that have negative effects on the viability of the cells. However, we did not have the chance to perform any tests to support this assumption or try the effects of another control protein.
  • Testing AMPs with different mechanisms, e.g. peptides that attack the bacterium from the inside.
  • Exploring alternative plasmid backbones to extend the effectiveness of the AMPs.

Lipid-based Nanocarriers

When we were developing our project idea, one of the main questions was how we could deliver the AMP-encoding plasmid directly to the target bacteria. We needed a system that we could produce relatively easily in the lab and that was capable of taking up DNA and releasing it again at a specific location, and we ultimately opted for lipid-based nanocarriers as one of the two delivery systems we were working on in parallel. Read more about the engineering process of outer membrane vesicles (OMVs) as our alternative system here.

Cycle 1: Producing Lipid-based Nanocarriers

eng-cycle

Design

Lipid-based nanocarriers are vesicular structures that can be synthesized in the lab and have been extensively researched and adapted for drug and gene delivery applications. Their properties, such as lipid composition, size, and surface modifications, can be tailored to enhance encapsulation, stability, and target specificity. There are several sub-categories of lipid nanocarriers, but we focused particularly on liposomes and lipid nanoparticles (LNPs) (see Figure 5). Liposomes (50 - 500 nm) have one or more lipid bilayers, and form a spherical structure to encapsulate the cargo in an aqueous solution. LNPs (100 - 400 nm), on the other hand, have a lipid monolayer and their payload is enveloped by reverse micelles. Their interior does not necessarily have to be aqueous.

Liposome vs LNP

Figure 5: Liposome (left) and Lipid Nanoparticle (Right). Image created with BioRender.com.

Our liposomes and LNPs had to be able to encapsulate DNA and fuse with biological membranes in order to release the payload. In particular, we wanted to use cationic lipids as building blocks for the vesicles, as these would ideally ensure enhanced encapsulation of the negatively charged plasmid DNA and optimized fusion with the negatively charged bacterial membranes. Through research and consultation with experts (see our Integrated Human Practices page), we eventually decided on 2-dioleoyl-3-trimethylammonium propane (DOTAP), a cationic synthetic lipid that is commonly used for lipid-based carrier formulations for the delivery of genetic material. In addition to this, we also decided on using the neutral helper lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), which is often used in combination with DOTAP. However, as DOTAP and DOPE were not yet available to us at the start of the project, we first started our work with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), a phosphatidylcholine that is commonly used as a model lipid for biophysical experiments, as it mimics the mammalian phospholipid composition [10].

Build

We produced liposomes using a method called PVA swelling, followed by extrusion for size control. LNPs were prepared using a simple pipetting-mixing protocol or microfluidic mixing (MFM). The carriers were then tested and characterized in regards to size distribution, lipid composition and plasmid encapsulation efficiency.

Test

After developing the lipid-based nanocarriers, we conducted various tests to evaluate their effectiveness, focusing on size distribution, stability, and plasmid encapsulation efficiency.

Our test phase focused on three critical aspects of our lipid-based nanocarriers: size distribution, stability, and plasmid encapsulation efficiency. The results were both promising and insightful:

We successfully produced Giant Unilamellar Vesicles (GUVs) using PVA swelling and tested different lipid compositions. DOTAP/DOPE ratios of 52:48 and 26:74 showed larger diameters (up to 100 µm) and an enhanced stability compared to other lipid compositions (Figure 6).

PVA Swelling

Figure 6: GUV formation using PVA swelling method with various lipid compositions. (A) 100% POPC, (B) 100% DOTAP, (C) 100% DOPE, (D) DOTAP/DOPE molar ratio 52:48, (E) DOTAP/DOPE molar ratio 26:74, (F) DOTAP/DOPE molar ratio 76:24. GUVs were pipetted off the gel and imaged using transmitted light microscopy. Scale bars represent 100 µm.

GUVs were downsized to Large Unilamellar Vesicles (LUVs) of approximately 200 nm using extrusion (Figure 7). We selected DOTAP/DOPE (52:48) for further experiments due to optimal size and stability.

Extrusion Lipid Ratios

Figure 7: Impact of extrusion on GUVs size and polydispersity for various lipid compositions. GUVs were prepared from different lipid compositions: POPC, DOPE, DOTAP and DOTAP/DOPE molar ratios 52:48, 26:74 and 76:24. Extrusion was performed with a 1 µm pore size polycarbonate membrane. Average diameter was measured by DLS after varying numbers of passages through the polycarbonate membrane.

We successfully encapsulated plasmids into GUVs using PVA swelling with best encapsulation in POPC liposomes.

Extrusion Lipid Ratios

Figure 8: GUVs were prepared with POPC using PVA swelling. The DAPI-stained plasmid was added into the PVA before drying and before adding the lipid solution. Controls were done with DAPI-stained H2O instead of plasmid. Scale bar represents 100 µm. Experiment was done in triplicate.

Plasmid-loaded pLNPs were produced using both simple pipetting-mixing and MFM, with average diameters of 150 nm and 170 nm, respectively.

During the process of pLNP production, we developed a novel, cost-effective Midori Green assay for quantification of DNA-encapsulation. Measured results closely aligned with the established PicoGreen assay, validating our new method (Details on our Measurement page).

PicoGreen vs. Midori Green

Figure 9: Comparison of encapsulation efficiencies using the Midori Green and PicoGreen assays. Encapsulation efficiency was measured in triplicate for pLNPs composed of DOTAP/DOPE/POPC at a molar ratio of 50:30:20. The samples were stained with Midori Green to ensure equal production conditions, and then divided into two batches, with each batch tested using either the Midori Green or PicoGreen assay. (A) The graph shows the mean encapsulation efficiencies from the triplicate measurements. Error bars represent the standard error of the mean (SEM). (B) A direct comparison of the encapsulation efficiencies for individual replicates, tested with both assays. Error bars represent the SEM from technical duplicate measurements.

Moreover, we could show that there is a positive correlation between DOTAP molar ratio and encapsulation efficiency (Figure 10) using PicoGreen assay. This finding highlights the importance of balancing lipid composition and N/P ratio for optimal performance (Figure 10) using PicoGreen assay. This finding highlights the importance of balancing lipid composition and N/P ratio for optimal performance.

It is critical to optimize both the lipid composition and the N/P ratio to achieve maximal encapsulation efficiency while preserving fusion capabilities and minimizing cytotoxicity, which will be discussed further in Cycle 2.

PicoGreen Assay

Figure 10: Efficiency of plasmid encapsulation in pLNPs as determined by the PicoGreen assay. pLNPs were formulated from DOTAP/DOPE/POPC in various molar ratios, specifically 0:30:70, 10:30:60, 20:30:50, 30:30:40, 40:30:30, 50:30:20, 60:30:10, and 70:30:0. The experiment was performed only once.

Overall, these tests confirmed the viability of both liposomes and LNPs as carriers, with significant encapsulation efficiency and stability, particularly with optimized lipid compositions and production protocols.

Learn

During our experiments, it became evident that LNPs are superior in terms of production efficiency compared to liposomes. While liposomes require labor-intensive processes such as PVA swelling and subsequent extrusion for size control, LNPs can be synthesized using simple mixing techniques, resulting in a significantly higher yield of particles.

As a result, we shifted our focus to LNPs, exploring different lipid compositions to optimize size distribution and encapsulation efficiency. Our findings indicated that lipid composition plays a crucial role in determining the performance of the nanocarriers:

LNPs with a higher molar ratio of DOTAP in the lipid composition exhibited increased encapsulation efficiencies, likely due to the cationic nature of DOTAP, which facilitates the interaction with the negatively charged plasmid DNA. POPC, appeared to potentially enhance the stability of the nanocarriers. Its inclusion in the lipid formulation contributed to more consistent size distribution and reduced aggregation over time.

Through further testing, we found that a combination of DOTAP, DOPE, and POPC in a molar ratio of 50:30:20 was an effective formulation. This mixture combined the advantages of each lipid: the high encapsulation efficiency from DOTAP, the membrane fusion properties of DOPE, and the stability imparted by POPC. This optimized composition was subsequently used in our later assays, to achieve robust and reliable results. However, while size, stability, and encapsulation efficiency are critical parameters, they are not the only considerations for the intended therapeutic application. An equally important factor is the fusion capability of the nanocarriers:

  • Targeted Fusion: Ensuring effective fusion with the target bacterium, P. aeruginosa, is essential for delivering the therapeutic payload to the infection site.
  • Minimizing Off-Target Effects: Avoiding unwanted fusion with human lung cells, which are in close proximity to the bacteria, is crucial to prevent potential cytotoxicity and ensure the safety of the delivery system.

This insight led us to the next phase of our project, where we will investigate the impact of different lipid compositions on the fusion behavior of LNPs, aiming to fine-tune the formulation for optimal therapeutic efficacy.

Cycle 2: Fusion

eng-cycle

Design

The goal of Cycle 2 is to optimize our LNPs to selectively fuse with P. aeruginosa while avoiding fusion with human lung cells. This targeted fusion is essential for delivering therapeutic payloads directly to pathogens in the lungs, minimizing off-target effects and cytotoxicity to human cells. The LNPs should demonstrate high fusion efficiency with P. aeruginosa while exhibiting low interaction and fusion with human lung cells, and should not cause significant cytotoxicity at therapeutically relevant concentrations. To achieve this selective fusion, we employed a targeting mechanism inspired by the interaction of the bacterial lectin LecA on P. aeruginosa with the glycosphingolipid Gb3 on mammalian cells. Including Gb3 in the LNP formulations could enhance binding to bacteria that express such lectins, allowing selective targeting.

Build

The LNPs were formulated from DOTAP/DOPE/POPC in a molar ratio of 50:30:20, based on findings from Cycle 1. Gb3 was integrated into the lipid composition to enhance specificity for P. aeruginosa. This was inspired by the binding mechanism of LecA on P. aeruginosa with Gb3, aiming to enhance selectivity for bacterial cells [11,12,13]. To test the interaction, we added Atto647N-labeled DOPE, a red-fluorescent lipid, into the lipid composition, while the bacteria were labeled with GFP. This setup allowed us to observe interactions using confocal microscopy. Additionally, we conducted cytotoxicity assays (MTT and SYTOX Green) on human lung tissue cells and performed a fusion assay to evaluate the uptake of a GFP plasmid from pLNPs into human cells.

Test

Fusion Assays with P. aeruginosa aimed to observe interaction and fusion behavior between engineered LNPs and the target bacteria using fluorescence microscopy. Co-localization of fluorescence signals (Figure 11) for LNPs (red) and bacteria (green) suggests that our Gb3-incorporated LNPs can successfully target P. aeruginosa. This provides initial validation of our design strategy. However, further quantitative analysis is needed to determine the efficiency and specificity of these interactions compared to control LNPs without Gb3.

Confocal Microscopy Image

Figure 11: Co-localization of lipid nanoparticles (LNPs) and P. aeruginosa in liquid culture. Confocal microscopy image showing the interaction between fluorescently labeled LNPs and P. aeruginosa after 30 minutes of incubation. LNPs were formulated with the fluorescent lipid Atto 647N (red), while P. aeruginosa expressed GFP in its membrane (green). Merged image demonstrating co-localization of LNPs and bacteria. Scale bar represents 10 µm.

The MTT assay evaluated the viability of A549 cells after treatment with various concentrations of liposomes, eLNPs, and pLNPs. The results indicated cytotoxicity with liposomes, while LNPs showed a mild dose-dependent cytotoxic effect only at higher concentrations​.

MTT Assay

Figure 12: MTT assay assessing A549 cell viability after treatment with varying concentrations of pLNPs. (A) pLNPs were formulated using a DOTAP/DOPE lipid composition in a 52:48 molar ratio. (B) pLNPs were formulated using a DOTAP/DOPE/POPC lipid composition in a 50:30:20 ratio. A549 cells were cultured in 100 µL DMEM, representing 100% cell viability. Controls included water dilutions (30 µL H2O + 70 µL DMEM and 60 µL H2O + 40 µL DMEM) to assess any effects of the solvent, with a 100 µL H2O positive control, where complete cell death is expected. Cells were then treated with increasing volumes of pLNPs (3 µL, 6 µL, 15 µL, 30 µL, and 60 µL) suspended in water. Error bars represent the standard error of the mean (SEM) from duplicate measurements.

The SYTOX Green assay was used to assess membrane integrity of lung cells following treatment with LNPs. Only a minimal impact of eLNPs on cell membrane integrity was observed, affirming their relative biosafety compared to liposomes​.

SYTOX Green Assay

Figure 13: SYTOX Green assay assessing A549 cell viability after treatment with varying concentrations of eLNPs. eLNPs were formulated from DOTAP/DOPE/POPC in a molar ratio of 50:30:20. A549 cells were cultured in 100 µL DMEM, representing 0% cell cytotoxicity. Water controls were included to determine the effects of dilution, with treatments of 30 µL H2O + 70 µL DMEM and 60 µL H2O + 40 µL DMEM. A positive control with 100 µL H2O was used, representing 100% cytotoxicity. Cells were then exposed to increasing volumes of eLNPs (6 µL, 15 µL, 30 µL, and 60 µL) suspended in water. Sample size n = 3. Error bars represent the population standard deviation.

The eGFP Fusion assay investigated the potential fusion and plasmid uptake by human lung cells. The data indicated minimal fusion of pLNPs with A549 cells, supporting the hypothesis that these LNPs are less likely to fuse with mammalian cells, which is desirable for biosafety.

Fusion Assay Plot

Figure 14: Fluorescence intensity measurement of eGFP expression in A549 cells following treatment with pLNPs. All fluorescence intensities were normalized by subtracting the background measurement obtained from phenol red-free DMEM. The cells were treated with DMEM as a negative control, lipofectamine-mediated transfection with the eGFP plasmid as a positive control, 3 µg plasmid DNA alone, or different concentrations of pLNPs containing the eGFP plasmid, respectively. The pLNP formulations consisted of either DOTAP/DOPE in a molar ratio of 52:48 (A) or DOTAP/DOPE/POPC in a molar ratio of 50:30:20 (B). ND indicates non-detectable fluorescence intensities, meaning they were below the background level. Experiment was conducted in duplicate. Error bars represent the standard error of the mean (SEM).

Learn

Our preliminary findings indicated some interaction between the LNPs and P. aeruginosa; however, further investigation is required to validate the efficacy and specificity of the targeting mechanism. These preliminary findings were constrained by time limitations and the need for collaboration with a partner laboratory, as our team was not certified to handle the S2-level pathogen P. aeruginosa, while the partner lab possessed the necessary certification. Future work should focus on optimizing the incorporation of Gb3 into the lipid formulation to enhance selectivity for the target bacterium, while ensuring stability and minimizing off-target effects.

Our pLNPs exhibited no significant cytotoxicity at lower concentrations, but a dose-dependent increase in cytotoxicity was observed at higher concentrations, highlighting the necessity for careful dose optimization in therapeutic applications. Importantly, the minimal fusion observed with A549 cells in the eGFP assay suggests that the current pLNP formulation is effective in minimizing unintended interactions with human cells.

The lipid composition should be further optimized to minimize cytotoxic effects. Cytotoxicity can be assessed by testing different lipid formulations and determining which lipids, and at what concentrations, contribute most to the formulation’s toxicity. Subsequently, the formulation can be further modified, carefully balancing other critical properties such as fusogenicity, specificity, and encapsulation efficiency against potential adverse effects. This process requires extensive data collection. Computational modeling may be employed to reduce the data needed, identifying correlations between lipid ratios and the desired LNP properties.

Outer Membrane Vesicles

Cycle 1: Constitutive Promoter for eCPX expression

eng-cycle

Design

In the development phase of our project, we realized that our liposomal delivery system would entail significant production costs that could harm overall market adoption. To circumvent this bottleneck, we decided to look into a promising alternative for plasmid delivery: outer membrane vesicles (OMV).

OMVs are vesicles produced by Gram-negative bacteria [14], among other things encapsulating genetic material to mediate horizontal gene transfer, this fact alone makes ideal candidates for It forms the basis for our efficient and cost effective delivery solution. We have used the E. coli BL21(DE3) omp8 strain (Human Practices) , a variant of the BL21(DE3) strain with the following deletions: OmpA, OmpC, OmpF and LamB, in order to isolate a larger amount of OMVs that display a greater proportion of our outer membrane protein eCPX [15].

eCPX or the enhanced circularly permuted OmpX, derived from the native E. coli OmpX, was genetically fused to a SpyTag peptide. This construct is controlled via an inducible LacI promoter and allows us to functionalize our OMVs with multiple phage tail proteins that act as targeting ligands(see our human practices). This approach is tailored to maximize targeting efficiency while limiting specificity towards P. aeruginosa.

We hypothesized that when using an inducible promoter to express eCPX-SpyTag, OMVs produced pre-induction without the eCPX-SpyTag alongside the OMVs displaying eCPX-SpyTag would lead to a heterogenous OMV population that would be impossible to separate. We decided to circumvent this issue by replacing the inducible promoter with one of 2 constitutive promoters; LacIq (BBa_K3257003) and AmpR (BBa_K2796022).

Build

We were able to clone both promoters into our plasmid via PCR using primers containing our promoter regions. This was followed by a Gel-electrophoresis and subsequent transformation into E. coli BL21(DE3) omp8 strains. We then evaluated all 3 promoters in terms of cell viability by measuring growth curves, after which, we isolated OMVs from the aforementioned cultures to measure protein expression and functionality by performing standardized SDS Gels and Western Blots.

Test

Overexpression of outer membrane proteins typically overloads protein translocation machinery [16], leading to cell stress and sub-optimal bioproduction capabilities. However, overexpression of eCPX has been shown to not inhibit cell viability [17]. Our growth curves confirmed these findings, however the Amp promoter seemingly showed higher reduction of cell viability compared to the LacIq promoter.

growth curve lac amp

Figure 15: Growth Curve showing growth rates of E. coli Omp8 strains containing either pTrc99-LacIq-eCPX-SpyTag OR pTrc99a-Amp-eCPX-SpyTag. Both cultures were grown in 50 mL LB alongside kanamycin and ampicillin, OD600 was measured once an hour for 29 hours, 1:1, 1:10, 1:20 dilutions were performed depending on measured OD600

However, cell viability does not give the full picture with respect to protein expression. Therefore, we performed SDS-PAGEs followed by Western Blots with OMVs that had been isolated with optimized protocols in order to maximize both OMV production and protein expression (See our Results).

Our SDS Gels and Western Blots illustrated that the LacIq promoter lead to higher levels of eCPX-SpyTag expression and functionality compared to the Amp promoter, however comparing it to the inducible Trc Promoter (induced with 100 µM IPTG) the same level of protein expression was not achieved.

OMV SDS gel

Figure 16: SDS-PAGE of OMVs Incubated with SC-P2 Post-CoomassieStaining. OMVs exhibiting eCPX-SpyTag(eCPX-ST) were functionalized with SpyCatcher-Phagetail2 (SC-P2). Samples adjusted to protein concentration of 10 µg for all OMVs, except Amp-OMVs, which were adjusted to 15 µg.

OMV western blot gel

Figure 17: Western blot of OMVs incubated with SC-P2, bound by His-antibodies. Visualized with chemiluminescence. OMVs diluted to 10 µg total protein per sample (Exception : Amp-OMVs w/15 µg per sample), 1.5 µg P2 used per sample.

Learn

We concluded this cycle with the knowledge that the LacIq constitutive promoter used in place of the inducible Trc Promoter could achieve similar levels of protein expression without the need for induction, this however wasn’t the case with what was observed with the Amp promoter.

Our results could be partly attributed to the reduced cellular impact associated with eCPX overexpression compared to other outer membrane proteins [17] and partly due to the higher expression levels associated with LacIq compared to the Amp promoter.

Cycle 2: Nonspecific fusion

eng-cycle

Design

OMVs can mediate horizontal gene transfer among other processes (See our Descriptions). Our goal was to engineer OMVs to deliver AMP coding plasmids while displaying specificity towards P.aeruginosa. This specificity could be achieved through the functionalization of phage-tail proteins on the OMV surface. In order to ascertain the efficiency of OMV-mediated plasmid delivery, numerous “non-specific fusion” experiments were carried out with OMVs that had not been functionalized with targeting ligands.

Build

OMVs encapsulating plasmids with diverse copy numbers were extracted from various donor strains using standard extraction protocol for use in fusion experiments (See our Notebook/Protocols).

Test

We conducted “non-specific” fusion experiments with differing OMV concentrations alongside dissimilar recipient strains using divergent experimental protocols[18][19][20], in an effort to measure the efficiency of OMV-mediated transformation (See more on the Results).

OMVs were screened for plasmid encapsulation via DNAse coupled PCR. (See more on the Results) prior to their use within the fusion experiment. After numerous attempts at non-specific fusion, we were not able to transform our recipient E. coli BL21(DE3) strains using OMVs encapsulating plasmids. However, an interesting phenomenon was observed when E. coli BL21(DE3) strains were incubated with OMVs produced by a strain carrying an ampicillin resistance plasmid. Bacteria would seemingly grow when incubated overnight in LB with ampicillin only to “regain” susceptibility on further inoculations in the same media.

Learn

After having observed the same result in experimental repeats, we thought about possible reasons. Our final hypothesis hinges on the periplasmic localisation of β-Lactamase[21], the enzyme responsible for degrading β-Lactams such as ampicillin. We speculate that OMVs encapsulating β-Lactamase alongside the plasmid, are able to degrade the ampicillin present, thereby allowing bacterial growth without plasmid uptake.

Design

This hypothesis was thought up by eliminating possibilities such as contaminations through heavy use of controls. However, in order to prove our hypothesis, we had to perform new experiments that would definitively prove that false-positive results arose out of an encapsulation of β-Lactamase.

Build

In order to restrict the cause of antibiotic degradation to OMVs, the fusion protocol was modified in order to remove OMVs prior to incubation in LB with ampicillin. After stationary incubation of bacteria and OMVs, the solution was centrifuged down in order to remove the OMVs present within the supernatant. The bacterial pellet was then resuspended and incubated further in LB media containing antibiotics.

A second experiment was required in order to verify the ampicillin degrading capabilities of OMVs, wherein OMVs would be incubated overnight in LB containing antibiotics, followed by the inoculation of a bacterial culture in the same media.

Test

No growth was observed at all proving the susceptibility of the wild type strain.

In contrast, growth was observed when a wild type culture was inoculated in media that had previously been inoculated with OMVs indicating the lack of ampicillin within the media post-OMV incubation. OMV controls showed no change in optical density; were not contaminated.

Learn

Taken together, these results confirm our theory regarding β-Lactamase encapsulation and the ability of OMVs to degrade ampicillin present in the media. Unfortunately, these results invalidate our ability to perform further “non-specific fusion” experiments using our isolated OMVs, due to the exclusive encapsulation of plasmids carrying ampicillin resistance.

After discussing with our primary investigators, we devised a strategy in order to circumvent ampicillin resistance that relied upon replacing the β-Lactamase gene with one coding for chloramphenicol acyltransferase Due to the fact that chloramphenicol acyltransferase inhibits chloramphenicol present within the cytoplasm, we would see lower encapsulation of antibiotic degrading enzymes into the OMVs. However, due to time constraints, we were not able to integrate these improvements into our project.

References

[1] Li P, Sun M, Wohland T, Yang D, Ho B, Ding JL. Molecular Mechanisms that Govern the Specificity of Sushi Peptides for Gram-Negative Bacterial Membrane Lipids. Biochemistry. 2006 Sep 1;45(35):10554–62.

[2] Yau YH, Ho B, Tan NS, Ng ML, Ding JL. High Therapeutic Index of Factor C Sushi Peptides: Potent Antimicrobials against Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2001 Oct;45(10):2820–5.

[3] Li A, Lee PY, Ho B, Ding JL, Lim CT. Atomic force microscopy study of the antimicrobial action of Sushi peptides on Gram negative bacteria. Biochim Biophys Acta BBA - Biomembr. 2007 Mar;1768(3):411–8.

[4] Tan NS, Ng MLP, Yau YH, Chong PKW, Ho B, Ding JL. Definition of endotoxin binding sites in horseshoe crab Factor C recombinant sushi proteins and neutralization of endotoxin by sushi peptides. FASEB J. 2000 Sep;14(12):1801–13.

[5] Leptihn S, Guo L, Frecer V, Ho B, Ding JL, Wohland T. One step at a time: Action mechanism of Sushi1 antimicrobial peptide and derived molecules. Virulence. 2010 Jan;1(1):42–4.

[6] Sockolosky JT, Szoka FC. Periplasmic production via the pET expression system of soluble, bioactive human growth hormone. Protein Expr Purif. 2013 Feb;87(2):129–35.

[7] Freudl R. Signal peptides for recombinant protein secretion in bacterial expression systems. Microb Cell Factories. 2018 Dec;17(1):52.

[8] NCCLS Approved Standard: ASM-2, Performance Standards for Antimicrobic disc Susceptibility Tests, 2nd ed., National Committee for Clin. Lab. Standards (1979).

[9] Ghimire J, Hart RJ, Soldano A, Chen CH, Guha S, Hoffmann JP, et al. Optimization of Host Cell-Compatible, Antimicrobial Peptides Effective against Biofilms and Clinical Isolates of Drug-Resistant Bacteria. ACS Infect Dis. 2023 Apr 14;9(4):952–65.

[10] Avanti Research [Internet]. [cited 2024 Oct 1]. 16:0-18:1 PC (POPC) | 26853-31-6 | Avanti. Available from: https://avantiresearch.com/product/850457

[11] Eierhoff T, Bastian B, Thuenauer R, Madl J, Audfray A, Aigal S, et al. A lipid zipper triggers bacterial invasion. Proceedings of the National Academy of Sciences. 2014 Sep 2;111(35):12895–900.

[12] Schubert T, Sych T, Madl J, Xu M, Omidvar R, Patalag LJ, et al. Differential recognition of lipid domains by two Gb3-binding lectins. Sci Rep. 2020 Jun 16;10(1):9752.

[13] Brandel A, Aigal S, Lagies S, Schlimpert M, Meléndez AV, Xu M, et al. The Gb3-enriched CD59/flotillin plasma membrane domain regulates host cell invasion by Pseudomonas aeruginosa. Cell Mol Life Sci. 2021 Apr 1;78(7):3637–56.

[14] Schwechheimer, C., Kuehn, M. Outer-membrane vesicles from Gram-negative bacteria: biogenesis and functions. Nat Rev Microbiol 13, 605–619 (2015).

[15] Thoma, J., Manioglu, S., Kalbermatter, D. et al. Protein-enriched outer membrane vesicles as a native platform for outer membrane protein studies. Commun Biol 1, 23 (2018).

[16] Wagner S, Baars L, Ytterberg AJ, Klussmeier A, Wagner CS, Nord O, et al. Consequences of membrane protein overexpression in Escherichia coli. Mol Cell Proteomics. 2007 Sep;6(9):1527–50.

[17] Rice JJ, Daugherty PS. Directed evolution of a biterminal bacterial display scaffold enhances the display of diverse peptides. Protein Eng Des Sel. 2008 Jul;21(7):435-42. doi: 10.1093/protein/gzn020. Epub 2008 May 13. PMID: 18480093; PMCID: PMC2427320.

[18] Somdatta Chatterjee, Ayan Mondal, Shravani Mitra, Sulagna Basu, Acinetobacter baumannii transfers the blaNDM-1 gene via outer membrane vesicles, Journal of Antimicrobial Chemotherapy, Volume 72, Issue 8, August 2017, Pages 2201–2207,

[19] Li P, Luo W, Xiang TX, Jiang Y, Liu P, Wei DD, Fan L, Huang S, Liao W, Liu Y, Zhang W. Horizontal gene transfer via OMVs co-carrying virulence and antimicrobial-resistant genes is a novel way for the dissemination of carbapenem-resistant hypervirulent Klebsiella pneumoniae. Front Microbiol. 2022 Dec 1;13:945972. doi: 10.3389/fmicb.2022.945972. PMID: 36532464; PMCID: PMC9751880.

[20] Tran, F., Boedicker, J.Q. Genetic cargo and bacterial species set the rate of vesicle-mediated horizontal gene transfer. Sci Rep 7, 8813 (2017).

[21] Oana Ciofu, Terry J. Beveridge, Jagath Kadurugamuwa, Jan Walther-Rasmussen, Niels Høiby, Chromosomal β-lactamase is packaged into membrane vesicles and secreted from Pseudomonas aeruginosa, Journal of Antimicrobial Chemotherapy, Volume 45, Issue 1, January 2000, Pages 9–13,

gitlab.igem.org/2024/freiburg | Creative Commons Attribution 4.0 License