Engineering

loading

Introduction

Inflammatory bowel disease (IBD) is a chronic and recurrent intestinal disease, mainly including Crohn's Disease (CD) and Ulcerative Colitis (UC). Although these two diseases have some similarities in clinical manifestations, such as chronic diarrhea, abdominal pain, and possible extraintestinal complications, they have significant differences in pathological features. CD mainly manifests as segmental inflammation of the intestine, accompanied by granuloma formation and fissure like ulcers. UC, on the other hand, presents as continuous mucosal inflammation without the formation of granulomas

The etiology of IBD is complex, involving multiple aspects such as genetic susceptibility, immune system dysfunction, imbalance of gut microbiota ecology, and environmental factors. These factors, either individually or in combination, contribute to the occurrence of IBD. Especially the disruption of intestinal barrier function and abnormal immune response to gut microbiota are considered key links in the development of diseases.

Figure:Metagenomic, metatranscriptomic, and stool metabolomic profiles are disrupted during IBD activity.

a,Relative abundance distributions for ten of the most cross-sectionally significantly differentially abundant metabolites in samples from individuals with IBD, as a ratio to the median relative abundance in individuals without IBD. Left, fraction of samples below detection limit. b, Relationships between two measures of disease activity: patient-reported (Harvey–Bradshaw index (HBI) in CD; simple clinical colitis activity index (SCCAI) in UC and host molecular (faecal calprotectin (cal)43. Linear regression shown with 95% confidence bound. c, d, Distribution of microbial dysbiosis scores as a measure of disease activity. e, Kaplan-Meier curves for the distributions of the durations of (left) and intervals between (right) dysbiotic episodes in UC and CD. Both are approximately exponential (fits in dashed lines), with means of 4.1 and 17.2 weeks, respectively, for UC, and 7.8 and 12.8 weeks for CD. f, Relative abundance distributions of significantly different metagenomic species, metabolites, and microbial transcribers in dysbiotic samples compared to non-dysbiotic samples from the same disease group . Also shown are antibody titres for ANCA, ASCA (IgG or IgA), anti-OmpC, and anti-CBir1 antibodies. (from Jason Lloyd-Price, Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases)

Recent studies have revealed multiple gene loci associated with IBD, such as NOD2/CARD15, IL23R, and IL10. These genes are involved in immune regulation, intestinal barrier function, and microbial community balance. Among them, the variation of NOD2/CARD15 is significantly associated with the risk of CD, while the polymorphism of IL23R and IL10 affects the susceptibility of UC. In addition, the reduction of gut microbiota diversity is also one of the important factors contributing to the onset of IBD.

The incidence rate and prevalence of IBD are on the rise worldwide, especially in western developed countries. It is reported that there are more than 3 million IBD patients in the United States, with 85387 new cases in 2019, accounting for 21% of the global incidence rate. Although the incidence of IBD is similar between males and females, genetic factors are significantly associated with familial clustering, especially in CD. Environmental factors such as diet, smoking, and lifestyle are also key external drivers of IBD.

In terms of clinical treatment, although Crohn's Disease (CD) and Ulcerative Colitis (UC) both belong to inflammatory bowel disease (IBD), there are significant differences in treatment plans and prognosis between the two. Traditional anti-inflammatory drugs have poor therapeutic effects on CD patients and may require the use of immunosuppressants or biologics to assist in treatment. In contrast, UC patients typically have a better response to aminosalicylic acid drugs and glucocorticoids. In terms of prognosis, due to the complex pathological process and varied clinical manifestations of CD, its prognosis is relatively poor and it is more likely to require surgical intervention, while UC has a relatively better prognosis, especially in early diagnosis and treatment.

In general, current treatment options for IBD are often non-specific and diffuse, and can only target a single factor in the treatment of the disease. In addition, the treatment is expensive and the process is often very painful. Therefore, we hope to use the technology of synthetic biology to design an engineered bacterium that can treat with spatiotemporal specificity, solve the problem from the source and is not limited to a single factor, reduce the side effects of treatment, simplify the complex course of treatment, and provide a cost-effective option!

After reviewing the literature, we took the gastrointestinal tract, the pathogenic location of IBD, as the entry point. Combined with the studies of intestinal microflora in recent years, we expected to design a plasmid that could express IBD therapeutic drugs on the basis of Escherichia coli as engineering bacteria.

We started by selecting the products of the genes. The most common symptom of IBD patients is inflammation of the digestive tract. From this point of view, anti-inflammatory therapy is a direct and rapid treatment choice. Therefore, we chose the base sequence encoding the anti-inflammatory factor as the main genetic element.

Once the bacteria were able to express the anti-inflammatory factors needed for treatment, the next question was how to make the modified bacteria survive in the gut. The intestinal environment is complex, in which there are a large number of intestinal flora. It is required that engineered bacteria should be at advantage in this community. Based on the original design, we added the adhesion factor which can adhere bacteria to the gut, so that the engineered bacteria can stay in the gut for a long time for recruitment and multiplication.

Gene expression requires not only its own coding sequence, but also the regulation of cis-acting elements and trans-acting factors on both sides of genes. Promoters play an important role in fast responding to the inflammatory environment and initiating gene expression. After meeting these conditions, the engineered bacteria is able to play a therapeutic effect in the right place and time, and reduce the adverse effects of the expressed anti-inflammatory factors in other parts of the human body. Therefore, we divided the selection range of promoters into genes that can respond to ROS, that is, select an ROS promoter.

The following schematic diagram of this engineered bacterial plasmid shows our three main components. Our whole component design process runs perfectly in the Design-Build-Test-Learn cycle, where we do a lot of literature research, design a lot of ideas, discuss the most appropriate ones to carry out the construction of the components, and test the effectiveness of the components, and then choose the most appropriate ones for optimization.Below we will introduce the design process of our components, and the ideas that were not selected will also be introduced in the idea library. Please follow our text through our Design-Build-Test-Learn cycle!

After selecting the target gene, we need a suitable "switch" to make it express at the right place and time. According to the characteristic that the concentration of ROS in the intestine of patients with enteritis is often higher than that in other parts of the body, this "switch" should be able to increase the expression of target genes in response to ROS, and hardly enhance the expression in the absence of ROS environment.

ROS Promotor

ROS, or reactive oxygen species, are molecules commonly found in organisms undergoing aerobic respiration. Under normal circumstances, it does not threaten the health of the organism. Typically, a single electron is initially transferred to oxygen to form a superoxide (O2-), which is then catalyzed by superoxide dismutase to become a hydroperoxide (H2O2). Superoxide is less likely to cross the membrane, which limits its range of influence. However, H2O2can easily spread to other sites. When the human body is in a state of disease, ROS production is greatly increased, and H2O2 which is of high diffusion will spread from the lesion to other healthy parts, causing oxidative damage to the Escherichia coli in this place [1].

Fig1. ROS are highly reactive and quickly transition between species.

(Picture resource: Brieger K, Schiavone S, Miller FJ Jr, Krause KH. Reactive oxygen species: from health to disease. Swiss Med Wkly. 2012 Aug 17;142:w13659. doi: 10.4414/smw.2012.13659. PMID: 22903797.)

To counter this adverse effect, organisms have evolved relative clearance systems. The well-known ways in which Escherichia coli eliminate H2O2 are glutathione reduction, the thioredoxin system, and the gludoxin system. The genes that respond to ROS and encode related enzymes differs within organisms. In E. coli, the key regulators of adaptive responses to hydrogen peroxide and superoxide anions are OxyR and SoxR and SoxS. Among them, OxyR direct oxidation is the mechanism by which Escherichia coli sense hydrogen peroxide and induce the production of OxyR regulon. OxyR proteins are homologous to the LysR-NodD family of bacterial regulatory proteins and bind to the promoter of the oxyR regulatory gene. OxyR protein can activate the transcription of oxyR regulatory genes in vitro [2]. As shown in Fig2, there are multiple cysteine residues on the reduced OxyR protein, containing many sulfhydryl groups. These groups can be oxidized by H2O2 to form disulfide bonds. Under the action of glutathione and gludoxin, OxyR can be regenerated in reduced form [3]. This suggests that oxidation of the OxyR protein brings about conformational changes [2]. The changes are mainly due to the intramolecular disulfide bond Cys199-Cys208[3]. The activated OxyR binds upstream of the promoters −35 sequence of the target genes such as dps,katG and gorA in the form of tetramers [3][4]. Transcription is stimulated by direct interaction with the carboxyl terminal domain of the alpha subunit of RNA polymerase [4]. In addition, both states of OxyR is capable to bind to the oxyR promoter as repressors that limit the amount of regulatory protein synthesis [3]. OxyR regulates at least eight genes, including gorA encoding glutathione, dps encoding non-specific DNA-binding protein, katG encoding hydroperoxidase I and ahpCF encoding alkyl hydroperoxide reductase [5]. When the expression of OxyR regulators increased, the expression of these genes also augmented. Therefore, we conducted comparative analysis in the genes of katG, dps, gorA and ahpCF to select the final promoter base sequence.

Fig2. The oxyR regulon

(Picture resource: Pomposiello PJ, Demple B. Redox-operated genetic switches: the SoxR and OxyR transcription factors. Trends Biotechnol. 2001 Mar;19(3):109-14. doi: 10.1016/s0167-7799(00)01542-0. PMID: 11179804.)

In a series of related studies, we founded that CARMEN MICHAÂN et al. used wild-type Escherichia coli in a culture medium containing 10μM H2O2 and measured gene expression of katG,dps,gorA and ahpCF over a period of time [5]. As shown in Fig3, we can find that katG has a rapid response to reactive oxygen stress and the longest duration among the genes to be selected. Therefore, it is the first choice as the promoter of engineered bacteria.

Fig3.In vivo transcription of the Escherichia coli oxyR regulon

Wild-type Escherichia coli from an overnight culture in M9 minimal medium were diluted in fresh medium and incubated at 37°C with shaking at 150 rpm. At an OD600 of 0.2, H2O2 was added to half of the culture, to make a final 10 M solution, and the rest was used as a control. Samples were collected immediately (1min) after the addition of H2O2 and at 5, 10, 15, and 20 min of exposure. Samples were frozen with liquid nitrogen, and total RNA was purified. The fluorescence signal of each PCR product was compared to that of gapA. Data are from an average of eight MPCR amplifications. Values from treated samples were divided by those from the corresponding control. All genes were analyzed, but only those genes for which statistically significant (P<0.05) increases were observed at a given time are represented. Error bars were estimated from the corresponding SEM. (Picture resource: Michán, C., Manchado, M., Dorado, G., & Pueyo, C. (1999). In vivo transcription of the Escherichia coli oxyR regulon as a function of growth phase and in response to oxidative stress. Journal of bacteriology, 181(9), 2759–2764. https://doi.org/10.1128/JB.181.9.2759-2764.1999)

There is an experimental analysis of reactive oxygen stress response of katG gene conducted separately. we could see that with the multiplication of bacteria (the curve indicated the change of OD600 value), the expression of katG gradually increased at first, and then showed a trend of decline after the number of bacteria was saturated. The activity of peroxidase, a gene expression product, had the same trend as the gene, but remained stable instead of decreasing later. This phenomenon further indicates that the expression of katG is related to H2O2 concentration. When H2O2 increased, the expression of katG went up. While the reaction between peroxidase and H2O2 reaches equilibrium, the expression of katG will gradually decrease. This mechanism can reduce the extra energy expenditure of cells. Therefore, the promoter of katG is suitable for the design of engineered bacteria.

In this paper, the changes of katG gene expression due to the increase of H2O2 caused by aerobic respiration during bacterial proliferation were analyzed by the RNA content expressed by katG. Escherichia coli cultured overnight in LB broth were diluted in fresh medium and incubated at 37°C and shaken at 150 rpm. Samples are collected at regular intervals and quickly cooled to 0°C. The culture growth is monitored by measuring OD600 for a specified time and is measured in the usual RNA purification and measurement manner.There is an experimental analysis of reactive oxygen stress response of katG gene conducted separately. With the multiplication of bacteria (the curve indicated the change of OD600 value), the expression of katG gradually increased at first, and then showed a trend of decline after the number of bacteria was saturated. The initial upregulation was the result of increased H2O2 output by aerobic metabolism under favorable conditions. The activity of peroxidase, a gene expression product, had the same trend as the gene, but remained stable instead of decreasing later. This phenomenon further indicates that the expression of katG is related to H2O2 concentration. When H2O2 increased, the expression of katG went up. While the reaction between peroxidase and H2O2 reaches equilibrium, the expression of katG will gradually decrease. This mechanism can reduce the extra energy expenditure of cells. Therefore, the promoter of katG is suitable for the design of engineered bacteria [5].

Fig4.Peroxidase activity during course of growth of wild-type bacteria in nutrient LB medium.

Activity data (darkly shaded bars) are from an average of four independent determinations. Error bars represent SEM. The katG expression data are shown in light bars. Statistical significance (P<0.05), for comparisons with minimal values at 1.5 h of outgrowth, is marked with asterisks. Bacterial growth, monitored as OD600, is indicated by the line. (Picture resource: Michán, C., Manchado, M., Dorado, G., & Pueyo, C. (1999). In vivo transcription of the Escherichia coli oxyR regulon as a function of growth phase and in response to oxidative stress. Journal of bacteriology, 181(9), 2759–2764.https://doi.org/10.1128/JB.181.9.2759-2764.1999)

[1] Brieger K, Schiavone S, Miller FJ Jr, Krause KH. Reactive oxygen species: from health to disease. Swiss Med Wkly. 2012 Aug 17;142:w13659. doi: 10.4414/smw.2012.13659. PMID: 22903797.

[2] Storz G, Tartaglia LA, Ames BN. The OxyR regulon. Antonie Van Leeuwenhoek. 1990 Oct;58(3):157-61. doi: 10.1007/BF00548927. PMID: 2256675.

[3] Pomposiello PJ, Demple B. Redox-operated genetic switches: the SoxR and OxyR transcription factors. Trends Biotechnol. 2001 Mar;19(3):109-14. doi: 10.1016/s0167-7799(00)01542-0. PMID: 11179804.

[4] Tao K. In vivo oxidation-reduction kinetics of OxyR, the transcriptional activator for an oxidative stress-inducible regulon in Escherichia coli. FEBS Lett. 1999 Aug 20;457(1):90-2. doi: 10.1016/s0014-5793(99)01013-3. PMID: 10486570.

[5] Michán C, Manchado M, Dorado G, Pueyo C. In vivo transcription of the Escherichia coli oxyR regulon as a function of growth phase and in response to oxidative stress. J Bacteriol. 1999 May;181(9):2759-64. doi: 10.1128/JB.181.9.2759-2764.1999. PMID: 10217765; PMCID: PMC93716.


Ideas Of Anti-inflammatory Factors


How to determine effective drugs?


As mentioned before, the engineered bacteria we designed have three important elements: reactive oxygen species initiation factors, adhesion factors, and anti-inflammatory factors. Among them, anti-inflammatory factors are the key to the therapeutic effect of engineered bacteria. How to determine effective and easy-to-express anti-inflammatory drug molecules is an important issue we have to face.

In the early stages of the project, we considered a variety of drugs that may have anti-inflammatory effects, such as Short chain fatty acids (SCFAs), DL-endopeptidase, IL-10, etc(We will display these ideas in the idea library). However, taking into account factors such as the absorption capacity of the pharmaceutical molecules, their anti-inflammatory capabilities and the size of the introduced gene, we believe that peptide-based drugs should be chosen. Peptides are small in molecular weight, easily absorbed, have a small gene length to save vector space, they are ideal pharmaceutical molecules.

Peptides with anti-inflammatory effects are currently less applied, and we have screened two peptides with anti-inflammatory effects through extensive literature review, namely melittin (MEL) and Indolicidin (Indo). Among them, melittin is the main active substance of bee venom, which has long been used in traditional Chinese medicine to treat inflammatory diseases, and modern researchers have also confirmed the anti-inflammatory effects of melittin. This has inspired us to apply melittin in the treatment of IBD. Below are the introductions to these two drugs.


Basic information on melittin:


Figure 1

Melittin is an amphipathic, water-soluble peptide consisting of 26 amino acid residues and is the principal active component of natural bee venom. The N-terminal region contains four positively charged residues, while the C-terminal region contains two positively charged residues. Due to the uneven distribution of polar and non-polar amino acid residues, melittin exhibits both hydrophilic and lipophilic amphipathic properties. The first 20 amino acids (from the N-terminus) are hydrophobic, whereas the subsequent 6 amino acids are hydrophilic. The complete sequence is as follows:

NH2-Gly-ile-Gly-Ala-Val-Leu-Lys-Val-Leu-Thr-Gly-Leu-Pro-Ala-Leu-ile-Ser-Trp-ile-Lys-Arg-Lys-Arg-Gln-Gln-CONH2

Figure 2

Reference: Ceremuga M, Stela M, Janik E, Gorniak L, Synowiec E, Sliwinski T, Sitarek P, Saluk-Bijak J, Bijak M. Melittin-A Natural Peptide from Bee Venom Which Induces Apoptosis in Human Leukaemia Cells. Biomolecules. 2020 Feb 6;10(2):247.

Each melittin chain exhibits a helix-hinge-helix motif or bent rod structure. Amino acid residues 3-10 form an α-helix, while residues 12-14 create a hinge region that connects to another α-helix containing residues 15-24. This unique structure, along with its amphipathic characteristics, enables melittin to penetrate cell membranes and interact with cellular substructures at the molecular level.


Action principle of melittin:


Effect of melittin on cell membrane:


Figure 3

Reference: Zhang HQ, Sun C, Xu N, Liu W. The current landscape of the antimicrobial peptide melittin and its therapeutic potential. Front Immunol. 2024 Jan 22;15:

Melittin monomers can attach to the membrane surface and spontaneously integrate into natural or artificial phospholipid bilayers, thereby reducing the rigidity between the polar and non-polar regions and decreasing the permeability barrier. It has been proposed that melittin-induced membrane permeability may result from the formation of toroidal pores or fissures within the membrane.


Anti-inflammatory effect of melittin:


Figure 4

Reference: Liu Minchen & Du Ruofei, Chinese Journal of Modern Applied Pharmacy, 2023

In terms of anti-inflammatory effects, melittin plays a crucial role in regulating inflammatory signaling pathways. It inhibits the activity of the NF-κB pathway, reducing the expression of inflammatory cytokines such as TNF-α, IL-1β, and IL-6. Melittin can suppress the phosphorylation of IKK and IκB, thereby blocking the nuclear translocation of NF-κB and interfering with inflammatory signal transduction. Additionally, melittin affects the MAPK signaling pathway by inhibiting the phosphorylation of JNK and p38, thus regulating the production of pro-inflammatory cytokines and reducing the activity of inflammatory cells and the secretion of inflammatory mediators. Furthermore, melittin interferes with the JAK/STAT pathway, inhibiting the activity of STAT transcription factors decreasing the secretion of inflammatory cytokines, which helps alleviate inflammatory diseases. Melittin also reduces inflammation by inhibiting Akt phosphorylation, which interferes with the expression of inflammatory proteins and suppresses the production of inflammatory mediators such as COX-2, iNOS, and cPLA2. Additionally, melittin promotes the recruitment of immune cells to the site of inflammation and induces T-cell apoptosis, thereby modulating the immune response and further alleviating inflammation.

Current experiments have demonstrated that melittin can be used to treat various inflammatory diseases, such as arthritis, skin inflammation, ulcerative colitis, and other inflammatory conditions.

table 1: The inflammatory diseases can be treated by melittin.

Reference:Liu Minchen &Du Ruofei, Chinese Journal of Modern Applied Pharmacy, 2023

下载图标

This project focuses on the treatment of IBD, with particular attention to ulcerative colitis (UC), a form of IBD characterized by persistent inflammation primarily affecting the superficial layers of the rectum and colon.

Interviews with clinicians reveal that conventional therapies for UC often fail to achieve satisfactory efficacy and have notable side effects. Therefore, more effective treatments for UC are needed. We think melittin may be a good choice for treatment, our and other experiments have proved this.


Therapeutic effect of melittin on ulcerative colitis:


Ahmedy et al. (2020) evaluated the anti-inflammatory effects of melittin through oral administration in an acetic acid-induced colitis mouse model. Melittin treatment ameliorated acetic acid-triggered decreases of body weight and an increase of colon mass index in colitis model mice (Figure 5). Compared to untreated colitis model mice, the melittin-treated group showed significantly reduced levels of tumor necrosis factor (TNF-α) and IL-6 (Figure 6). Melittin amended changes induced by acetic acid in the upstream inflammatory signaling pathways in colon such as NF-κB, Toll-like receptor 4 (TLR4), p38 MAPK, TNF receptor-associated factor 6 (TRAF6) (Figure 7). Melittin also reversed acetic acid-induced alterations in PGE2 and enzymes involved in its synthesis pathway in colon inculding secretory phospholipase A2 (sPLA2), and cyclooxygenase-2 (COX-2) (Figure 8). Furthermore, Melittin attenuated acetic acid-induced oxidative stress (Figure 9) and protected colon mucosa and submucosa against acetic acid-induced damage (Figure 10).

5-10Refernce:Ahmedy OA, Ibrahim SM, Salem HH, Kandil EA. Antiulcerogenic effect of melittin via mitigating TLR4/TRAF6 mediated NF-κB and p38MAPK pathways in acetic acid-induced ulcerative colitis in mice. Chem Biol Interact. 2020 Nov 1;331:109276.

Figure 5

Melittin inverted changes induced by acetic acid in body weight and colon mass index. a Change in body weight from day 0 to day 5 induced by acetic acid and melittin administration. b Effect of acetic acid and melittin on colon mass index in mice. Each bar with vertical line represents the mean ± S.E.M. of 10 mice per group; * significantly different from control group at p < 0.05, ** significantly different from control group at p < 0.001, *** significantly different from control group at p < 0.0001, significantly different from acetic acid group at p < 0.05, significantly different from acetic acid group at p < 0.0001 using One-Way ANOVA followed by Tukey multiple comparisons test.####

Figure 6

Melittin mitigated acetic acid-induced alterations in TNF-α and IL-6 levels in colon. Each bar with vertical line represents the mean ± S.E.M. of 6 mice per group; *** significantly different from control group at p < 0.0001, significantly different from acetic acid group at p < 0.0001 using One-Way ANOVA followed by Tukey multiple comparisons test.###

Figure 7

Melittin amended changes induced by acetic acid in the upstream inflammatory signaling pathways in colon. a Western blot analysis for the protein expression levels of TLR4, TRAF6 and p38 MAPK using β-actin as loading control (n = 3) b NF-κB level as measured by ELISA (n = 6). Each bar with vertical line represents the mean ± S.E.M.; * significantly different from control group at p < 0.05, ** significantly different from control group at p < 0.001, *** significantly different from control group at p < 0.0001, significantly different from acetic acid group at p < 0.0001 using One-Way ANOVA followed by Tukey multiple comparisons test.###

Figure 8

Melittin reversed acetic acid-induced alterations in PGE2 and enzymes involved in its synthesis pathway in colon. a PGE2 level as estimated by ELISA (n = 6). b Western blot analysis for the protein expression level of sPLA2 using β-actin as loading control (n = 3). c COX-2 level as estimated by ELISA (n = 6). Each bar with vertical line represents the mean ± S.E.M.; * significantly different from control group at p < 0.05, ** significantly different from control group at p < 0.001, *** significantly different from control group at p < 0.0001, significantly different from acetic group at p < 0.001 using One-Way ANOVA followed by Tukey multiple comparisons test.###

Figure 9

Melittin attenuated acetic acid-induced oxidative stress. Each bar with vertical line represents the mean ± S.E.M. of 6 mice per group; *** significantly different from control group at p < 0.0001, significantly different from acetic acid group at p < 0.05, significantly different from acetic acid group at p < 0.001, significantly different from acetic acid group at p < 0.0001 using One-Way ANOVA followed by Tukey multiple comparisons test.######

Figure 10

Melittin protected colon mucosa and submucosa against acetic acid-induced damage. (A) H & E staining of histological sections in the colon of different experimental groups. Control mice as well as those receiving melittin alone showed normal histological structures of colon wall with apparent intact mucosal lining including glandular elements and lining enterocytes, normal submucosa as well as outer muscular coat with minimal inflammatory cells infiltrates and normal vasculatures. However, mouse received acetic acid showed focal areas of erosion and damaged lining mucosa including glandular elements (arrows) with abundant intraluminal necrotic depresses and marked congestion of mucosal as well as submucosal blood vessels (dotted arrows), moderate submucosal edema and inflammatory cells infiltrates (star). On the other hand, mouse treated with melittin showed apparent intact intestinal mucosa, intact submucosal and outer muscular layers with occasional focal mononuclear inflammatory cells aggregates (H & E X 200 and X 50). (B) Histological scoring of colon mucosal damage. Results are expressed as median and range of 3 mice per group; ** significantly different from control group at p < 0.001, # significantly different from acetic acid group at p < 0.05, using Kruskal–Wallis one-way ANOVA followed by Dunn's multiple comparisons test.

Additionally, melittin reduced oxidative stress in colitis model mice. Atieh Yaghoubi et al. (2022) evaluated the anti-inflammatory properties of melittin in a dextran sulfate sodium (DSS)-induced colitis model.(Figure 11) They also analyzed the synergistic effects of melittin and sulfasalazine (SSZ) to assess whether combination therapy could improve clinical symptoms in UC model mice. The results indicated that both melittin alone and in combination with sulfasalazine significantly alleviated pathological damage in the colon tissues of UC model mice.(Figure 12-15) Melittin treatment exhibited antioxidant effects, suggesting that melittin could be a potential alternative therapy for UC intervention.

11-15Reference:Yaghoubi A, Amel Jamehdar S, Reza Akbari Eidgahi M, Ghazvini K. Evaluation of the therapeutic effect of melittin peptide on the ulcerative colitis mouse model. Int Immunopharmacol. 2022 Jul;108:108810. 

Figure 11

Melittin peptide improve the clinical symptoms of DSS-induced colitis in mice. (A, B, and D) Therapy with melittin peptide effect on colon weight and colon shortening that induced by DSS; (C) Evaluation of the colon weight to length ratio in a different group in comparison to the colitis group. [(*p < 0.05; ***p < 0.001 compare to the control group); (#p < 0.05; ##p < 0.01; ### p < 0.001 compare to the colitis group)]. (DSS=dextran sulfate sodium; SSZ=Sulfasalazine).

Figure 12

Melittin peptide ameliorated the disease activity index. (A and B) Therapy with melittin peptide (2.4 mg/kg/day) alone or along with SSZ (100 mg/kg/day) was measured in a different group in comparison to colitis group. [(**p < 0.01; ***p < 0.001 compare to the control group); (##p < 0.01; ### p < 0.001 compare to the colitis group); (@@p < 0.01; @@@p < 0.001 compare to the Melittin group); (++p < 0.01; +++p < 0.001 compare to the Sulfasalazine group); ($p < 0.05; $$p < 0.01; $$$p < 0.001 compare to the Melittin in combination with sulfasalazine group)].

Figure 13

Melittin peptide attenuated edema and inflammation of colon tissue induced by DSS in colitis mice. Evaluation of the effect of the therapy with melittin peptide alone or along with SSZ on (A) inflammation score; (B) Mucosal damage score; (C) and crypt loss in different groups. [(*p < 0.05; **p < 0.01; ***p < 0.001 compare to the control group); (##p < 0.01; ### p < 0.001 compare to the colitis group); (+p < 0.05; ++p < 0.01 compare to the Sulfasalazine group)].

Figure 14

Melittin peptide effect on histological damage to colon tissue in DSS-induced colitis mice. (A) Pathological damage ranges were measured in the different groups; (B) Evaluation of histological score of the colon in different treatment groups; (C) Submucosal edema and infiltration of the inflammatory cell in the histological result of different groups. [(*p < 0.05; **p < 0.01; ***p < 0.001 compare to the control group); (### p < 0.001 compare to the colitis group); (+p < 0.05; ++p < 0.01 compare to the Sulfasalazine group)].

Figure 15

Melittin effect on oxidative stress markers in DSS-induced colitis mice. The outcomes of melittin and melittin along with SSZ (A) MDA; (B) SOD activity; (C) catalase enzyme activity; and (D) total thiol of the serum in comparison to the colitis group. [(***p < 0.001 compare to the control group); (#p < 0.05; ### p < 0.001 compare to the colitis group); (@@p < 0.01; @@@p < 0.001 compare to the Melittin group); (+++p < 0.001 compare to the Sulfasalazine group)] (DSS=dextran sulfate sodium; SSZ=Sulfasalazine; MDA=malonyl dialdehyde; SOD=Superoxide dismutase).


Artificial modification of melittin


To further enhance the performance of melittin, we conducted in-depth research and precise modifications. In the modification process, we employed an innovative approach by connecting two melittin monomers with a linker to form a unique hairpin structure. This modification resulted in multiple performance improvements. The specific amino acid and polypeptide sequences are shown in Figure 16.

Figure 16

The artificial modification significantly reduced the cytotoxicity of melittin. By introducing this structure, we effectively decreased the peptide's toxicity to normal cells, greatly enhancing its application safety. This improvement allows melittin to be used in a wider range of scenarios, particularly in treatments requiring prolonged contact with biological tissues. Additionally, the modified melittin shows a marked increase in immunostimulatory capacity (building on previous laboratory work). This feature enhances its potential in regulating immune and inflammatory responses. The enhanced immunostimulatory ability not only helps activate the immune system more effectively but may also facilitate appropriate inflammation modulation, offering new possibilities for treating IBD.


Conclusion


Therefore, due to its anti-inflammatory and antioxidant properties, we have selected melittin as one of the drug molecules for this project.


Indolicidin


Indolicidin, as a naturally occurring compound with significant bioactivity, is gradually revealing its potential applications in the treatment of colitis. Studies indicate that due to its unique anti-inflammatory and immunomodulatory properties, indolicidin can act through various mechanisms, particularly excelling in modulating the gut microbiota and enhancing intestinal barrier function. The synergistic effects of these functions enable indolicidin to effectively alleviate symptoms of colitis, thereby improving patients' quality of life.


Basic information:


Figure 17

Indolicidin, discovered by Selsted et al. (1992), is a natural peptide belonging to the cathelicidin family isolated from bovine neutrophils. This peptide consists of 13 amino acid residues, nearly half of which are hydrophobic, including 5 tryptophan residues. These tryptophan residues are responsible for insertion and distribution within biological membranes, leading to the peptide's hemolytic activity (Subbalakshmi et al., 1996). The physicochemical properties are shown in Table 2.

Table 2: Sequence and physicochemical properties of indolicidin

Reference:Batista Araujo J, Sastre de Souza G, Lorenzon EN. Indolicidin revisited: biological activity, potential applications and perspectives of an antimicrobial peptide not yet fully explored. World J Microbiol Biotechnol. 2022 Jan 12;38(3):39.


Action principle:


Representation of indolicidin's mechanism of action after transient membrane perturbation,intracellular potential targets are proteins and nucleic acids (a), indolicidin-LPS interactionrelated to anti-sepsis and anti-biofilm activity (b) and the indolicidin anion carrier mechanismis thought to explain hemolytic activity(c).

Figure 18

(a)The bacteriostatic effect of indolicotin: Indolicidin has a broad range of biological targets, including both Gram-positive and Gram-negative bacteria (Vergis et al., 2020). Additionally, Indolicidin can disrupt established biofilms and inhibit biofilm formation. At the molecular level, although Indolicidin can insert into the plasma membrane, it does not directly cause membrane rupture. Instead, it enters the cytoplasm, binds to DNA, and inhibits the synthesis of proteins and DNA.

(b)Indolicidin neutralizes the pro-inflammatory effects of LPS: Lipopolysaccharide (LPS) is considered an endotoxin and a major outer membrane component of almost all Gram-negative bacteria, acting as a potent immune system stimulant (Cohen 2002). LPS activates inflammatory cells, such as monocytes and macrophages, leading to the release of cytokines (e.g., TNF-α, IL-1β, and IL-6) and nitric oxide, thereby triggering an inflammatory cascade (Nan et al., 2009a; Park et al., 2009). Indolicidin can bind and neutralize LPS, making it a potential anti-inflammatory agent.

(c)The indolicidin anion carrier mechanismis thought to explain hemolytic activity.

Reference:Batista Araujo J, Sastre de Souza G, Lorenzon EN. Indolicidin revisited: biological activity, potential applications and perspectives of an antimicrobial peptide not yet fully explored. World J Microbiol Biotechnol. 2022 Jan 12;38(3):39.


Conclusion:


Melittin and indolicidin, as novel peptide-based drugs, exhibit significant anti-inflammatory potential. Both inhibit inflammatory responses through different mechanisms, improving gut health. Our project focuses on these two anti-inflammatory agents, aiming to fully utilize their properties and enhance clinical efficacy and safety, providing new therapeutic options for IBD patients.


Anti-inflammatory Factors


Peptide cell experiment


Next, we will use the cell model constructed above for preliminary drug screening experiments. Thaw macrophages and culture them in wells on a 24 well plate after two passages. Wait for the cells to grow full again and perform the same starvation treatment on the cells. After infection, different concentrations of Mel and Indo were applied to the experimental group, and the cells were collected after four hours of cultivation. Compared with the control group treated only with PBS solution, the experimental group cells showed varying degrees of decreased levels of reactive oxygen species(ROS), decreased levels of some inflammatory factors, and increased expression of anti-inflammatory factor IL-10.

Use flow cytometry to detect cellular ROS levels. As shown in Figure 1A, the experimental groups treated with 5, 10, and 15 ppm Mel and 5 and 15 ppm Indo showed a significant decrease in the level of ROS in cells. However, the experimental results of applying different concentrations of peptides to uninfected macrophages showed that Mel and Indo did not reduce the level of ROS in healthy cells (Figure 1B). The above results indicate that appropriate concentrations of Mel and Indo can significantly reduce the levels of ROS in inflammatory cells.

图片1 图片2 图片3 图片4

Figure 1A

图片1 图片2 图片3 图片4

Figure 1B

The expression levels and changes of four inflammatory factors in cells were detected by RT-qPCR technology, and the results are shown in Figure 2A. Only concentrations of 5 and 10 ppm in the Indo group showed significant decrease in IL-1 β. In the Mel group, a concentration of 15ppm of Mel significantly upregulated all four inflammatory factors, while a concentration of 10ppm significantly downregulated the expression levels of IL-1 β and IL-6. We speculate that both peptides at appropriate concentrations can downregulate the expression of some inflammatory factors. Excessive concentrations may lead to peptides acting as antigenic substances to stimulate macrophages to generate inflammatory response, resulting in a significant increase in the expression level of inflammatory factors.

In addition, we also conducted cell counts and intracellular cell counts after infection. As shown in Figure 2B, there was no significant change in the number of cells after experimental treatment in all groups. Except for a significant decrease in the number of intracellular bacteria in cells treated with Mel at concentrations of 5 and 10 ppm, there was no significant change in the number of intracellular bacteria in the other groups.

Figure 2A

图片1 图片2
图片1 图片2

Figure 2B


In vivo peptide experiments


The above experiments have demonstrated that Mel and Indo at appropriate concentrations have certain anti-inflammatory effects in an in vitro cellular inflammation model. Next, we will further validate the anti-inflammatory effects of these two drugs in mice.

Firstly, a mouse intestinal inflammation model was constructed. A batch of female Balb/cJNju-Foxn1nu/Nju Hfh11nu mice were ordered and experimental procedures were carried out according to the table and all fluids were administered into the mice by gavage. The concentration of Salmonella is , Mel and Indo is 10ppm. The intestinal specimens taken were from the cecum to the colon.

We measured the colon length of each group of mice to characterize the level of intestinal inflammation. The results showed that both Mel and Indo could significantly alleviate the shortened colon length in mice caused by intestinal inflammation, and Mel had better alleviating effect (Figure 3A).

图片1 图片2

Figure 3A

Figure 3B

Take some colon tissue for microscopic observation. After dehydration, embedding, and sectioning of colon tissue, HE staining was performed. The results are shown in Figure 3B. It can be seen that compared with the PBS group, the S.Tm group had damaged intestinal epithelial cells and disrupted intestinal mucosal integrity. However, the symptoms of the treatment group were significantly relieved, and the Mel group had better effects. Some tissue sections were processed for immunofluorescence staining. The results are shown in Figure 3C. It can be seen that the IL-6 fluorescence signal intensity of the tissues treated with Salmonella by gavage was significantly higher than that of the PBS group, indicating more severe intestinal inflammation. However, the IL-6 fluorescence intensity of the Mel and Indo groups was significantly lower than that of the S.Tm group, and both Mel and Indo had certain anti-inflammatory effects.

Figure 3C

Cut off a portion of colon tissue, completely dissociate it, stain immune factors and immune cell marker proteins, and then measure their expression levels using flow cytometry. As shown in Figure 3D, Mel can significantly reduce the levels of IFN-γ, IL-6, TNF-α, F4/80, and Ly6G in the colon tissue of mice with enteritis, while Indo can only reduce the levels of IFN-γ, TNF-α, and F4/80

图片1 图片2
图片1 图片2
图片1 图片2
图片1 图片2
图片1 图片2

Figure 3D

Take a portion of colon tissue, extract cellular RNA, and perform RT qPCR to detect the expression levels of various inflammatory factors inside the cells. The results are shown in Figure 3E. Compared with the control group, the expression levels of IL-6, IL-1 β, and IL-8 in the intestinal tissue after Mel treatment were significantly reduced, while after Indo treatment, only IL-6 expression levels were significantly reduced, and other inflammatory factors showed no significant changes or significant increases. In addition, compared to the positive control group, there was no significant change in IL-10 levels in the Mel group, while the Indo group showed a significant increase.

Through in vitro and in vivo experiments with the two peptides mentioned above, we found that compared to Indo, Mel exhibited better anti-inflammatory performance in all the indicators we tested. Therefore, we selected Mel for further experimental research.

图片1 图片2 图片3
图片1 图片2 图片3

Figure 3E


Adhesion Factor CMC


The key to the efficient functioning of FMK is to make the engineered bacteria FMK stay in the intestine longer and express anti-inflammatory factors in response. Therefore, the team simulated the binding between the glucan binding protein and the glucan substrate, and designed the artificial glucan binding protein CMC.

Glucan is an extremely important polysaccharide produced by a large number of bacteria and fungi. At the same time, the surface of intestinal epithelial cells secretes mucus, which plays a protective role as a barrier. The main component of mucus is the highly glycosylated glycoprotein MUC2, which has various polysaccharide structures such as Core1, Core2 and Core3 [1]. At the same time, the beneficial properties of probiotics are related to the extracellular polysaccharide (EPS) they produce [2], and the surface of most probiotics can be exposed to glucan. Lactobacillus in Firmicutes has various functions, such as regulating the balance of intestinal microbial community, promoting food digestion and nutrient absorption, alleviating intestinal inflammation and infection, etc. Its cell wall is mainly composed of peptidoglycan and polysaccharide. For example, Lactobacillus can produce glucan sucrase (GS), and synthesize EPSs through glucose-transfer glycogen [3], such as α-glucan [4]. Salmonella is a common food-borne pathogen that uses a variety of virulence factors to overcome colonization resistance and induce intestinal inflammation [5], all of which are Gram-negative bacteria. Lipopolysaccharide, an important component of cell wall, is an endotoxin and contains O antigen [6]. S.typhimurium is a zoonotic pathogen that causes a large proportion of non-typhoid Salmonella (NTS) infections, accounting for a quarter of infections, second only to Salmonella enteritidis serotype in incidence [7]. Therefore, we thought of combining this characteristic of glucan as the core idea for designing synthetic proteins, so that the adhesion factors we designed can serve as a "bridge" between intestinal microorganisms and the intestines, recruit intestinal probiotics, and achieve stable attachment of engineered bacteria to the intestines. This idea inspired us to design CBMcipc domain, which is derived from the scaffold protein CipC of Clostridium cellulolyticum and contains a group III cellulose binding domain (CBD), a hydrophilic domain and two hydrophobic domains. The CBD domain confers CBMcipc with the ability to bind to glucan[8].

In order to select the best glucan-binding protein, fluorescent protein mCherry sequence was introduced in the experiment, and proteins M, CM, CMC and CMCC with different copy numbers of Clostridium cellulolyticum CipC (CBMcipc) were designed. By introducing pET-28a plasmid carrying corresponding genes (namely M, CM, CMC, CMC, CMCC) into Escherichia coli BL21, the synthetic Escherichia coli strains EcM, EcCM, EcCMC, EcCMCC were constructed. The proteins M, CM, CMC and CMCC are composed of mCherry, mCherry and one copy of CBMcipc, mCherry and two copies of CBMcipc, mCherry and three copies of CBMcipc (Fig1a). The N-terminal connected outer membrane protein A (OmpA) signal peptide helps the protein to pass through the intima and locate on the outer membrane surface [8], thus giving the protein surface display ability.

To verify the above properties, cells were stained with FITC-labeled glucan for flow cytometry and confocal microscopy. After incubation with FITC-glucan for 10 minutes, EcM cells showed almost no FITC fluorescence, while EcCM, EcCMC and EcCMCC cells showed obvious FITC fluorescence. Among the three CBMcipc containing strains, EcCMC cells showed the highest FITC fluorescence (Fig 1b, c), indicating that EcCMC had the highest glucan-binding capacity.

Confocal microscopy images and fluorescence quantification further revealed that EcM, EcCM, and EcCMCC cells had a whole-cell distribution of mCherry fluorescence, while EcCMC cells had a bilateral distribution of mCherry, indicating that CMC exhibited a stronger surface display ability(Fig1d). Since the cell-surface display of the binding protein is necessary for it to bind to dextran outside the cell, this property of EcCMC may help enhance its dextran binding ability.

Fig1. Design and characterization of the synthetic bacteria used for glucan binding[7]. (Based on the preliminary research findings from the laboratory)

Gavage experiments with EcCM, EcCMC, EcCMCC mixed with Lactobacillus plantarum (probiotics) were done on mice to assess the ratio of the number of beneficial intestinal bacteria to the amount of beneficial bacteria in the gavage after 12 hours of gavage (adherence rate) and the results showed that EcCMC had the highest adherence rate (Fig2). Whereas, after replacing the probiotics with Salmonella enterica serovar Typhimurium subsp. enterica (pathogenic bacteria), the adhesion rate was lower in all cases, which reflects the preference of adhesion factors for binding to probiotics.

Fig2 The adhesion rate of exogenous yeast was detected by intragastric administration.

Based on the above characteristics, CMC was selected as a adhesion factor in the experiment, hoping to achieve the colonization of engineered bacteria in the intestine and the recruitment of intestinal probiotics through its expression in the intestine, so as to better alleviate intestinal inflammation and regulate the intestinal microenvironment.

Reference:

[1].NIE Shuo, WEN Zhengshun. Secretion, Structure, Synthesis Regulation of Intestinal Mucin 2 and Its Role in Development of Intestinal Diseases. Chinese Journal of Animal Nutrition, 2020, 32(6): 2521-2532.

[2]. Pourjafar, Hadi et al. “Functional and health-promoting properties of probiotics' exopolysaccharides; isolation, characterization, and applications in the food industry.” Critical reviews in food science and nutrition vol. 63,26 (2023): 8194-8225. doi:10.1080/10408398.2022.2047883

[3].Yu, Liansheng et al. “Glucansucrase Produced by Lactic Acid Bacteria: Structure, Properties, and Applications.” Fermentation (2022): n. pag.

[4].Chen, Ziwei et al. “Lactic acid bacteria-derived α-glucans: From enzymatic synthesis to miscellaneous applications.” Biotechnology advances vol. 47 (2021): 107708. doi:10.1016/j.biotechadv.2021.107708

[5].Whitfield, Chris et al. “Lipopolysaccharide O-antigens-bacterial glycans made to measure.” The Journal of biological chemistry vol. 295,31 (2020): 10593-10609. doi:10.1074/jbc.REV120.009402

[6].Park, Jeong Soon et al. “Mechanism of anchoring of OmpA protein to the cell wall peptidoglycan of the gram-negative bacterial outer membrane.” The FASEB Journal 26 (2012): 219 - 228.

[7]. Yin, Hongda et al. “Synthetic physical contact-remodeled rhizosphere microbiome for enhanced phytoremediation.” Journal of hazardous materials vol. 433 (2022): 128828. doi:10.1016/j.jhazmat.2022.128828