R

e

s

u

l

t

s

RESULT


1 Screening of suitable hypoxia-inducible promoters


1.1 Construction of plasmids and characterization of HIPs


Escherichia coli Nissle 1917 (EcN), as a probiotic, has tumour-targeting properties to some extent. So, our aim was to enhance this tendency by specifically targeting EcN to the hypoxic tumour microenvironment (TME). Therefore, we focused on hypoxia-inducible promoters (HIPs), hoping to achieve explicit hypoxia targeting of EcN.


HIPs are regulated by the Fumarate and Nitrate Reduction regulator (FNR)1^1. Under hypoxic conditions, FNR homodimerizes through a 4Fe–4S2+ cluster and binds to a specific DNA site termed ‘FNR box’. Upon its binding, the homodimer interacts with the RNA polymerase binding to TATA box at -10 region, thus activating transcription (Fig. 3). Oxygen can inhibit this process. Previous study2^2 has shown that pPepT, as an endogenous HIP of EcN, can produce a large amount of expression under hypoxic conditions. Mengesha et al3^3 made improvements to the pPepT from Salmonella by mutating its FNR box and TATA box, resulting in an enhanced hypoxia-responsive curve.


Figure 2

Figure 1 | Mechanism of pPepT. FNR homodimer binds to FNR binding site as it interacts with RNA polymerase to activate transcription.


Thus, on the base of wild type PepT promoter, we strived to change the intensity of its expression by mutating TATA box, mutating FNR box, and changing the RBS. We used the PJUMP41-2A (sfGFP) plasmid (BBa_J428365, this plasmid referred below are abbreviated as O12, as it corresponds to the O12 well on the biobrick) as our vector This plasmid is characterized as a low-copy plasmid with spectinomycin resistance and carries the BBa_J23100 promoter along with the sfGFP sequence. Through reverse PCR and Gibson assembly (details are shown in “Engineering”), we successfully replaced the J23100 promoter and RBS with the following 10 promoters + RBS (see BBa_K4713116 - BBa_K4713125):


Table 1 | The promoter + RBS sequences used in screening.

Table 1

P.S. WT: wild type. FF+20 variant is also an FNR-binding-dependent promoter, which used for parallel comparison.


We used sfGFP as the fluorescence output and tested the intensity of 10 variants. In order to achieve normalization of fluorescence intensity, the empty plasmid was used as a blank control, and the ratio of the intensity of sfGFP fluorescence controlled by BBa_J23100 under normoxic and anaerobic conditions was used to explain the insufficient folding and weak fluorescence of sfGFP under anaerobic conditions (coefficient R in formula)


Relative Expression level was analyzed with the following formula:

Relative Expression Level=Fluorescence(Sample)Fluorescence(Blank)OD(Sample)OD(Culture Medium)×R{Relative \ Expression \ Level} = {{Fluorescence(Sample)-Fluorescence(Blank)} \over {OD(Sample) - OD(Culture \ Medium)}} \times R
Under normoxia condition,R=1; while under anaerobic conditions,Under \ normoxia \ condition, R = 1; \ while \ under \ anaerobic \ conditions,
R=expression level of O12 undernormoxic conditionsexpression level of O12 under anaerobic conditionsR = {{expression \ level \ of \ O12 \ under normoxic \ conditions} \over {expression \ level \ of \ O12 \ under \ anaerobic \ conditions}}

The result was shown in Fig. 2.


Figure 2

Figure 2 | Characterization results of 10 HIP variants. (a) Outcome of relative sfGFP / OD600 nm. N = 8 biological replicates. Data are mean ± SEM. (b) Fold changes of biological replicates. N = 8 biological replicates. Data are mean ± SEM.


Our results show that FF+20 variant exhibits the strongest sfGFP fluorescence under anaerobic conditions, although it also has the highest baseline expression level. Mutating the TATA box enhances expression levels under anaerobic conditions, but it also leads to increased expression under normoxic conditions. Similar effects are observed when changing the RBS. However, altering the FNR box did not result in significant expression distinction.


Meanwhile, we acknowledged that our characterization was conducted on a macroscopic scale, and some measurement errors may be inherent. Therefore, we aimed to observe individual or small groups of cells at the microscopic level to obtain more precise data. To achieve this, we employed microfluidic technology.


1.2 Microfluidics for Precise Characterization at the Single-cell Scale


In order to measure fluorescence intensity with single cell accuracy at precise oxygen concentrations, we specially designed a microfluidic chip. With the help of Prof. Chunxiong Luo, we designed a chip that can simultaneously measure the fluorescence intensity of 4 E. coli strains under 6 oxygen concentration gradients4^4, as shown in Fig. 3.


Figure 2

Figure 3 | Schematic diagram of microfluidic chip. The above figure shows the details of the culture layer (Mold 1) and loading layer (Mold 2). The culture layer (Mold 1) is composed of 3 masks on the left, and the loading layer (Mold 2) on the right is composed of 2 masks. To clearly show the detailed pattern of each layer of the two molds, the size in the figure does not represent the true proportion of the structure. All our masks are drawn using L-edit software, and the tdb format file is also uploaded in Gitlab.


We have made the microfluidic chip according to the drawing, and the real picture is shown in Fig. 4.

Figure 4

Figure 4 | Actual diagram of microfluidic chip. (a) The actual effect of the microfluidic chip. (b) the chip under the microscope. We use the lithography machine and coating machine in Chunxiong Luo's lab for chip fabrication.

We demonstrated the viability of the microfluidic chip by verifying the presence of an oxygen concentration gradient and the capture of E. coli by the trap chamber. As shown in Fig. 5a, when the mixture was changed from air to nitrogen air at 10 minutes, the fluorescence intensity was found to be increased, and the 6 channels were different. The following data (Fig. 5b - c, Video 1) further confirms the reliability of our chips


Figure 5

Figure 5 | On-machine test of our microfluidic chip. (a) The oxygen concentration changes over time, switching from total air to air nitrogen mixture at 10 minutes. (b - c) Green fluorescent of E. coli in a trap chamber.


Figure 5

Video 1 | Sample loading process of our microfluidic chip. This video was captured under a microscope.


Unfortunately, we didn't have enough time to apply our microfluidic chip on formal measurement. However, we believe that microscopic result will be similar to the macroscopic result.


2 Success on construction of a hybrid promoter sensing both hypoxia and high lactate


In order to reduce the escape rate and improve targeting to the tumour microenvironment, our initial idea was to build an AND-gate system to sense hypoxia and high lactate simultaneously. Fig. 6a shows one of the traditional AND-gate systems5^5. This system involves two signal inputs (Ara and aTc), which produce protein sicA and invF respectively that can bind and act as a transcriptional activator to initiate the expression of downstream rfp gene. We initially wanted to imitate this AND-gate structure and replace the downstream output with the necessary gene (asd) for EcN. The asd gene expresses aspartate semialdehyde dehydrogenase, which is required for the biosynthesis of lysine, threonine and methionine. Deletion of asd causes bacterial cell wall rupture and death. Supplementation with diaminopimelic acid (DAP) can promote the growth of asd knockout strains. Notably, DAP cannot be produced or metabolized from the host cell environment, making it an ideal strategy to reduce escape rates. High lactate is sensed through the native lldPRD lactate operon of EcN6^6. But we are worried that this AND-gate structure may bring a greater expression burden to EcN.


However, after gaining a deeper understanding of the structure of the lactate operon, we came up with a fantastic idea. Because the principle of the lactate operon (Fig. 6b) is that when lactate is low, lldR acts as an inhibitory protein and inhibits promoter expression by binding to the O1 and O2 sites. When the lactate concentration rises, lactic acid molecules trigger an allosteric reaction by binding to the lldR protein, making the inhibitory effect disappear. Additionally, the lldR protein that binds lactic acid molecules can also bind to the O1 site and act as a transcription activator. Therefore, we wanted to replace the promoter between the O1 and O2 sites with our pPepT (Fig. 6c), allowing it to achieve an AND-gate-like structure and sense hypoxia and high lactate simultaneously. Facts have proved that we successfully constructed this type of promoter through the following experiments.


Figure 6

Figure 6 | Iteration of project model. (a) shows a traditional AND-gate architecture, based on chaperone proteins. (b) shows our original idea, using the AND-gate structure to realize the perception of hypoxia and high lactate. (c) is a more elegant structure that has been proven to successfully achieve the equivalent of an AND-gate through a compact hybrid promoter.


2.1 Knockout of asd gene in EcN

To enable EcN to respond to downstream output from the bybrid promoter, we deleted the asd gene of EcN. The two-plasmid system of Yang et al 7^7 8^8 was applied for gene knockout of EcN. Plasmids of pEcCas and pTarget were the gift from Prof. Changtao Jiang. The N20 sequence was designed via chopchop (chopchop.cbu.uib.no).


Through this method, the asd gene was successfully knocked out in four EcN strains (EcN ∆asd 1 ~ 4). (Fig. 7a) By exogenously supplementing DAP at different concentrations (Fig. 7b), we found that concentrations of 50 μg/ml and above could enable the survival of asd knockout strains.


Figure 7

Figure 7 | asd knockout results of EcN. (a) Nucleic acid electrophoresis results of colony PCR. Compared with the wild type, EcN ∆asd showed a shorter PCR band, consistent with the result of the designed knockout of 975 bp. (b) DAP exogenous supplementation results. 50 μg/ml is a concentration suitable for EcN ∆asd growth.


2.2 Hybrid promoter responds to hypoxia and high lactate


Before constructing the hybrid promoter, we imagined two failure situations (Fig. 8a): first, the lldR protein binding at the O1 site can cover up the FNR binding site of PepT, so that the FNR protein cannot bind to the FNR box to initiate downstream transcription due to steric hindrance (condition 1); second, as the distance between promoters O1 and O2 lengthens (pPepT is 19 bp longer than the original promoter), the active lldR protein bound to the O1 site cannot activate transcription (condition 2).


Figure 8

Figure 8 | Rational design of hybrid promoters. (a) According to the principle of two promoters, we may encounter two problems when combining. The first one is that FNR protein cannot bind normally due to steric hindrance effect. The second is that the distance between the promoter and O1 will affect the intensity of induced activation. (b) Based on this, we designed a total of 24 versions of promoters.


Therefore, we constructed 6 versions of the plldR operon through shortening or lengthening the distance between O1 and O2. Combined with 4 variants of pPepT (BBa_K4713111 to BBa_K4713114), a total of 24 candidates was constructed (Fig. 8b). We replaced the BBa_J23100 promoter of O12 plasmid with our candidate. In order to improve the transformation efficiency (the transformation efficiency of EcN ∆asd is about one-tenth that of EcN), we replaced the original LB medium with SOC medium and appropriately increased the supplementation amount of DAP (100 μg/ml). All promoters were successfully constructed, but only 14 candidates were successfully transformed into EcN ∆asd (the remaining 10 were also successfully transformed recently, but have not yet been characterized). Subsequently, we evaluated the promoter strength under different oxygen (normoxic or anaerobic) and lactate concentrations (0, 0.1 mM, 1 mM or 10 mM), with the results showing as follows:


Figure 9

Figure 9 | Heatmap of hybrid promoter intensity under different conditions. We defined the intensity (expressed as OD600nm) of each variant under normoxic and lactic acid-free conditions as 1, and the relative values under other conditions were obtained by division. N = 3 biological replicates.


It can be seen from the results that appropriately shortening the distance between O1 and O2 (version S1, S3) shows a better response. We finally selected the best-performing promoter S3-FA (BBa_K4713008), which exhibited both low expression at baseline and high expression under conditions of low oxygen and high lactate.


At the same time, due to the low expression of endogenous lldR protein, the promoter's response to lactate concentration is weak, resulting in a certain escape expression at the baseline level. In order to further reduce this expression, we also transferred the lldR overexpression plasmid into EcN ∆asd, and inserted the T5 promoter and lldR sequence into the vector pET28a through gibson assembly, so that it can induce the controllable expression of lldR through IPTG. The EcN ∆asd co-transformed with pET28a_T5-lldR and O12-plldR-J23117 (this plasmid replaces the BBa_J23100 promoter with the BBa_K1847008, a lactate-responsive promoter, whose promoter sequence between O1 and O2 is BBa_J23117) significantly increased the level of lldR protein after induction of expression with IPTG, thereby inhibiting the expression of the plldR promoter, manifested by a significant decrease in OD600nm (Fig. 10).


Figure 10

Figure 10 | Overexpression result of lldR protein. Using different concentrations of IPTG to induce lldR expression significantly reduced the growth of the strain, and this inhibitory effect was dose-dependent. N = 3 biological replicates.


Unfortunately, due to time limits, we did not experiment with the effect of expressing lldR on our hybrid promoter, however, we believe that such expression could significantly reduce baseline levels of escape expression.


3 Expressing vesicle proteins enables precise spatial positioning


After the construction of hybrid promoter, our engineered bacteria had the ability to colonize specifically in the tumour tissues. According to our plan, the next step we need to find a method that can detect the location of the presence of bacteria. Bourdeau et al9^9 genetically modified the vesicular protein from a photosynthetic autotrophic bacterium cyanobacterium Anabaena flos-aquae and transformed it into E. coli BL21(AI) to successfully express the vesicular protein, which could be detected by medical ultrasound probes. However, this article still has the shortcoming that the bacteria can only reach the tumour by in situ injection. The ability of engineered bacteria to self-localize tumours is necessary if this method is to be applied to clinical treatment.


Figure 11

Figure 11 | Ultrasound approach tracks engineered bacteria. 10^{10} Bourdeau et al. 9^9 genetically engineered bacteria to express what they term acoustic response genes (ARG), which encode the components of hollow structures called gas vesicles that scatter sound waves and generate an echo that can be detected by ultrasound.


Fortunately, the scheme proposed by the Peking iGEM team to target the tumour microenvironment by engineering bacteria can make up for this deficiency. We hypothesized that ARG could be introduced into the engineered bacteria containing and induce its expression vesicles. Since only the engineered bacteria colonized in the tumour tissue could survive, the signal would only be detected in the tumour tissue when detected by a medical ultrasound probe.


To prove this idea, pET28a_T7-ARG1 plasmid (addgene #106473, purchased from Miaoling Biology) was transformed into BL21(DE3) and BL21(AI). Vesicle protein expression was induced according to the method of Bourdeau et al9^9, and a range of IPTG concentrations (0.1 mM, 0.4 mM, 1 mM, 10 mM) were used. As a negative control, BL21(DE3) transduced with pET28a_T7-eGFP (gift from A.P. Qingsong Wang) was characterized in the same way, which could only express GFP after induction.


After centrifuging for 2h at 300g, the results are shown as Fig. 11. The results for other induction concentrations are similar and will not be shown here.


Figure 12

Figure 12 | Culture centrifugation results. From the pictures, we can find that although the BL21(AI) transformed with ARG1 plasmid has little cells obviously floating on the upper layer, the bacterial liquid is still turbid; as a control, the bacteria containing pET28a-eGFP completely sink to the bottom. However, no similar phenomenon has been observed in BL21(DE3).


Then, we used a transmission electron microscope (TME) (JEOL, JEM-F200) to observe the sample, and the results are as follows:


Figure 13

Figure 13 | Representative TEM images of characterization. Compared with the control (a), aggregated bubble-like structures can be seen in the E. coli BL21(AI) cells transformed with pET28a-T7-ARG1 plasmid (b) and E. coli BL21(DE3) transformed with pET28a-T7-ARG1 plasmid (c), which are vesicle proteins.


After confirming that the pET28a_T7-ARG1 plasmid was available, we managed to express vesicle proteins in EcN. Considering the lack of T7RNA polymerase in EcN, we replaced the T7 promoter with the T5 promoter through enzyme digestion and ligation, and transformed it (pET28a_T5-ARG1) into EcN through through electroporation. Induction was characterized in a similar way, except that we used 5 μM IPTG for induction and grew 150 ml cultures. The results are as follows:


Figure 14

Figure 14 | Results after one day of natural sedimentation of the culture. Most of the bacteria have still not settled to the bottom, and a suspicious floating bacterial film can be seen where the red arrow points.


Figure 15

Figure 15 | Representative TEM images of characterization. Compared with the control (a), aggregated white spots can be seen in the EcN cells transformed with pET28a-T5-ARG1 plasmid (b), which are vesicle proteins.


Based on the above presentation, we successfully expressed vesicular proteins in EcN that ultrasound could detect. The pET28a_T5-ARG plasmid, combined with the hybrid promoter, is expected to obtain engineered bacteria that can specifically colonize tumours and be detected by ultrasound. The engineered bacteria are expected to be used as an alternative to PET-CT in the future to provide help for the systemic detection of metastatic lesions in advanced pancreatic cancer.


4 Construction and debugging of dynamic encapsulation circuits


Though tumour-targeting and ultrasound-sensing has been achieved, controlled immunogenicity during in vivo delivery is still a problem. In order to avoid viremia as much as possible, we expect to mask epitopes of EcN before ultrasound imaging and achieve rapid clearance after diagnosis. Thus, we focused on engineering the capsular polysaccharide (CAP). CAP is a loose mucous substance located on the surface of the cell wall, protecting bacteria from environmental influences. It is worth mentioning that the thickness of CAP changes dynamically, and if not generated endogenously, its thickness will naturally become thinner. Harimoto et al 11^{11} have designed an inducible capsular polysaccharide (iCAP) system. They used the kfiC gene to regulate the expression of CAP. As an endogenous gene in EcN, kfiC encodes a glycotransferase essential in CAP biosynthesis, thus its expression determines the rate of CAP production, and then impacts the thickness of cellular outer layer. Based on the previous work, we designed the following circuit (Fig. 16a): kfiC under the control of ParaB (an arabinose-inducible promoter) would be transformed into EcN ∆asd ∆kfic. Prior to delivery, the bacteria would be induced by arabinose to start kfiC expression, and then encapsulation from CAP production, preparing it for host immune evasion and tumour focus localization. As CAP gradually degrades and cellular outer layer becomes thinner over time, the remaining bacteria in blood would be rapidly cleared, thus reducing long-term inflammatory response and toxicity risks (Fig. 16b) 11^{11}.


Figure 16

Figure 16 | The iCAP system. (a) Inducible gene circuit diagram wherein the kfiC gene was cloned under the control of ParaB (an arabinose-inducible promoter) to allow inducible CAP expression via the small molecule arabinose. (b) Workflow and parameter adjustment of iCAP system: prior to delivery, the bacteria are induced by arabinose to produce CAP and become encapsulated (with thickened OP). After intravenous injection, OP thickness gradually declines as CAP is being degraded, and bacteria elimination rate surges, allowing transition from immune evasion phase to rapid clearance phase, separated by the clearance cutoff. Ideally, by modulating arabinose concentration and consequently modulating CAP production rate, the timepoint corresponding to clearance cutoff should coincide with timepoint of ultrasound exposure, so as to preserve the bacteria for ultrasound detection, and eliminate it quickly after utilization as a 'disposable product'. OP, outer polysaccharide.


We tried to knockout the kfiC gene from the genome of EcN ∆asd in the same way as the asd gene (see Result 2.1), yet without success. A possible cause may be that both asd and kfiC genes are essential to bacterial cell wall synthesis, thus the absence of both would be fatal to EcN. We propose to change the output necessary gene of hybrid promoter to ThyA12^{12}. ThyA encodes thymidylate synthase in E. coli, which is required in nucleotide biosynthesis pathways, and does not interfere with cell wall synthesis. Nevertheless, we demonstrated the feasibility of our iCAP system through modelling. By changing the elimination rate kek_e in our MC model (see Dry Lab - Model), we were able to simulate an enrichment to the tumour compartment as well as rapid clearance in other compartments with suitable elimination rates determined by CAP thickness. This design can be utilized to optimize therapeutic bacteria administration to maximize curative effects and minimize side effects.


Reference


  1. Wing, H. J., Williams, S. M. & Busby, S. J. Spacing requirements for transcription activation by Escherichia coli FNR protein. Journal of Bacteriology 177, 6704–6710 (1995).
  2. Chien, T. et al. Enhancing the tropism of bacteria via genetically programmed biosensors. Nat Biomed Eng 6, 94–104 (2022).
  3. Mengesha, A. et al. Development of a flexible and potent hypoxia-inducible promoter for tumour-targeted gene expression in attenuated salmonella. Cancer Biology & Therapy 5, 1120–1128 (2006).
  4. Sun, Y. et al. Two-Layered Microfluidic Devices for High-Throughput Dynamic Analysis of Synthetic Gene Circuits in E. coli. ACS Synth. Biol. 11, 3954–3965 (2022).
  5. Moon, T. S., Lou, C., Tamsir, A., Stanton, B. C. & Voigt, C. A. Genetic programs constructed from layered logic gates in single cells. Nature 491, 249–253 (2012).
  6. Aguilera, L. et al. Dual Role of LldR in Regulation of the lldPRD Operon, Involved in l-Lactate Metabolism in Escherichia coli. Journal of Bacteriology 190, 2997–3005 (2008).
  7. Jiang, Y. et al. Multigene Editing in the Escherichia coli Genome via the CRISPR-Cas9 System. Appl Environ Microbiol 81, 2506–2514 (2015).
  8. Li, Q. I. et al. A modified pCas/pTargetF system for CRISPR-Cas9-assisted genome editing in Escherichia coli. ABBS 53, 620–627 (2021).
  9. Bourdeau, R. W. et al. Acoustic reporter genes for noninvasive imaging of microorganisms in mammalian hosts. Nature 553, 86–90 (2018).
  10. Solé, R. & Conde-Pueyo, N. Ultrasound approach tracks gut microbes. Nature 553, 36–37 (2018).
  11. Harimoto, T. et al. A programmable encapsulation system improves delivery of therapeutic bacteria in mice. Nat Biotechnol 40, 1259–1269 (2022).
  12. Fu, X. & Xu, J.-G. Development of a chromosome-plasmid balanced lethal system for Lactobacillus acidophilus with thyA gene as selective marker. Microbiology and immunology 44, 551–556 (2000).